1
|
Sidky H, Hansen KA, Girvin AT, Hotaling N, Michael SG, Gersing K, Sahner DK. Assessing the effect of selective serotonin reuptake inhibitors in the prevention of post-acute sequelae of COVID-19. Comput Struct Biotechnol J 2024; 24:115-125. [PMID: 38318198 PMCID: PMC10839808 DOI: 10.1016/j.csbj.2023.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 02/07/2024] Open
Abstract
Background Post-acute sequelae of COVID-19 (PASC) produce significant morbidity, prompting evaluation of interventions that might lower risk. Selective serotonin reuptake inhibitors (SSRIs) potentially could modulate risk of PASC via their central, hypothesized immunomodulatory, and/or antiplatelet properties although clinical trial data are lacking. Materials and Methods This retrospective study was conducted leveraging real-world clinical data within the National COVID Cohort Collaborative (N3C) to evaluate whether SSRIs with agonist activity at the sigma-1 receptor (S1R) lower the risk of PASC, since agonism at this receptor may serve as a mechanism by which SSRIs attenuate an inflammatory response. Additionally, determine whether the potential benefit could be traced to S1R agonism. Presumed PASC was defined based on a computable PASC phenotype trained on the U09.9 ICD-10 diagnosis code. Results Of the 17,908 patients identified, 1521 were exposed at baseline to a S1R agonist SSRI, 1803 to a non-S1R agonist SSRI, and 14,584 to neither. Using inverse probability weighting and Poisson regression, relative risk (RR) of PASC was assessed.A 29% reduction in the RR of PASC (0.704 [95% CI, 0.58-0.85]; P = 4 ×10-4) was seen among patients who received an S1R agonist SSRI compared to SSRI unexposed patients and a 21% reduction in the RR of PASC was seen among those receiving an SSRI without S1R agonist activity (0.79 [95% CI, 0.67 - 0.93]; P = 0.005).Thus, SSRIs with and without reported agonist activity at the S1R were associated with a significant decrease in the risk of PASC.
Collapse
Affiliation(s)
- Hythem Sidky
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kristen A. Hansen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
- Axle Research and Technologies, Rockville, MD, USA
| | | | - Nathan Hotaling
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
- Axle Research and Technologies, Rockville, MD, USA
| | - Sam G. Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
- Palantir Technologies, Denver, CO, USA
- Axle Research and Technologies, Rockville, MD, USA
| | - Ken Gersing
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - David K. Sahner
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
- Axle Research and Technologies, Rockville, MD, USA
| | - on behalf of the N3C consortium
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
- Palantir Technologies, Denver, CO, USA
- Axle Research and Technologies, Rockville, MD, USA
| |
Collapse
|
2
|
Basting CM, Langat R, Broedlow CA, Guerrero CR, Bold TD, Bailey M, Velez A, Schroeder T, Short-Miller J, Cromarty R, Mayer ZJ, Southern PJ, Schacker TW, Safo SE, Bramante CT, Tignanelli CJ, Schifanella L, Klatt NR. SARS-CoV-2 infection is associated with intestinal permeability, systemic inflammation, and microbial dysbiosis in hospitalized patients. Microbiol Spectr 2024; 12:e0068024. [PMID: 39345212 PMCID: PMC11537016 DOI: 10.1128/spectrum.00680-24] [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: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) and its associated severity have been linked to uncontrolled inflammation and may be associated with changes in the microbiome of mucosal sites including the gastrointestinal tract and oral cavity. These sites play an important role in host-microbe homeostasis, and disruption of epithelial barrier integrity during COVID-19 may potentially lead to exacerbated inflammation and immune dysfunction. Outcomes in COVID-19 are highly disparate, ranging from asymptomatic to fatal, and the impact of microbial dysbiosis on disease severity is unclear. Here, we obtained plasma, rectal swabs, oropharyngeal swabs, and nasal swabs from 86 patients hospitalized with COVID-19 and 12 healthy volunteers. We performed 16S rRNA sequencing to characterize the microbial communities in the mucosal swabs and measured concentrations of circulating cytokines, markers of gut barrier integrity, and fatty acids in the plasma samples. We compared these plasma concentrations and microbiomes between healthy volunteers and COVID-19 patients, some of whom had unfortunately died by the end of the study enrollment, and performed a correlation analysis between plasma variables and bacterial abundances. Rectal swabs of COVID-19 patients had reduced abundances of several commensal bacteria including Faecalibacterium prausnitzii and an increased abundance of the opportunistic pathogens Eggerthella lenta and Hungatella hathewayi. Furthermore, the oral pathogen Scardovia wiggsiae was more abundant in the oropharyngeal swabs of COVID-19 patients who died. The abundance of both H. hathewayi and S. wiggsiae correlated with circulating inflammatory markers including IL-6, highlighting the possible role of the microbiome in COVID-19 severity and providing potential therapeutic targets for managing COVID-19.IMPORTANCEOutcomes in coronavirus disease 2019 (COVID-19) are highly disparate and are associated with uncontrolled inflammation; however, the individual factors that lead to this uncontrolled inflammation are not fully understood. Here, we report that severe COVID-19 is associated with systemic inflammation, microbial translocation, and microbial dysbiosis. The rectal and oropharyngeal microbiomes of COVID-19 patients were characterized by a decreased abundance of commensal bacteria and an increased abundance of opportunistic pathogens, which positively correlated with markers of inflammation and microbial translocation. These microbial perturbations may, therefore, contribute to disease severity in COVID-19 and highlight the potential for microbiome-based interventions in improving COVID-19 outcomes.
Collapse
Affiliation(s)
| | - Robert Langat
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Candace R. Guerrero
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Tyler D. Bold
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Melisa Bailey
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adrian Velez
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ty Schroeder
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonah Short-Miller
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minnesota, Minneapolis, USA
| | - Zachary J. Mayer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Peter J. Southern
- Department of Microbiology and Immunology, University of Minnesota, Minnesota, Minneapolis, USA
| | - Timothy W. Schacker
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Sandra E. Safo
- Department of Biostatistics and Health Data Science, University of Minnesota, Minnesota, Minneapolis, USA
| | - Carolyn T. Bramante
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | | | - Luca Schifanella
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- National Cancer Institute, Center for Cancer Research, Vaccine Branch, Animal Models and Retroviral Vaccines Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Nichole R. Klatt
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Johnson SG, Abedian S, Stürmer T, Huling JD, Lewis V C, Buse JB, Brosnahan SB, Mudumbi PC, Erlandson KM, McComsey GA, Arnold J, Wiggen TD, Wong R, Murphy S, Rosen C, Kaushal R, Weiner MG, Bramante C. Prevalent Metformin Use in Adults With Diabetes and the Incidence of Long COVID: An EHR-Based Cohort Study From the RECOVER Program. Diabetes Care 2024; 47:1930-1940. [PMID: 39287394 PMCID: PMC11502533 DOI: 10.2337/dca24-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Studies show metformin use before and during SARS-CoV-2 infection reduces severe COVID-19 and postacute sequelae of SARS-CoV-2 (PASC) in adults. Our objective was to describe the incidence of PASC and possible associations with prevalent metformin use in adults with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS This is a retrospective cohort analysis using the National COVID Cohort Collaborative (N3C) and Patient-Centered Clinical Research Network (PCORnet) electronic health record (EHR) databases with an active comparator design that examined metformin-exposed individuals versus nonmetformin-exposed individuals who were taking other diabetes medications. T2DM was defined by HbA1c ≥6.5 or T2DM EHR diagnosis code. The outcome was death or PASC within 6 months, defined by EHR code or computable phenotype. RESULTS In the N3C, the hazard ratio (HR) for death or PASC with a U09.9 diagnosis code (PASC-U09.0) was 0.79 (95% CI 0.71-0.88; P < 0.001), and for death or N3C computable phenotype PASC (PASC-N3C) was 0.85 (95% CI 0.78-0.92; P < 0.001). In PCORnet, the HR for death or PASC-U09.9 was 0.87 (95% CI 0.66-1.14; P = 0.08), and for death or PCORnet computable phenotype PASC (PASC-PCORnet) was 1.04 (95% CI 0.97-1.11; P = 0.58). Incident PASC by diagnosis code was 1.6% metformin vs. 2.0% comparator in the N3C, and 2.1% metformin vs. 2.5% comparator in PCORnet. By computable phenotype, incidence was 4.8% metformin and 5.2% comparator in the N3C and 24.7% metformin vs. 26.1% comparator in PCORnet. CONCLUSIONS Prevalent metformin use is associated with a slightly lower incidence of death or PASC after SARS-CoV-2 infection. PASC incidence by computable phenotype is higher than by EHR code, especially in PCORnet. These data are consistent with other observational analyses showing prevalent metformin is associated with favorable outcomes after SARS-CoV-2 infection in adults with T2DM.
Collapse
Affiliation(s)
- Steven G. Johnson
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN
| | - Sajjad Abedian
- Information Technologies and Services Department, Weill Cornell Medicine, New York, NY
| | - Til Stürmer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jared D. Huling
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN
| | - Colby Lewis V
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY
| | - John B. Buse
- Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Shari B. Brosnahan
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Praveen C. Mudumbi
- Department of Population Health, New York University Grossman School of Medicine, New York, NY
| | | | | | - Jonathan Arnold
- Division of General Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Talia D. Wiggen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN
| | - Rachel Wong
- Department of Biomedical Informatics and the Department of Internal Medicine, Stony Brook University, Stony Brook, NY
| | | | | | - Rainu Kaushal
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY
| | - Mark G. Weiner
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY
| | - Carolyn Bramante
- Division of General Internal Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | | |
Collapse
|
4
|
Biswal S, Mallick B. Unlocking the potential of signature-based drug repurposing for anticancer drug discovery. Arch Biochem Biophys 2024; 761:110150. [PMID: 39265695 DOI: 10.1016/j.abb.2024.110150] [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: 04/21/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Cancer is the leading cause of death worldwide and is often associated with tumor relapse even after chemotherapeutics. This reveals malignancy is a complex process, and high-throughput omics strategies in recent years have contributed significantly in decoding the molecular mechanisms of these complex events in cancer. Further, the omics studies yield a large volume of cancer-specific molecular signatures that promote the discovery of cancer therapy drugs by a method termed signature-based drug repurposing. The drug repurposing method identifies new uses for approved drugs beyond their intended initial therapeutic use, and there are several approaches to it. In this review, we discuss signature-based drug repurposing in cancer, how cancer omics have revolutionized this method of drug discovery, and how one can use the cancer signature data for repurposed drug identification by providing a step-by-step procedural handout. This modern approach maximizes the use of existing therapeutic agents for cancer therapy or combination therapy to overcome chemotherapeutics resistance, making it a pragmatic and efficient alternative to traditional resource-intensive and time-consuming methods.
Collapse
Affiliation(s)
- Sruti Biswal
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India.
| |
Collapse
|
5
|
Du W, Chen H, Gróf I, Lemaitre L, Bocsik A, Perdyan A, Mieczkowski J, Deli MA, Hortobágyi T, Wan Q, Glebov OO. Antidepressant-induced membrane trafficking regulates blood-brain barrier permeability. Mol Psychiatry 2024; 29:3590-3598. [PMID: 38816584 PMCID: PMC11541205 DOI: 10.1038/s41380-024-02626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
As the most prescribed psychotropic drugs in current medical practice, antidepressant drugs (ADs) of the selective serotonin reuptake inhibitor (SSRI) class represent prime candidates for drug repurposing. The mechanisms underlying their mode of action, however, remain unclear. Here, we show that common SSRIs and selected representatives of other AD classes bidirectionally regulate fluid-phase uptake at therapeutic concentrations and below. We further characterize membrane trafficking induced by a canonical SSRI fluvoxamine to show that it involves enhancement of clathrin-mediated endocytosis, endosomal system, and exocytosis. RNA sequencing analysis showed few fluvoxamine-associated differences, consistent with the effect being independent of gene expression. Fluvoxamine-induced increase in membrane trafficking boosted transcytosis in cell-based blood-brain barrier models, while a single injection of fluvoxamine was sufficient to enable brain accumulation of a fluid-phase fluorescent tracer in vivo. These findings reveal modulation of membrane trafficking by ADs as a possible cellular mechanism of action and indicate their clinical repositioning potential for regulating drug delivery to the brain.
Collapse
Affiliation(s)
- Wenjia Du
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Huanhuan Chen
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ilona Gróf
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Lucien Lemaitre
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Alexandra Bocsik
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Adrian Perdyan
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Jakub Mieczkowski
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Mária A Deli
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Tibor Hortobágyi
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Oleg O Glebov
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
| |
Collapse
|
6
|
Yumoto T, Coopersmith CM. Targeting AMP-activated protein kinase in sepsis. Front Endocrinol (Lausanne) 2024; 15:1452993. [PMID: 39469575 PMCID: PMC11513325 DOI: 10.3389/fendo.2024.1452993] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Sepsis is a global health challenge marked by limited clinical options and high mortality rates. AMP-activated protein kinase (AMPK) is a cellular energy sensor that mediates multiple crucial metabolic pathways that may be an attractive therapeutic target in sepsis. Pre-clinical experimental studies have demonstrated that pharmacological activation of AMPK can offer multiple potential benefits during sepsis, including anti-inflammatory effects, induction of autophagy, promotion of mitochondrial biogenesis, enhanced phagocytosis, antimicrobial properties, and regulation of tight junction assembly. This review aims to discuss the existing evidence supporting the therapeutic potential of AMPK activation in sepsis management.
Collapse
Affiliation(s)
- Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
7
|
Wu K, Yin L, Han J, Cai Q, Guo Y, Jin X, Wu J, Cheng Y. Case-control study on risk factors for in-hospital mortality in patients with severe COVID-19. Front Public Health 2024; 12:1424720. [PMID: 39440172 PMCID: PMC11493594 DOI: 10.3389/fpubh.2024.1424720] [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: 04/28/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Objective The purpose of this study was to identify independent risk factors affecting patient survival and explore predictors of severe cases of coronavirus disease 2019 (COVID-19). Methods We conducted a retrospective, observational, case-control study on adult patients with severe COVID-19 who were admitted to affiliated hospitals in Tianjin between December 18, 2022, and January 31, 2023. We used univariate and multifactorial logistic regression analyses to analyze demographic indicators, comorbidity profiles, and laboratory parameters in two groups of patients (deceased and surviving) to identify independent risk factors for death in patients with severe COVID-19. Results Patients in the deceased group were older than those in the survival group (p = 0.018), and there were more cases of coexisting respiratory insufficiency in the deceased group (p = 0.002). Additionally, laboratory test results for white blood cell count (WBC) and creatine kinase (CK) showed significantly higher values in the deceased group (p = 0.047 and p = 0.029, respectively), while arterial oxygen partial pressure (PAO2) showed significantly lower values compared to the survival group (p = 0.021). Age, respiratory insufficiency, WBCH (highest WBC value), CKH (highest CK value), and PAO2F (first PAO2 value) had area under curve (AUC) values of 0.698, 0.838, 0.721, 0.744, and 0.633, respectively. Conclusion The main risk factors for mortality in patients with severe COVID-19 that we identified in this study were the advanced age of patients, coexisting respiratory insufficiency, elevated levels of WBC and CK, and decreased levels of PAO2. Elevated WBC and CK laboratory parameters, in particular, demonstrated good predictive value for in-hospital mortality risk.
Collapse
Affiliation(s)
- Kemei Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lili Yin
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangqin Han
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuhan Cai
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Guo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Jin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinling Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yupei Cheng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
8
|
Rothman RL, Stewart TG, Mourad A, Boulware DR, McCarthy MW, Thicklin F, Garcia Del Sol IT, Garcia JL, Bramante CT, Shah NS, Singh U, Williamson JC, Rebolledo PA, Jagannathan P, Schwasinger-Schmidt T, Ginde AA, Castro M, Jayaweera D, Sulkowski M, Gentile N, McTigue K, Felker GM, DeLong A, Wilder R, Collins S, Dunsmore SE, Adam SJ, Hanna GJ, Shenkman E, Hernandez AF, Naggie S, Lindsell CJ. Time to Sustained Recovery Among Outpatients With COVID-19 Receiving Montelukast vs Placebo: The ACTIV-6 Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2439332. [PMID: 39422912 DOI: 10.1001/jamanetworkopen.2024.39332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Importance The effect of montelukast in reducing symptom duration among outpatients with mild to moderate COVID-19 is uncertain. Objective To assess the effectiveness of montelukast compared with placebo in treating outpatients with mild to moderate COVID-19. Design, Setting, and Participants This randomized clinical trial (Accelerating COVID-19 Therapeutic Interventions and Vaccines [ACTIV]-6) was conducted from January 27 through June 23, 2023, during the circulation of Omicron subvariants. Participants aged 30 years or older with confirmed SARS-CoV-2 infection and 2 or more acute COVID-19 symptoms for less than 7 days were included across 104 US sites. Interventions Participants were randomized 1:1 to receive montelukast, 10 mg once daily, or matched placebo for 14 days. Main Outcomes and Measures The primary outcome was time to sustained recovery (defined as ≥3 consecutive days without symptoms). Secondary outcomes included time to death; time to hospitalization or death; a composite of health care utilization events (hospitalization, urgent care clinic visit, emergency department visit, or death); COVID-19 clinical progression scale score; and difference in mean time unwell. A modified intention-to-treat approach was used for the analysis. Results Among 1250 participants who were randomized and received the study drug or placebo, the median age was 53 years (IQR, 42-62 years), 753 (60.2%) were female, and 704 (56.3%) reported receiving 2 or more doses of a SARS-CoV-2 vaccine. Among 628 participants who received montelukast and 622 who received placebo, differences in time to sustained recovery were not observed (adjusted hazard ratio [AHR], 1.02; 95% credible interval [CrI], 0.92-1.12; P = .63 for efficacy). Unadjusted median time to sustained recovery was 10 days (95% CI, 10-11 days) in both groups. No deaths occurred, and hospitalizations were reported for 2 participants (0.3%) in each group; the composite of health care utilization events was reported for 18 participants (2.9%) in the montelukast group and 18 (2.9%) in the placebo group (AHR, 1.01; 95% CrI, 0.45-1.84; P = .48 for efficacy). Five participants (0.4%) experienced serious adverse events (3 [0.5%] in the montelukast group and 2 [0.3%] in the placebo group). Conclusions and Relevance In this randomized clinical trial of outpatients with mild to moderate COVID-19, treatment with montelukast did not reduce duration of COVID-19 symptoms. These findings do not support the use of montelukast for the treatment of mild to moderate COVID-19. Trial Registration ClinicalTrials.gov Identifier: NCT04885530.
Collapse
Affiliation(s)
| | | | - Ahmad Mourad
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - David R Boulware
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | | | | | | | | | - Carolyn T Bramante
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | | | | | - John C Williamson
- Section on Infectious Diseases, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Paulina A Rebolledo
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | | | | | - Adit A Ginde
- University of Colorado School of Medicine, Aurora
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City
| | - Dushyantha Jayaweera
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Mark Sulkowski
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland
| | - Nina Gentile
- Department of Emergency Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kathleen McTigue
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - G Michael Felker
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | - Allison DeLong
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | - Rhonda Wilder
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | - Sean Collins
- Vanderbilt University Medical Center, Nashville, Tennessee
- Geriatric Research Education and Clinical Center, Veterans Affairs Tennessee Valley Healthcare System, Nashville
| | - Sarah E Dunsmore
- National Center for Advancing Translational Sciences, Bethesda, Maryland
| | - Stacey J Adam
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - George J Hanna
- Biomedical Advanced Research and Development Authority, Washington, DC
| | - Elizabeth Shenkman
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville
| | - Adrian F Hernandez
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | - Susanna Naggie
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
- Department of Medicine, University of Minnesota Medical School, Minneapolis
| | | |
Collapse
|
9
|
Bonnet U, Juckel G. [The Impact of Antidepressants on COVID-19 and Post-Acute COVID-19 Syndrome: A Scoping-Review Update]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024. [PMID: 39313202 DOI: 10.1055/a-2374-2218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Introduction Preclinically, fluvoxamine and other antidepressants (AD) exerted antiviral and anti-inflammatory properties also against SARS-COV-2. Therfore, It makes sense to test the clinical effect of AD against COVID-19 and Long COVID. METHODS On May 20, 2024, this systematic scoping review in PUBMED identified 1016 articles related to AD and COVID-19, Long COVID and SARS-COV-2. These included 10 retrospective "large scale" studies (> 20000 chart reviews), 8 prospective clinical trials (plus 4 regarding Long COVID), 11 placebo-controlled randomized (RCT) (plus 2 regarding Long COVID) and 15 meta-analyses. RESULTS COVID-19: Retrospective studies with cohorts taking AD primarily for psychiatric comorbidities or chronic pain conditions directly prior to SARS-COV-2 infection described that this substance class (most studied: Selective Serotonin Re-Uptake Inhibitors (SSRI) and Selective Serotonin Noradrenaline Re-Uptake Inhibitors (SSNRI)) were associated with (i) significantly fewer SARS-COV-2 infections and (ii) a milder course of COVID-19 ("COVID-19 protection"). Ten of the 11 RCTs found regarding COVID-19 tested fluvoxamine, as this old AD appeared suitable as a prophylactic agent against severe COVID-19, taking into account its in vitro potency against the progression of intracellular sepsis cascades. Therefore, most (12 out of 15) meta-analyses also referred to fluvoxamine. They found (iii) a significant (40-70% reduction) in mortality, intubation and hospitalization rates when fluvoxamine was used as an add-on to standard therapy for mild to moderate COVID-19. When this AD was used in the early stages of the disease, it was more successful than when it was given later in advanced, severe COVID-19 (e.g. severe pneumonia, final sepsis stages). A dose dependency was observed: 2x50 mg fluvoxamine over 15 days was less effective than 2x100 or even 3x100 mg with an adverse event profile still at the placebo level. Direct comparisons with drugs approved for COVID-19 do not yet exist. A first indirect meta-analytical comparison showed an advantage of paxlovid or molnupiravir versus fluvoxamine against the development of severe COVID-19: risk reduction of 95% (I2 = N/A, but only one study) or 78% (I2=0) versus 5+-5% (I2=48). However, an add-on of fluvoxamine was still significantly more efficacious than symptom-oriented standard therapy alone. Long COVID: A common Long COVID phenotype with dominant anxiety and depression symptoms, which responds to AD, relaxation therapy and/or psychotherapy, has now been identified. Casuistics report positive effects of AD on fatigue, cognitive and autonomic dysfunctions. A first large prospective open-label RCT has just shown significantly more favourable courses, less viral load and less pro-inflammatory cytokines in the treatment of mild to moderate COVID-19 with fluvoxamine versus standard treatment, also with regard to the subsequent development of neuropsychiatric and pulmonary Long COVID or fatigue. CONCLUSION Overall, there is promising evidence of a preventive effect of AD (especially fluvoxamine) against progression to severe COVID-19 and against the development of Long COVID. It is likely, that the entire AD substance class could be effective here. This assumption is based on the results of retrospective large scale studies, but awaits verification by better controlled studies. The potential effectiveness/efficacy (currently low and moderate confidence of the evidence for the entire substance class and specifically fluvoxamine, respectively) of fluvoxamine as an add-on against COVID-19 and possibly also directly against Long COVID could stimulate similar projects in other infectious diseases that also have the potential to pose a lasting threat to the health of those affected. We consider the evidence to date to be sufficient to be able to emphasize a possible positive effect of these substances in the psychoeducation of patients with COVID-19 or Long COVID who are already receiving AD for other conditions - especially also against the symptoms associated with the viral disease or its consequences. In regions where neither vaccines nor antiviral agents currently approved for the prevention or treatment of COVID-19 are available, AD and in particular fluvoxamine would be a cost-effective alternative to protect against a severe course, even if this AD appears to have a smaller effect against COVID-19 than the currently approved antiviral agents, but with presumably better tolerability. A direct comparative clinical trial with approved antiviral agents is still pending and should be positive to further open the door for a guideline-based recommendation of fluvoxamine (or perhaps even AD) for COVID-19 or its aftermath.
Collapse
Affiliation(s)
- Udo Bonnet
- Klinik für Seelische Gesundheit, Akademisches Lehrkrankenhaus der Universität Duisburg-Essen, Evangelisches Krankenhaus Castrop-Rauxel, Castrop-Rauxel, Germany
- LVR-Universitätsklinikum Essen, Klinik für Psychiatrie und Psychotherapie, Medizinische Fakultät, Universität Duisburg-Essen, Essen, Germany
| | - Georg Juckel
- LWL-Universitätsklinikum der Ruhr-Universität Bochum, Klinik für Psychiatrie, Psychotherapie und Präventivmedizin, Bochum, Germany
| |
Collapse
|
10
|
Harmon DC, Levene JA, Rutlen CL, White ES, Freeman IR, Lapidus JA. Preadmission Metformin Use Is Associated with Reduced Mortality in Patients with Diabetes Mellitus Hospitalized with COVID-19. J Gen Intern Med 2024:10.1007/s11606-024-08864-x. [PMID: 39299975 DOI: 10.1007/s11606-024-08864-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Observational studies have reported an association between metformin and improved outcomes in COVID-19, but most have been small and with significant limitations. OBJECTIVE To evaluate the association between preadmission metformin exposure and mortality in patients with diabetes mellitus hospitalized with coronavirus disease 2019 (COVID-19) infection. DESIGN Retrospective cohort analysis using electronic health records extracted from the American Heart Association COVID-19 Registry. PARTICIPANTS Adults (n = 11,993) with diabetes mellitus but without chronic kidney disease (CKD) or need for hemodialysis who were hospitalized with COVID-19 between January 25, 2020, and February 9, 2022. MAIN MEASURES We used propensity score modeling to address differences between metformin and non-metformin users prior to multivariable log-binomial models to examine the association between metformin use at time of hospital admission for COVID-19 infection and in-hospital death; composite of in-hospital death or discharge to hospice; composite of in-hospital death, discharge to hospice, or ICU admission; and composite of in-hospital death, discharge to hospice, ICU admission, or mechanical ventilation. KEY RESULTS Compared to metformin non-use, pre-admission metformin use was associated with lower risk of in-hospital death (risk ratio (RR) 0.81 [95% CI 0.75-0.90]); composite of in-hospital death or discharge to hospice (RR 0.79 [95% CI 0.74-0.87]); composite of in-hospital death, discharge to hospice, or ICU admission (RR 0.90 [95% CI 0.86-0.95]); and composite of in-hospital death, discharge to hospice, ICU admission, or mechanical ventilation (RR 0.9 [95% CI 0.84-0.98]). Metformin use was also associated with lower risk of death due to respiratory cause (RR 0.86 [95% CI 0.74-0.97]) but not cardiovascular (RR 0.84 [95% CI 0.58-1.2]) or other (RR 0.78 [95% CI 0.60-1.0]) causes. CONCLUSIONS Pre-admission metformin use was associated with lower risk of in-hospital mortality and markers of disease severity among adults with diabetes mellitus without CKD and not requiring hemodialysis who were hospitalized with COVID-19 infection.
Collapse
Affiliation(s)
- David C Harmon
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA.
| | - Jacqueline A Levene
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Christine L Rutlen
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Elizabeth S White
- Oregon Health & Science University - Portland State University (OHSU-PSU) School of Public Health, Portland, OR, USA
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Ilana R Freeman
- Oregon Health & Science University - Portland State University (OHSU-PSU) School of Public Health, Portland, OR, USA
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Jodi A Lapidus
- Oregon Health & Science University - Portland State University (OHSU-PSU) School of Public Health, Portland, OR, USA
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Claveau S, Mahmood F, Amir B, Kwan JJW, White C, Vipond J, Iannattone L. COVID-19 and Cancer Care: A Review and Practical Guide to Caring for Cancer Patients in the Era of COVID-19. Curr Oncol 2024; 31:5330-5343. [PMID: 39330021 PMCID: PMC11431468 DOI: 10.3390/curroncol31090393] [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: 06/25/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
COVID-19, a novel infectious disease caused by the emergence of the SARS-CoV-2 virus in 2020, has had a profound impact on healthcare, both at the individual and population level. The impact at the population level was felt most acutely during the emergency phase of the pandemic, with hospital capacity issues leading to widespread disruptions and delays in the delivery of healthcare services such as screening programs and elective surgeries. While hospitals are no longer being acutely overwhelmed by COVID-19 patients, the impact of the virus on vulnerable patient populations such as cancer patients continues to be of ongoing consequence. Cancer patients remain at high risk of hospitalization, ICU admission, and death due to COVID-19, even in the era of vaccination. Infection prevention and risk mitigation strategies such air quality control, masking, testing, vaccination, and treatment should therefore be integrated into the usual care and counseling of cancer patients moving forward to avoid preventable morbidity and mortality from this infection and ensure the safety of this vulnerable cohort as they navigate their cancer diagnosis and treatment in the era of COVID-19.
Collapse
Affiliation(s)
- Simon Claveau
- Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Farhan Mahmood
- Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Baraa Amir
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | | | - Cheryl White
- Independent Researcher, Toronto, ON M6P 3X9, Canada
| | - Joe Vipond
- Department of Emergency Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Lisa Iannattone
- Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
12
|
Peters AE, Jones WS, Anderson B, Bramante CT, Broedl U, Hornik CP, Kehoe L, Knowlton KU, Krofah E, Landray M, Locke T, Patel MR, Psotka M, Rockhold FW, Roessig L, Rothman RL, Schofield L, Stockbridge N, Trontell A, Curtis LH, Tenaerts P, Hernandez AF. Framework of the strengths and challenges of clinically integrated trials: An expert panel report. Am Heart J 2024; 275:62-73. [PMID: 38795793 PMCID: PMC11330722 DOI: 10.1016/j.ahj.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
The limitations of the explanatory clinical trial framework include the high expense of implementing explanatory trials, restrictive entry criteria for participants, and redundant logistical processes. These limitations can result in slow evidence generation that is not responsive to population health needs, yielding evidence that is not generalizable. Clinically integrated trials, which integrate clinical research into routine care, represent a potential solution to this challenge and an opportunity to support learning health systems. The operational and design features of clinically integrated trials include a focused scope, simplicity in design and requirements, the leveraging of existing data structures, and patient participation in the entire trial process. These features are designed to minimize barriers to participation and trial execution and reduce additional research burdens for participants and clinicians alike. Broad adoption and scalability of clinically integrated trials are dependent, in part, on continuing regulatory, healthcare system, and payer support. This analysis presents a framework of the strengths and challenges of clinically integrated trials and is based on a multidisciplinary expert "Think Tank" panel discussion that included representatives from patient populations, academia, non-profit funding agencies, the U.S. Food and Drug Administration, and industry.
Collapse
Affiliation(s)
- Anthony E Peters
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - W Schuyler Jones
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | | | - Carolyn T Bramante
- Departmentd of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | | | - Christoph P Hornik
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC; Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Lindsay Kehoe
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Kirk U Knowlton
- Intermountain Medical Center Heart Institute, Salt Lake City, UT
| | | | | | - Trevan Locke
- Margolis Institute for Health Policy, Duke University, Durham, NC
| | - Manesh R Patel
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | | | - Frank W Rockhold
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC; Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC
| | | | | | | | - Norman Stockbridge
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Anne Trontell
- Patient-Centered Outcomes Research Institute (PCORI), Washington, DC
| | - Lesley H Curtis
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | | | - Adrian F Hernandez
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
13
|
Liang Y, Quan X, Gu R, Meng Z, Gan H, Wu Z, Sun Y, Pan H, Han P, Liu S, Dou G. Repurposing existing drugs for the treatment ofCOVID-19/SARS-CoV-2: A review of pharmacological effects and mechanism of action. Heliyon 2024; 10:e35988. [PMID: 39247343 PMCID: PMC11379597 DOI: 10.1016/j.heliyon.2024.e35988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Following the coronavirus disease-2019 outbreak caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), there is an ongoing need to seek drugs that target COVID-19. First off, novel drugs have a long development cycle, high investment cost, and are high risk. Second, novel drugs must be evaluated for activity, efficacy, safety, and metabolic performance, contributing to the development cycle, investment cost, and risk. We searched the Cochrane COVID-19 Study Register (including PubMed, Embase, CENTRAL, ClinicalTrials.gov, WHO ICTRP, and medRxiv), Web of Science (Science Citation Index, Emerging Citation Index), and WHO COVID-19 Coronaviral Disease Global Literature to identify completed and ongoing studies as of February 20, 2024. We evaluated the pharmacological effects, in vivo and in vitro data of the 16 candidates in the paper. The difficulty of studying these candidates in clinical trials involving COVID-19 patients, dosage of repurposed drugs, etc. is discussed in detail. Ultimately, Metformin is more suitable for prophylactic administration or mildly ill patients; the combination of Oseltamivir, Tamoxifen, and Dexamethasone is suitable for moderately and severely ill patients; and more clinical trials are needed for Azvudine, Ribavirin, Colchicine, and Cepharanthine to demonstrate efficacy.
Collapse
Affiliation(s)
- Yutong Liang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiaoxiao Quan
- Beijing Institute of Radiation Medicine, Beijing, China
- Scientific Experimental Center of Guangxi University of Chinese Medicine, Nanning, China
| | - Ruolan Gu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhiyun Meng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhuona Wu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Huajie Pan
- General Internal Medicine Department, Jingnan Medical District, PLA General Hospital, Beijing, China
| | - Peng Han
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Guifang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
14
|
Bramante CT, Beckman KB, Mehta T, Karger AB, Odde DJ, Tignanelli CJ, Buse JB, Johnson DM, Watson RHB, Daniel JJ, Liebovitz DM, Nicklas JM, Cohen K, Puskarich MA, Belani HK, Siegel LK, Klatt NR, Anderson B, Hartman KM, Rao V, Hagen AA, Patel B, Fenno SL, Avula N, Reddy NV, Erickson SM, Fricton RD, Lee S, Griffiths G, Pullen MF, Thompson JL, Sherwood NE, Murray TA, Rose MR, Boulware DR, Huling JD. Favorable Antiviral Effect of Metformin on SARS-CoV-2 Viral Load in a Randomized, Placebo-Controlled Clinical Trial of COVID-19. Clin Infect Dis 2024; 79:354-363. [PMID: 38690892 PMCID: PMC11327787 DOI: 10.1093/cid/ciae159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Metformin has antiviral activity against RNA viruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The mechanism appears to be suppression of protein translation via targeting the host mechanistic target of rapamycin pathway. In the COVID-OUT randomized trial for outpatient coronavirus disease 2019 (COVID-19), metformin reduced the odds of hospitalizations/death through 28 days by 58%, of emergency department visits/hospitalizations/death through 14 days by 42%, and of long COVID through 10 months by 42%. METHODS COVID-OUT was a 2 × 3 randomized, placebo-controlled, double-blind trial that assessed metformin, fluvoxamine, and ivermectin; 999 participants self-collected anterior nasal swabs on day 1 (n = 945), day 5 (n = 871), and day 10 (n = 775). Viral load was quantified using reverse-transcription quantitative polymerase chain reaction. RESULTS The mean SARS-CoV-2 viral load was reduced 3.6-fold with metformin relative to placebo (-0.56 log10 copies/mL; 95% confidence interval [CI], -1.05 to -.06; P = .027). Those who received metformin were less likely to have a detectable viral load than placebo at day 5 or day 10 (odds ratio [OR], 0.72; 95% CI, .55 to .94). Viral rebound, defined as a higher viral load at day 10 than day 5, was less frequent with metformin (3.28%) than placebo (5.95%; OR, 0.68; 95% CI, .36 to 1.29). The metformin effect was consistent across subgroups and increased over time. Neither ivermectin nor fluvoxamine showed effect over placebo. CONCLUSIONS In this randomized, placebo-controlled trial of outpatient treatment of SARS-CoV-2, metformin significantly reduced SARS-CoV-2 viral load, which may explain the clinical benefits in this trial. Metformin is pleiotropic with other actions that are relevant to COVID-19 pathophysiology. CLINICAL TRIALS REGISTRATION NCT04510194.
Collapse
Affiliation(s)
- Carolyn T Bramante
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kenneth B Beckman
- Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tanvi Mehta
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amy B Karger
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Darrell M Johnson
- Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ray H B Watson
- Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jerry J Daniel
- Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - David M Liebovitz
- General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jacinda M Nicklas
- General Internal Medicine, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Ken Cohen
- UnitedHealth Group, Optum Labs, Minnetonka, Minnesota, USA
| | - Michael A Puskarich
- Emergency Medicine, Hennepin County Medical Center, Minneapolis, Minnesota, USA
| | - Hrishikesh K Belani
- Department of Medicine, Olive View-University of California, Los Angeles, California, USA
| | - Lianne K Siegel
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nichole R Klatt
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Blake Anderson
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Katrina M Hartman
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Via Rao
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aubrey A Hagen
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Barkha Patel
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sarah L Fenno
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nandini Avula
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Neha V Reddy
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Spencer M Erickson
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Regina D Fricton
- General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Samuel Lee
- General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gwendolyn Griffiths
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew F Pullen
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jennifer L Thompson
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nancy E Sherwood
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas A Murray
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael R Rose
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David R Boulware
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jared D Huling
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Siedner MJ, Sax PE. Repurposing Revisited: Exploring the Role of Metformin for Treatment of COVID-19. Clin Infect Dis 2024; 79:292-294. [PMID: 38690870 DOI: 10.1093/cid/ciae154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Indexed: 05/03/2024] Open
Affiliation(s)
- Mark J Siedner
- Medical Practice Evaluation Center and Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Clinical Research Department, Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Paul E Sax
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases and Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Wimalawansa SJ. Unlocking insights: Navigating COVID-19 challenges and Emulating future pandemic Resilience strategies with strengthening natural immunity. Heliyon 2024; 10:e34691. [PMID: 39166024 PMCID: PMC11334859 DOI: 10.1016/j.heliyon.2024.e34691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024] Open
Abstract
The original COVID-19 vaccines, developed against SARS-CoV-2, initially mitigated hospitalizations. Bivalent vaccine boosters were used widely during 2022-23, but the outbreaks persisted. Despite this, hospitalizations, mortality, and outbreaks involving dominant mutants like Alpha and Delta increased during winters when the population's vitamin D levels were at their lowest. Notably, 75 % of human immune cell/system functions, including post-vaccination adaptive immunity, rely on adequate circulatory vitamin D levels. Consequently, hypovitaminosis compromises innate and adaptive immune responses, heightening susceptibility to infections and complications. COVID-19 vaccines primarily target SARS-CoV-2 Spike proteins, thus offering only a limited protection through antibodies. mRNA vaccines, such as those for COVID-19, fail to generate secretory/mucosal immunity-like IgG responses, rendering them ineffective in halting viral spread. Additionally, mutations in the SARS-CoV-2 binding domain reduce immune recognition by vaccine-derived antibodies, leading to immune evasion by mutant viruses like Omicron variants. Meanwhile, the repeated administration of bivalent boosters intended to enhance efficacy resulted in the immunoparesis of recipients. As a result, relying solely on vaccines for outbreak prevention, it became less effective. Dominant variants exhibit increased affinity to angiotensin-converting enzyme receptor-2, enhancing infectivity but reducing virulence. Meanwhile, spike protein-related viral mutations do not impact the potency of widely available, repurposed early therapies, like vitamin D and ivermectin. With the re-emergence of COVID-19 and impending coronaviral pandemics, regulators and health organizations should proactively consider approval and strategic use of cost-effective adjunct therapies mentioned above to counter the loss of vaccine efficacy against emerging variants and novel coronaviruses and eliminate vaccine- and anti-viral agents-related serious adverse effects. Timely implementation of these strategies could reduce morbidity, mortality, and healthcare costs and provide a rational approach to address future epidemics and pandemics. This perspective critically reviews relevant literature, providing insights, justifications, and viewpoints into how the scientific community and health authorities can leverage this knowledge cost-effectively.
Collapse
Affiliation(s)
- Sunil J. Wimalawansa
- Medicine, Endocrinology, and Nutrition, B14 G2, De Soyza Flats, Moratuwa, Sri Lanka
| |
Collapse
|
17
|
Yoon H, Pirofski LA. Generating the Evidence Base for Convalescent Plasma Use for a New Infectious Disease. Curr Top Microbiol Immunol 2024. [PMID: 39117847 DOI: 10.1007/82_2024_275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) swept across the world in the waning months of 2019 and emerged as the cause of the coronavirus disease 19 (COVID-19) pandemic in early 2020. The use of convalescent plasma (CP) for prior respiratory pandemics provided a strong biological rationale for the rapid deployment of COVID-19 convalescent plasma (CCP) in early 2020 when no validated treatments or prior immunity existed. CCP is an antiviral agent, with its activity against SARS-CoV-2 stemming from specific antibodies elicited by the virus. Early efforts to investigate the efficacy of CCP in randomized clinical trials (RCTs) that targeted hospitalized patients with COVID-19 did not demonstrate the overall efficacy of CCP despite signals of benefit in certain subgroups, such as those treated earlier in disease. In contrast, studies adhering to the principles of antibody therapy in their study design, choice of patient population, and product qualification, i.e., those that administered high levels of specific antibody during the viral phase of disease in immunocompromised or very early in immunocompetent individuals, demonstrated benefits. In this chapter, we leverage the knowledge gained from clinical studies of CCP for COVID-19 to propose a framework for future studies of CP for a new infectious disease. This framework includes obtaining high-quality CP and designing clinical studies that adhere to the principles of antibody therapy to generate a robust evidence base for using CP.
Collapse
Affiliation(s)
- Hyunah Yoon
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
18
|
Hernandez AV, Liu A, Roman YM, Burela PA, Pasupuleti V, Thota P, Carranza-Tamayo CO, Retamozo-Palacios M, Benites-Zapata VA, Piscoya A, Vidal JE. Efficacy and safety of ivermectin for treatment of non-hospitalized COVID-19 patients: A systematic review and meta-analysis of 12 randomized controlled trials with 7,035 participants. Int J Antimicrob Agents 2024; 64:107248. [PMID: 38908535 DOI: 10.1016/j.ijantimicag.2024.107248] [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: 08/07/2023] [Revised: 03/15/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
INTRODUCTION We systematically assessed benefits and harms of the use of ivermectin in non-hospitalized patients with early COVID-19. METHODS Five databases were searched until October 17, 2023, for randomized controlled trials (RCTs) in adult patients with COVID-19 treated with ivermectin against standard of care (SoC), placebo, or active drug. Primary outcomes were hospitalization, all-cause mortality, and adverse events (AEs). Secondary outcomes included mechanical ventilation (MV), clinical improvement, clinical worsening, viral clearance, and severe adverse events (SAEs). Random effects meta-analyses were performed, with quality of evidence (QoE) evaluated using GRADE methods. Pre-specified subgroup analyses (ivermectin dose, control type, risk of bias, follow-up, and country income) and trial sequential analysis (TSA) were performed. RESULTS Twelve RCTs (n = 7,035) were included. The controls were placebo in nine RCTs, SoC in two RCTs, and placebo or active drug in one RCT. Ivermectin did not reduce hospitalization (relative risk [RR], 0.81, 95% confidence interval [95% CI] 0.64-1.03; 8 RCTs, low QoE), all-cause mortality (RR 0.98, 95% CI 0.73-1.33; 9 RCTs, low QoE), or AEs (RR 0.89, 95% CI 0.75-1.07; 9 RCTs, very low QoE) vs. controls. Ivermectin did not reduce MV, clinical worsening, or SAEs and did not increase clinical improvement and viral clearance vs. controls (very low QoE for secondary outcomes). Subgroup analyses were mostly consistent with main analyses, and TSA-adjusted risk for hospitalization was similar to main analysis. CONCLUSIONS In non-hospitalized COVID-19 patients, ivermectin did not have effect on clinical, non-clinical or safety outcomes versus controls. Ivermectin should not be recommended as treatment in non-hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Adrian V Hernandez
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs, CT, USA; Vicerrectorado de Investigación, Universidad San Ignacio de Loyola (USIL), Lima, Peru.
| | - Anna Liu
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs, CT, USA
| | - Yuani M Roman
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs, CT, USA
| | - Paula Alejandra Burela
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | - Manuel Retamozo-Palacios
- Department of Infectious Diseases, Hospital Regional de Taguatinga, Taguatinga, Brasília-DF, Brazil
| | - Vicente A Benites-Zapata
- Master Program in Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru
| | - Alejandro Piscoya
- Servicio de Gastroenterología, Departamento de Medicina, Hospital Guillermo Kaelin de la Fuente, Lima, Peru
| | - Jose E Vidal
- Division of Infectious Diseases, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Department of Neurology, Instituto de Infectologia Emílio Ribas, São Paulo, Brazil; Laboratory of Medical Investigation, Unit 49, Hospital das Clinicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Cousins HC, Nayar G, Altman RB. Computational Approaches to Drug Repurposing: Methods, Challenges, and Opportunities. Annu Rev Biomed Data Sci 2024; 7:15-29. [PMID: 38598857 DOI: 10.1146/annurev-biodatasci-110123-025333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Drug repurposing refers to the inference of therapeutic relationships between a clinical indication and existing compounds. As an emerging paradigm in drug development, drug repurposing enables more efficient treatment of rare diseases, stratified patient populations, and urgent threats to public health. However, prioritizing well-suited drug candidates from among a nearly infinite number of repurposing options continues to represent a significant challenge in drug development. Over the past decade, advances in genomic profiling, database curation, and machine learning techniques have enabled more accurate identification of drug repurposing candidates for subsequent clinical evaluation. This review outlines the major methodologic classes that these approaches comprise, which rely on (a) protein structure, (b) genomic signatures, (c) biological networks, and (d) real-world clinical data. We propose that realizing the full impact of drug repurposing methodologies requires a multidisciplinary understanding of each method's advantages and limitations with respect to clinical practice.
Collapse
Affiliation(s)
- Henry C Cousins
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Gowri Nayar
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Russ B Altman
- Departments of Genetics, Medicine, and Bioengineering, Stanford University, Stanford, California, USA
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| |
Collapse
|
20
|
Luo Y, Mao C, Sanchez‐Pinto LN, Ahmad FS, Naidech A, Rasmussen L, Pacheco JA, Schneider D, Mithal LB, Dresden S, Holmes K, Carson M, Shah SJ, Khan S, Clare S, Wunderink RG, Liu H, Walunas T, Cooper L, Yue F, Wehbe F, Fang D, Liebovitz DM, Markl M, Michelson KN, McColley SA, Green M, Starren J, Ackermann RT, D'Aquila RT, Adams J, Lloyd‐Jones D, Chisholm RL, Kho A. Northwestern University resource and education development initiatives to advance collaborative artificial intelligence across the learning health system. Learn Health Syst 2024; 8:e10417. [PMID: 39036530 PMCID: PMC11257059 DOI: 10.1002/lrh2.10417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction The rapid development of artificial intelligence (AI) in healthcare has exposed the unmet need for growing a multidisciplinary workforce that can collaborate effectively in the learning health systems. Maximizing the synergy among multiple teams is critical for Collaborative AI in Healthcare. Methods We have developed a series of data, tools, and educational resources for cultivating the next generation of multidisciplinary workforce for Collaborative AI in Healthcare. We built bulk-natural language processing pipelines to extract structured information from clinical notes and stored them in common data models. We developed multimodal AI/machine learning (ML) tools and tutorials to enrich the toolbox of the multidisciplinary workforce to analyze multimodal healthcare data. We have created a fertile ground to cross-pollinate clinicians and AI scientists and train the next generation of AI health workforce to collaborate effectively. Results Our work has democratized access to unstructured health information, AI/ML tools and resources for healthcare, and collaborative education resources. From 2017 to 2022, this has enabled studies in multiple clinical specialties resulting in 68 peer-reviewed publications. In 2022, our cross-discipline efforts converged and institutionalized into the Center for Collaborative AI in Healthcare. Conclusions Our Collaborative AI in Healthcare initiatives has created valuable educational and practical resources. They have enabled more clinicians, scientists, and hospital administrators to successfully apply AI methods in their daily research and practice, develop closer collaborations, and advanced the institution-level learning health system.
Collapse
Affiliation(s)
- Yuan Luo
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Chengsheng Mao
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Lazaro N. Sanchez‐Pinto
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of Critical Care, Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Faraz S. Ahmad
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Andrew Naidech
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Neurocritical Care, Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Luke Rasmussen
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jennifer A. Pacheco
- Center for Genetic MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Daniel Schneider
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
| | - Leena B. Mithal
- Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Division of Infectious Diseases, Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Scott Dresden
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Emergency MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Kristi Holmes
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Galter Health Sciences LibraryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Matthew Carson
- Galter Health Sciences LibraryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Sanjiv J. Shah
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Seema Khan
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Susan Clare
- Department of SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Richard G. Wunderink
- Division of Critical Care, Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Pulmonary and Critical Care Division, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Huiping Liu
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of PharmacologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of Hematology and Oncology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Theresa Walunas
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of General Internal Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Health Information PartnershipsInstitute for Public Health and Medicine, Northwestern UniversityChicagoIllinoisUSA
- Department of Microbiology‐ImmunologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Lee Cooper
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Feng Yue
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of Biochemistry and Molecular GeneticsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Firas Wehbe
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Deyu Fang
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - David M. Liebovitz
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of General Internal Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Health Information PartnershipsInstitute for Public Health and Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Michael Markl
- Department of RadiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Kelly N. Michelson
- Division of Critical Care, Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Center for Bioethics and Medical Humanities, Institute for Public Health and MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Susanna A. McColley
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Division of Pulmonary and Sleep Medicine, Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Marianne Green
- Division of General Internal Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Justin Starren
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Ronald T. Ackermann
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Division of General Internal Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Institute for Public Health and MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Richard T. D'Aquila
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Division of Infectious Diseases, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - James Adams
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Emergency MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Donald Lloyd‐Jones
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Epidemiology, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Rex L. Chisholm
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Health Information PartnershipsInstitute for Public Health and Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Abel Kho
- Northwestern University Clinical and Translational Sciences InstituteChicagoIllinoisUSA
- Institute for Augmented Intelligence in MedicineNorthwestern UniversityChicagoIllinoisUSA
- Division of Health and Biomedical Informatics, Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Division of General Internal Medicine, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Health Information PartnershipsInstitute for Public Health and Medicine, Northwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
21
|
Hartman KM, Patel B, Rao V, Hagen AA, Saveraid HG, Fricton R, Lee S, Snyder AT, Pullen MF, Boulware DR, Liebovitz DM, Belani HK, Niklas JM, Murray TA, Cohen K, Thompson JL, Erickson SM, Bramante CT. A Comparison of Recruitment Methods for a Remote, Nationwide Clinical Trial for COVID-19 Treatment. Open Forum Infect Dis 2024; 11:ofae224. [PMID: 38947738 PMCID: PMC11214097 DOI: 10.1093/ofid/ofae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/17/2024] [Indexed: 07/02/2024] Open
Abstract
This study describes decentralized recruitment and enrollment for a COVID-19 treatment trial, while comparing 5 primary recruitment methods: search engine ads, paid advertising within a national testing company, paid advertising within a regional testing company, electronic health record messages, and word of mouth. These are compared across patient demographics, efficiency, and cost. Clinical Trials Registration NCT04510194.
Collapse
Affiliation(s)
- Katrina M Hartman
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Barkha Patel
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Via Rao
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aubrey A Hagen
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hanna G Saveraid
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Regina Fricton
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, Northwestern University, Chicago, Illinois, USA
| | - Samuel Lee
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew T Snyder
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- M Health Fairview, Minneapolis, Minnesota, USA
| | - Matthew F Pullen
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - David R Boulware
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - David M Liebovitz
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hrishikesh K Belani
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Olive View - University of California, Los Angeles, California, USA
| | - Jacinda M Niklas
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Colorado, Denver, Colorado, USA
| | - Thomas A Murray
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ken Cohen
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- UnitedHealth Group, Optum Labs, Minnetonka, Minnesota, USA
| | - Jennifer L Thompson
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Spencer M Erickson
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Carolyn T Bramante
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- General Internal Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
22
|
Song ZH, Huang QM, Xu SS, Zhou JB, Zhang C. The Effect of Antihyperglycemic Medications on COVID-19: A Meta-analysis and Systematic Review from Observational Studies. Ther Innov Regul Sci 2024; 58:773-787. [PMID: 38683419 DOI: 10.1007/s43441-024-00633-6] [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: 09/06/2023] [Accepted: 02/09/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Diabetes, a chronic disease worldwide, may be associated with a poorer prognosis in patients with coronavirus disease 2019 (COVID-19). While some antihyperglycemic medications may be beneficial, others may increase the risk of adverse clinical outcomes of COVID-19. We aimed to analyze the effect of antihyperglycemic medications on COVID-19. METHODS We searched the Web of Science, Cochrane Library, EMBASE, PubMed, and Scopus databases from December 2019 to June 2022 to identify literature related to patients with COVID-19 and type 2 diabetes mellitus (T2DM) treated with antihyperglycemic medications. RESULTS 56 studies were included in the analysis. Metformin (OR 0.66; 95% CI 0.58-0.74; p < 0.05), Glucagon-like peptide-1 receptor agonist (GLP-1ra) (OR 0.73; 95% CI 0.59-0.91; p < 0.05), and sodium-dependent glucose transporters 2 inhibitor (SGLT 2i) (OR 0.77; 95% CI 0.69-0.87; p < 0.05) were associated with lower mortality risk, while insulin was associated with increased mortality risk (OR 1.40; 95% CI 1.26-1.55; p < 0.05). Meanwhile, metformin (OR 0.65; 95% CI 0.50-0.85; p < 0.05) and GLP-1ra (OR 0.84; 95% CI 0.76-0.94; p < 0.05) were significantly associated with decreased severe manifestation risk. What's more, metformin (OR 0.77; 95% CI 0.62-0.96; p < 0.05), GLP-1ra (OR 0.86; 95% CI 0.81-0.92; p < 0.05), and SGLT 2i (OR 0.87; 95% CI 0.79-0.97; p < 0.05) were also associated with a decreased risk of hospitalization, but insulin were associated with an increased risk of hospitalization (OR 1.31; 95% CI 1.12-1.52; p < 0.05). Nevertheless, the results of the subgroup analyses showed that the effects of different glucose-lowering agents on COVID-19 may be related to in-hospital use or out-hospital use, elderly or non-elderly patients use, and different geography. CONCLUSION Metformin, GLP-1ra, and SGLT 2i have shown a positive effect on clinical outcomes in COVID-19, particularly in non-elderly individuals. However, insulin use may pose a higher risk, especially in elderly patients, so need with caution. Meanwhile, DPP-4i, TZD, α-GLUi, and sulfonylureas appeared to have a neutral effect. These results need to be validated in future clinical studies.
Collapse
Affiliation(s)
- Zhi-Hui Song
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qiao-Ming Huang
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shan-Shan Xu
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian-Bo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Chao Zhang
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
23
|
Zeng G, Li J, Wang L, Zhang Z. Association of fluvoxamine with mortality and symptom resolution among inpatients with COVID-19. Mol Psychiatry 2024:10.1038/s41380-024-02635-0. [PMID: 38907019 DOI: 10.1038/s41380-024-02635-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Affiliation(s)
- Guangting Zeng
- Department of Pharmacy, The First People's Hospital of Chenzhou, Xiangnan University, Chenzhou, China
| | - Jianqiang Li
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Linlin Wang
- Department of Pharmacy, Cancer Center(Xiamen), Fudan University, Xiamen, China
| | - Zanling Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
24
|
Shao HH, Yin RX. Pathogenic mechanisms of cardiovascular damage in COVID-19. Mol Med 2024; 30:92. [PMID: 38898389 PMCID: PMC11186295 DOI: 10.1186/s10020-024-00855-2] [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: 03/09/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND COVID-19 is a new infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Since the outbreak in December 2019, it has caused an unprecedented world pandemic, leading to a global human health crisis. Although SARS CoV-2 mainly affects the lungs, causing interstitial pneumonia and severe acute respiratory distress syndrome, a number of patients often have extensive clinical manifestations, such as gastrointestinal symptoms, cardiovascular damage and renal dysfunction. PURPOSE This review article discusses the pathogenic mechanisms of cardiovascular damage in COVID-19 patients and provides some useful suggestions for future clinical diagnosis, treatment and prevention. METHODS An English-language literature search was conducted in PubMed and Web of Science databases up to 12th April, 2024 for the terms "COVID-19", "SARS CoV-2", "cardiovascular damage", "myocardial injury", "myocarditis", "hypertension", "arrhythmia", "heart failure" and "coronary heart disease", especially update articles in 2023 and 2024. Salient medical literatures regarding the cardiovascular damage of COVID-19 were selected, extracted and synthesized. RESULTS The most common cardiovascular damage was myocarditis and pericarditis, hypertension, arrhythmia, myocardial injury and heart failure, coronary heart disease, stress cardiomyopathy, ischemic stroke, blood coagulation abnormalities, and dyslipidemia. Two important pathogenic mechanisms of the cardiovascular damage may be direct viral cytotoxicity as well as indirect hyperimmune responses of the body to SARS CoV-2 infection. CONCLUSIONS Cardiovascular damage in COVID-19 patients is common and portends a worse prognosis. Although the underlying pathophysiological mechanisms of cardiovascular damage related to COVID-19 are not completely clear, two important pathogenic mechanisms of cardiovascular damage may be the direct damage of the SARSCoV-2 infection and the indirect hyperimmune responses.
Collapse
Affiliation(s)
- Hong-Hua Shao
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Rui-Xing Yin
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China.
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
25
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Prasanth MI, Wannigama DL, Reiersen AM, Thitilertdecha P, Prasansuklab A, Tencomnao T, Brimson S, Brimson JM. A systematic review and meta-analysis, investigating dose and time of fluvoxamine treatment efficacy for COVID-19 clinical deterioration, death, and Long-COVID complications. Sci Rep 2024; 14:13462. [PMID: 38862591 PMCID: PMC11166997 DOI: 10.1038/s41598-024-64260-9] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
There have been 774,075,242 cases of COVID-19 and 7,012,986 deaths worldwide as of January 2024. In the early stages of the pandemic, there was an urgent need to reduce the severity of the disease and prevent the need for hospitalization to avoid stress on healthcare systems worldwide. The repurposing of drugs to prevent clinical deterioration of COVID-19 patients was trialed in many studies using many different drugs. Fluvoxamine (an SSRI and sigma-1 receptor agonist) was initially identified to potentially provide beneficial effects in COVID-19-infected patients, preventing clinical deterioration and the need for hospitalization. Fourteen clinical studies have been carried out to date, with seven of those being randomized placebo-controlled studies. This systematic review and meta-analysis covers the literature from the outbreak of SARS-CoV-2 in late 2019 until January 2024. Search terms related to fluvoxamine, such as its trade names and chemical names, along with words related to COVID-19, such as SARS-CoV-2 and coronavirus, were used in literature databases including PubMed, Google Scholar, Scopus, and the ClinicalTrials.gov database from NIH, to identify the trials used in the subsequent analysis. Clinical deterioration and death data were extracted from these studies where available and used in the meta-analysis. A total of 7153 patients were studied across 14 studies (both open-label and double-blind placebo-controlled). 681 out of 3553 (19.17%) in the standard care group and 255 out of 3600 (7.08%) in the fluvoxamine-treated group experienced clinical deterioration. The estimated average log odds ratio was 1.087 (95% CI 0.200 to 1.973), which differed significantly from zero (z = 2.402, p = 0.016). The seven placebo-controlled studies resulted in a log odds ratio of 0.359 (95% CI 0.1111 to 0.5294), which differed significantly from zero (z = 3.103, p = 0.002). The results of this study identified fluvoxamine as effective in preventing clinical deterioration, and subgrouping analysis suggests that earlier treatment with a dose of 200 mg or above provides the best outcomes. We hope the outcomes of this study can help design future studies into respiratory viral infections and potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Department of Microbiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Thai Red Cross Society, Bangkok, Thailand
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Angela Michelle Reiersen
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Rama 1 Road, Pathumwan, Wang Mai, Bangkok, 10330, Thailand.
| |
Collapse
|
27
|
Chaudhary S, Kulkarni A. Metformin: Past, Present, and Future. Curr Diab Rep 2024; 24:119-130. [PMID: 38568468 DOI: 10.1007/s11892-024-01539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW This review provides the most recent update of metformin, a biguanide oral antihyperglycemic drug used as a first-line treatment in type 2 diabetes mellitus. RECENT FINDINGS Metformin continues to dominate in the world of antidiabetics, and its use will continue to rise because of its high efficiency and easy availability. Apart from type 2 diabetes, research is exploring its potential in other conditions such as cancer, memory loss, bone disorders, immunological diseases, and aging. Metformin is the most prescribed oral antidiabetic worldwide. It has been in practical use for the last six decades and continues to be the preferred drug for newly diagnosed type 2 diabetes mellitus. It reduces glucose levels by decreasing hepatic glucose production, reducing intestinal glucose absorption, and increasing insulin sensitivity. It can be used as monotherapy or combined with other antidiabetics like sulfonylureas, DPP-4 inhibitors, SGLT-2 inhibitors, or insulin, improving its efficacy. Metformin can be used once or twice daily, depending on requirements. Prolonged usage of metformin may lead to abdominal discomfort, deficiency of Vitamin B12, or lactic acidosis. It should be used carefully in patients with renal impairment. Recent studies have explored additional benefits of metformin in polycystic ovarian disease, gestational diabetes mellitus, cognitive disorders, and immunological diseases. However, more extensive studies are needed to confirm these additional benefits.
Collapse
|
28
|
Qiu S, Hubbard AE, Gutiérrez JP, Pimpale G, Juárez-Flores A, Ghosh R, de Jesús Ascencio-Montiel I, Bertozzi SM. Estimating the effect of realistic improvements of metformin adherence on COVID-19 mortality using targeted machine learning. GLOBAL EPIDEMIOLOGY 2024; 7:100142. [PMID: 38590914 PMCID: PMC10999684 DOI: 10.1016/j.gloepi.2024.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
Background Type 2 diabetes elevates the risk of severe outcomes in COVID-19 patients, with multiple studies reporting higher case fatality rates. Metformin is a widely used medication for glycemic management. We hypothesize that improved adherence to metformin may lower COVID-19 post-infection mortality risk in this group. Utilizing data from the Mexican Social Security Institute (IMSS), we investigate the relationship between metformin adherence and mortality following COVID-19 infection in patients with chronic metformin prescriptions. Methods This is a retrospective cohort study consisting of 61,180 IMSS beneficiaries who received a positive polymerase chain reaction (PCR) or rapid test for SARS-CoV-2 and had at least two consecutive months of metformin prescriptions prior to the positive test. The hypothetical intervention is improved adherence to metformin, measured by proportion of days covered (PDC), with the comparison being the observed metformin adherence values. The primary outcome is all-cause mortality following COVID-19 infection. We defined the causal parameter using shift intervention, an example of modified treatment policies. We used the targeted learning framework for estimation of the target estimand. Findings Among COVID-19 positive patients with chronic metformin prescriptions, we found that a 5% and 10% absolute increase in metformin adherence is associated with a respective 0.26% (95% CI: -0.28%, 0.79%) and 1.26% (95% CI: 0.72%, 1.80%) absolute decrease in mortality risk. Interpretation Subject to the limitations of a real-world data study, our results indicate a causal association between improved metformin adherence and reduced COVID-19 post-infection mortality risk.
Collapse
Affiliation(s)
- Sky Qiu
- University of California, School of Public Health, Berkeley, CA, USA
| | - Alan E. Hubbard
- University of California, School of Public Health, Berkeley, CA, USA
| | - Juan Pablo Gutiérrez
- Center for Policy, Population and Health Research, School of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ganesh Pimpale
- University of California, Department of Mechanical Engineering, Berkeley, CA, USA
| | - Arturo Juárez-Flores
- Center for Policy, Population and Health Research, School of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rakesh Ghosh
- Institute for Global Health Sciences, University of California, San Francisco, CA, USA
| | | | - Stefano M. Bertozzi
- University of California, School of Public Health, Berkeley, CA, USA
- University of Washington, School of Public Health, Seattle, WA, USA
- Instituto Nacional de Salud Pública, Cuernavaca, MOR, Mexico
| |
Collapse
|
29
|
Mikamo H, Takahashi S, Yamagishi Y, Hirakawa A, Harada T, Nagashima H, Noguchi C, Masuko K, Maekawa H, Kashii T, Ohbayashi H, Hosokawa S, Maejima K, Yamato M, Manosuthi W, Paiboonpol S, Suganami H, Tanigawa R, Kawamura H. Efficacy and safety of ivermectin in patients with mild COVID-19 in Japan and Thailand. J Infect Chemother 2024; 30:536-543. [PMID: 38154616 DOI: 10.1016/j.jiac.2023.12.012] [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: 10/24/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Ivermectin is an antiparasitic drug administered to hundreds of millions of people worldwide. Fundamental research suggests that ivermectin is effective against coronavirus disease 2019 (COVID-19); therefore, we investigated the efficacy and safety of ivermectin as a COVID-19 treatment option. METHODS This multi-regional (Japan and Thailand), multicenter, placebo-controlled, randomized, double-blind, parallel-group, Phase III study evaluated the efficacy and safety of ivermectin in patients with mild COVID-19 (IVERMILCO Study). The participants took a specified number of the investigational product (ivermectin or placebo) tablets of, adjusted to a dose of 0.3-0.4 mg/kg, orally on an empty stomach once daily for three days. The primary efficacy endpoint was the time at which clinical symptoms first showed an improving trend by 168 h after investigational product administration. RESULTS A total of 1030 eligible participants were assigned to receive the investigational product; 502 participants received ivermectin and 527 participants received a placebo. The primary efficacy endpoint was approximately 96 h (approximately four days) for both ivermectin and placebo groups, which did not show statistically significant difference (stratified log-rank test, p = 0.61). The incidence of adverse events and adverse drug reactions did not show statistically significant differences between the ivermectin and placebo groups (chi-square test, p = 0.97, p = 0.59). CONCLUSIONS The results show that ivermectin (0.3-0.4 mg/kg), as a treatment for patients with mild COVID-19, is ineffective; however, its safety has been confirmed for participants, including minor participants of 12 years or older (IVERMILCO Study ClinicalTrials.gov number, NCT05056883.).
Collapse
Affiliation(s)
- Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, 1-1, Yazakokarimata Nagakute-shi, Aichi, 480-1195, Japan.
| | - Satoshi Takahashi
- Department of Infection Control and Laboratory Medicine, Sapporo Medical University School of Medicine, 16-291, Minami1-jonishi, Chuo-ku, Sapporo-shi, Hokkaido, 060-8543, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Kochi Medical School, Kochi University, 185-1, Okocho-Kohasu, Nankoku-shi, Kochi, 783-8505, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Toshiyuki Harada
- Center for Respiratory Diseases, Department of Pulmonary Medicine, Japan Community Healthcare Organization Hokkaido Hospital, 1-8-3-18, Nakanoshima, Toyohira-ku, Sapporo, Hokkaido, 062-8618, Japan
| | | | - Chiaki Noguchi
- Sumida General Clinic, 3-4-8, Taihei, Sumida-ku, Tokyo, 130-0012, Japan
| | - Kentaro Masuko
- Shimamura Memorial Hospital, 2-4-1, Sekimachi-Kita, Nerima-ku, Tokyo, 177-0051, Japan
| | - Hiromitsu Maekawa
- Maekawa Medical Clinic, 1-1-8, Kitasaiwai, Nishi-ku, Yokohama-shi, Kanagawa, 220-0004, Japan
| | - Tatsuhiko Kashii
- Department of Oncology, Japan Organization of Occupational Health and Safety Toyama Rosai Hospital, 992, Rokuromaru, Uozu-shi, Toyama, 937-0042, Japan
| | - Hiroyuki Ohbayashi
- Department of Allergy and Respiratory Medicine, Tohno Chuo Clinic, 1-14-1, Matsugasecho, Mizunami-shi, Gifu, 509-6134, Japan
| | - Shinichiro Hosokawa
- Hosokawa Surgical Clinic, 1-75-2, Nishikomenocho, Nakamura-ku, Nagoya-shi, Aichi, 453-0812, Japan
| | - Katsuyuki Maejima
- Department of Internal Medicine, Diabetes Medicine, Maejima Clinic, 1-15-1, Midoricho, Showa-ku, Nagoya-shi, Aichi, 466-0013, Japan
| | - Masaya Yamato
- Department of General Internal Medicine, Infectious Disease, Rinku General Medical Center, 2-23, Rinku-Oraikita, Izumisano-shi, Osaka, 598-8577, Japan
| | - Weerawat Manosuthi
- Bamrasnaradura Infectious Diseases Institute, Ministry of Public Health, 38 Moo 4, Talat Khwan, Tiwanon Road, Mueang District, Nonthaburi 11000, Thailand
| | - Supachai Paiboonpol
- Department of Medicine, Ratchaburi Hospital, 85 Somboonkul Road, Na Mueang Subdistrict Muang District, Ratchaburi Provinc 70000, Thailand
| | - Hideki Suganami
- Global Data Science Center, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| | - Ryohei Tanigawa
- Global Clinical Development Department, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| | - Hitoshi Kawamura
- Medical Writing Department, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| |
Collapse
|
30
|
Olawore O, Turner LE, Evans MD, Johnson SG, Huling JD, Bramante CT, Buse JB, Stürmer T. Risk of Post-Acute Sequelae of SARS-CoV-2 Infection (PASC) Among Patients with Type 2 Diabetes Mellitus on Anti-Hyperglycemic Medications. Clin Epidemiol 2024; 16:379-393. [PMID: 38836048 PMCID: PMC11149650 DOI: 10.2147/clep.s458901] [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: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 06/06/2024] Open
Abstract
Background Observed activity of metformin in reducing the risk of severe COVID-19 suggests a potential use of the anti-hyperglycemic in the prevention of post-acute sequelae of SARS-CoV-2 infection (PASC). We assessed the 3-month and 6-month risk of PASC among patients with type 2 diabetes mellitus (T2DM) comparing metformin users to sulfonylureas (SU) or dipeptidyl peptidase-4 inhibitors (DPP4i) users. Methods We used de-identified patient level electronic health record data from the National Covid Cohort Collaborative (N3C) between October 2021 and April 2023. Participants were adults ≥ 18 years with T2DM who had at least one outpatient healthcare encounter in health institutions in the United States prior to COVID-19 diagnosis. The outcome of PASC was defined based on the presence of a diagnosis code for the illness or using a predicted probability based on a machine learning algorithm. We estimated the 3-month and 6-month risk of PASC and calculated crude and weighted risk ratios (RR), risk differences (RD), and differences in mean predicted probability. Results We identified 5596 (mean age: 61.1 years; SD: 12.6) and 1451 (mean age: 64.9 years; SD 12.5) eligible prevalent users of metformin and SU/DPP4i respectively. We did not find a significant difference in risk of PASC at 3 months (RR = 0.86 [0.56; 1.32], RD = -3.06 per 1000 [-12.14; 6.01]), or at 6 months (RR = 0.81 [0.55; 1.20], RD = -4.91 per 1000 [-14.75, 4.93]) comparing prevalent users of metformin to prevalent users of SU/ DPP4i. Similar observations were made for the outcome definition using the ML algorithm. Conclusion The observed estimates in our study are consistent with a reduced risk of PASC among prevalent users of metformin, however the uncertainty of our confidence intervals warrants cautious interpretations of the results. A standardized clinical definition of PASC is warranted for thorough evaluation of the effectiveness of therapies under assessment for the prevention of PASC.
Collapse
Affiliation(s)
- Oluwasolape Olawore
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lindsey E Turner
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN, USA
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, USA
| | - Michael D Evans
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, USA
| | - Steven G Johnson
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Jared D Huling
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Carolyn T Bramante
- Division of General Internal Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - John B Buse
- Division of Endocrinology, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Til Stürmer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - On behalf of the N3C Consortium
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN, USA
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, USA
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
- Division of General Internal Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Endocrinology, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
31
|
Lawson CA, Moss AJ, Arnold JR, Bagot C, Banerjee A, Berry C, Greenwood J, Hughes AD, Khunti K, Mills NL, Neubauer S, Raman B, Sattar N, Leavy OC, Richardson M, Elneima O, McAuley HJ, Shikotra A, Singapuri A, Sereno M, Saunders R, Harris V, Houchen-Wolloff L, Greening NJ, Harrison E, Docherty AB, Lone NI, Quint JK, Chalmers J, Ho LP, Horsley A, Marks M, Poinasamy K, Evans R, Wain LV, Brightling C, McCann GP. Long COVID and cardiovascular disease: a prospective cohort study. Open Heart 2024; 11:e002662. [PMID: 38802280 PMCID: PMC11131117 DOI: 10.1136/openhrt-2024-002662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Pre-existing cardiovascular disease (CVD) or cardiovascular risk factors have been associated with an increased risk of complications following hospitalisation with COVID-19, but their impact on the rate of recovery following discharge is not known. OBJECTIVES To determine whether the rate of patient-perceived recovery following hospitalisation with COVID-19 was affected by the presence of CVD or cardiovascular risk factors. METHODS In a multicentre prospective cohort study, patients were recruited following discharge from the hospital with COVID-19 undertaking two comprehensive assessments at 5 months and 12 months. Patients were stratified by the presence of either CVD or cardiovascular risk factors prior to hospitalisation with COVID-19 and compared with controls with neither. Full recovery was determined by the response to a patient-perceived evaluation of full recovery from COVID-19 in the context of physical, physiological and cognitive determinants of health. RESULTS From a total population of 2545 patients (38.8% women), 472 (18.5%) and 1355 (53.2%) had CVD or cardiovascular risk factors, respectively. Compared with controls (n=718), patients with CVD and cardiovascular risk factors were older and more likely to have had severe COVID-19. Full recovery was significantly lower at 12 months in patients with CVD (adjusted OR (aOR) 0.62, 95% CI 0.43 to 0.89) and cardiovascular risk factors (aOR 0.66, 95% CI 0.50 to 0.86). CONCLUSION Patients with CVD or cardiovascular risk factors had a delayed recovery at 12 months following hospitalisation with COVID-19. Targeted interventions to reduce the impact of COVID-19 in patients with cardiovascular disease remain an unmet need. TRAIL REGISTRATION NUMBER ISRCTN10980107.
Collapse
Affiliation(s)
| | - Alastair James Moss
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | | | - Catherine Bagot
- Department of Haemostasis and Thrombosis, Glasgow Royal Infirmary, Glasgow, Glasgow, UK
| | - Amitava Banerjee
- Farr Institute of Health Informatics Research, University College London, London, UK
| | - Colin Berry
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
- Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | - John Greenwood
- Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Alun D Hughes
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Kamlesh Khunti
- Leicester Real World Evidence Unit, Leicester Diabetes Centre, University of Leicester, Leicester, UK
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, Edinburgh, UK
| | - Stefan Neubauer
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Betty Raman
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Olivia C Leavy
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Michael Marks
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Rachael Evans
- The Institute for Lung Health, University of Leicester, Leicester, UK
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Chris Brightling
- The Institute for Lung Health, University of Leicester, Leicester, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, Glenfield Hospital, Leicester, UK
| |
Collapse
|
32
|
Patel S, Visotcky A, Devine A, Kode V, Kotlo S, Aljadah M, Sparapani R, Kulinski J. Prevalence, Predictors, and Outcomes of Type 2 NSTEMI in Hospitalized Patients With COVID-19. J Am Heart Assoc 2024; 13:e032572. [PMID: 38726904 PMCID: PMC11179823 DOI: 10.1161/jaha.123.032572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Data on the incidence of type 2 non-ST-segment-elevation myocardial infarction (T2MI) in hospitalized patients with COVID-19 has been limited to single-center studies. Given that certain characteristics, such as obesity and type 2 diabetes, have been associated with higher mortality in COVID-19 infections, we aimed to define the incidence of T2MI in a national cohort and identify pre-hospital patient characteristics associated with T2MI in hospitalized patients with COVID-19. METHODS AND RESULTS Using the national American Heart Association COVID-19 Cardiovascular Disease Quality Improvement Registry, we performed a retrospective 4:1 matched (age, sex, race, and body mass index) analysis of controls versus cases with T2MI. We performed (1) conditional multivariable logistic regression to identify predictive pre-hospital patient characteristics of T2MI for patients hospitalized with COVID-19 and (2) stratified proportional hazards regression to investigate the association of T2MI with morbidity and mortality. From January 2020 through May 2021, there were 709 (2.2%) out of 32 015 patients with T2MI. Five hundred seventy-nine cases with T2MI were matched to 2171 controls (mean age 70; 43% female). Known coronary artery disease, heart failure, chronic kidney disease, hypertension, payor source, and presenting heart rate were associated with higher odds of T2MI. Anti-hyperglycemic medication and anti-coagulation use before admission were associated with lower odds of T2MI. Those with T2MI had higher morbidity and mortality (hazard ratio, 1.40 [95% CI, 1.13-1.74]; P=0.002). CONCLUSIONS In hospitalized patients with COVID-19, those with a T2MI compared with those without had higher morbidity and mortality. Outpatient anti-hyperglycemic and anti-coagulation use were the only pre-admission factors associated with reduced odds of T2MI.
Collapse
Affiliation(s)
- Sahishnu Patel
- Division of Cardiovascular Medicine Rush University Medical Center Chicago IL USA
| | - Alexis Visotcky
- Division of Biostatistics Medical College of Wisconsin Milwaukee WI USA
| | - Adam Devine
- Division of Cardiovascular Medicine University of Minnesota Minneapolis MN USA
| | - Vishwajit Kode
- Department of Medicine California Pacific Medical Center San Francisco CA USA
| | - Srisha Kotlo
- Department of Medicine University of Chicago Chicago IL USA
| | - Michael Aljadah
- Division of Cardiovascular Medicine University of Minnesota Minneapolis MN USA
| | - Rodney Sparapani
- Division of Biostatistics Medical College of Wisconsin Milwaukee WI USA
| | - Jacquelyn Kulinski
- Division of Cardiovascular Medicine Medical College of Wisconsin Milwaukee WI USA
| |
Collapse
|
33
|
Xu K, He W, Yu B, Zhong K, Zhou D, Wang DW. Effects of different treatments for type 2 diabetes mellitus on mortality of coronavirus disease from 2019 to 2021 in China: a multi-institutional retrospective study. MOLECULAR BIOMEDICINE 2024; 5:18. [PMID: 38755442 PMCID: PMC11099001 DOI: 10.1186/s43556-024-00183-1] [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: 11/27/2023] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
The coronavirus disease (COVID-19) pandemic has continued for 5 years. Sporadic cases continue to occur in different locations. Type 2 diabetes mellitus (T2DM) is associated with a high risk of a poor prognosis in patients with COVID-19. Successful control of blood glucose levels can effectively decrease the risks of severe infections and mortality. However, the effects of different treatments were reported differently and even adversely. This retrospective study included 4,922 patients who have been diagnosed as COVID-19 and T2DM from 138 Hubei hospitals. The clinical characteristics and outcomes were compared and calculated their risk for death using multivariate Cox regression and Kaplan-Meier curves. After adjustment of age, sex, comorbidities, and in-hospital medications, metformin and alpha-glucosidase inhibitor (AGI) use performed lower all-cause mortality (adjusted hazard ratio [HR], 0.41; 95% confidence interval [CI]: 0.24-0.71; p = 0.001 for metformin; 0.53, 0.35-0.80, p = 0.002 for AGIs), while insulin use was associated with increased all-cause mortality (adjusted HR, 2.07, 95% CI, 1.61-2.67, p < 0.001). After propensity score-matched (PSM) analysis, adjusted HRs for insulin, metformin, and AGIs associated with all-cause mortality were 1.32 (95% CI, 1.03-1.81; p = 0.012), 0.48 (95% CI, 0.23-0.83, p = 0.014), and 0.59 (95% CI, 0.35-0.98, p = 0.05). Therefore, metformin and AGIs might be more suitable for patients with COVID-19 and T2DM while insulin might be used with caution.
Collapse
Affiliation(s)
- Ke Xu
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Bo Yu
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China
| | - Kaineng Zhong
- Hubei Provincial Health Commission, Wuhan, 430079, China
| | - Da Zhou
- Hubei Provincial Health Commission, Wuhan, 430079, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
34
|
Zhou Q, Zhao G, Pan Y, Zhang Y, Ni Y. The efficacy and safety of fluvoxamine in patients with COVID-19: A systematic review and meta-analysis from randomized controlled trials. PLoS One 2024; 19:e0300512. [PMID: 38753761 PMCID: PMC11098472 DOI: 10.1371/journal.pone.0300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/29/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Recently, several randomized controlled trials (RCTs) of fluvoxamine have been successfully conducted for the treatment of patients with coronavirus disease 2019 (COVID-19). This systematic review and meta-analysis was to evaluate the efficacy and safety of fluvoxamine in patients with COVID-19. METHODS MEDLINE, EMBASE, Cochrane Library and clinicaltrials.gov were searched for RCTs which were performed to evaluate fluvoxamine and placebo up to January 31, 2024. Review Manager 5.3 was used to perform meta-analysis. The risk ratio (RR) and mean difference (MD) was analyzed and calculated with a random effect model. RESULTS We pooled 4,711 participants from six RCTs (2,382 in the fluvoxamine group and 2,329 in the placebo group). Compared to the placebo group, the fluvoxamine group had a significantly lower rate of clinical deterioration (RR, 0.73; P = 0.004; 95% CI, 0.59 to 0.90; I2 = 0%) and hospitalization (RR, 0.76; P = 0.04; 95% CI, 0.59 to 0.99; I2 = 0%). In the meantime, compared with the placebo group, fluvoxamine group did not show any higher risk of AEs (P = 0.13 and 0.91, respectively) in safety outcomes analysis. The subgroup analysis showed that fluvoxamine treatment performed more than 200 mg daily appears to be more effective than those performed less than 200 mg daily in reducing clinical deterioration and hospitalization risks, while not exhibiting higher AE and SAE risks than placebo group. CONCLUSION Fluvoxamine for patients with COVID-19, especially those who take 200 mg or more daily, is superior to the placebo group in reducing clinical deterioration and hospitalization, and did not show any higher risk of AEs and SAEs in safety concerns, which might be a promising intervention for COVID-19.
Collapse
Affiliation(s)
- Qiufeng Zhou
- Department of Neurosurgery, Suzhou Ninth People’s Hospital, Suzhou, Jiangsu Province, China
| | - Guozheng Zhao
- Department of Neurosurgery, Suzhou Ninth People’s Hospital, Suzhou, Jiangsu Province, China
| | - Yu Pan
- Department of Emergency, Suzhou Ninth People’s Hospital, Suzhou, Jiangsu Province, China
| | - Ying Zhang
- Department of Emergency, Suzhou Ninth People’s Hospital, Suzhou, Jiangsu Province, China
| | - Yuehua Ni
- Department of Emergency, Suzhou Ninth People’s Hospital, Suzhou, Jiangsu Province, China
| |
Collapse
|
35
|
Singh S, Boyd S, Schilling WHK, Watson JA, Mukaka M, White NJ. The relationship between viral clearance rates and disease progression in early symptomatic COVID-19: a systematic review and meta-regression analysis. J Antimicrob Chemother 2024; 79:935-945. [PMID: 38385479 PMCID: PMC11062948 DOI: 10.1093/jac/dkae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Effective antiviral drugs accelerate viral clearance in acute COVID-19 infections; the relationship between accelerating viral clearance and reducing severe clinical outcomes is unclear. METHODS A systematic review was conducted of randomized controlled trials (RCTs) of antiviral therapies in early symptomatic COVID-19, where viral clearance data were available. Treatment benefit was defined clinically as the relative risk of hospitalization/death during follow-up (≥14 days), and virologically as the SARS-CoV-2 viral clearance rate ratio (VCRR). The VCRR is the ratio of viral clearance rates between the intervention and control arms. The relationship between the clinical and virological treatment effects was assessed by mixed-effects meta-regression. RESULTS From 57 potentially eligible RCTs, VCRRs were derived for 44 (52 384 participants); 32 had ≥1 clinical endpoint in each arm. Overall, 9.7% (R2) of the variation in clinical benefit was explained by variation in VCRRs with an estimated linear coefficient of -0.92 (95% CI: -1.99 to 0.13; P = 0.08). However, this estimate was highly sensitive to the inclusion of the recent very large PANORAMIC trial. Omitting this outlier, half the variation in clinical benefit (R2 = 50.4%) was explained by variation in VCRRs [slope -1.47 (95% CI -2.43 to -0.51); P = 0.003], i.e. higher VCRRs were associated with an increased clinical benefit. CONCLUSION Methods of determining viral clearance in COVID-19 studies and the relationship to clinical outcomes vary greatly. As prohibitively large sample sizes are now required to show clinical treatment benefit in antiviral therapeutic assessments, viral clearance is a reasonable surrogate endpoint.
Collapse
Affiliation(s)
- Shivani Singh
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Simon Boyd
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - William H K Schilling
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - James A Watson
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
- Biostatistics Department, Oxford University Clinical Research Unit, 764 Vo Van Kiet, Quan 5, Ho Chi Minh City, Vietnam
| | - Mavuto Mukaka
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - Nicholas J White
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| |
Collapse
|
36
|
Huang X, Liu X, Li Z. Bile acids and coronavirus disease 2019. Acta Pharm Sin B 2024; 14:1939-1950. [PMID: 38799626 PMCID: PMC11119507 DOI: 10.1016/j.apsb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/08/2023] [Accepted: 01/28/2024] [Indexed: 05/29/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been significantly alleviated. However, long-term health effects and prevention strategy remain unresolved. Thus, it is essential to explore the pathophysiological mechanisms and intervention for SARS-CoV-2 infection. Emerging research indicates a link between COVID-19 and bile acids, traditionally known for facilitating dietary fat absorption. The bile acid ursodeoxycholic acid potentially protects against SARS-CoV-2 infection by inhibiting the farnesoid X receptor, a bile acid nuclear receptor. The activation of G-protein-coupled bile acid receptor, another membrane receptor for bile acids, has also been found to regulate the expression of angiotensin-converting enzyme 2, the receptor through which the virus enters human cells. Here, we review the latest basic and clinical evidence linking bile acids to SARS-CoV-2, and reveal their complicated pathophysiological mechanisms.
Collapse
Affiliation(s)
- Xiaoru Huang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
- Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing 100191, China
| | - Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
- Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
- Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
37
|
Siripongboonsitti T, Tawinprai K, Avirutnan P, Jitobaom K, Auewarakul P. A randomized trial to assess the acceleration of viral clearance by the combination Favipiravir/Ivermectin/Niclosamide in mild-to-moderate COVID-19 adult patients (FINCOV). J Infect Public Health 2024; 17:897-905. [PMID: 38569269 DOI: 10.1016/j.jiph.2024.03.030] [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: 11/20/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND The efficacy of the viral clearance and clinical outcomes of favipiravir (FPV) in outpatients being treated for coronavirus disease 2019 (COVID-19) is unclear. Ivermectin (IVM), niclosamide (NCL), and FPV demonstrated synergistic effects in vitro for exceed 78% inhibiting severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) replication. METHODS A phase 2, open-label, 1:1, randomized, controlled trial was conducted on Thai patients with mild-to-moderate COVID-19 who received either combination FPV/IVM/NCL therapy or FPV alone to assess the rate of viral clearance among individuals with mild-to-moderate COVID-19. RESULTS Sixty non-high-risk comorbid patients with mild-to-moderate COVID-19 were randomized; 30 received FPV/IVM/NCL, and 30 received FPV alone. Mixed-effects multiple linear regression analysis of the cycle threshold value from SARS-CoV-2 PCR demonstrated no statistically significant differences in viral clearance rates between the combined FPV/IVM/NCL therapy group and the FPV-alone group. World Health Organization Clinical Progression scores and symptomatic improvement did not differ between arms on days 3, 6, and 10, and no adverse events were reported. No patients required hospitalization, intensive care unit admission, or supplemental oxygen or died within 28 days. C-reactive protein on day 3 was lower in the FPV/IVM/NCL group. CONCLUSION Viral clearance rates did not differ significantly between the FPV/IVM/NCL combination therapy and FPV-alone groups of individuals with mild-to-moderate COVID-19, although the combined regimen demonstrated a synergistic effect in vitro. No discernible clinical benefit was observed. Further research is required to explore the potential benefits of FVP beyond its antiviral effects. TRIAL REGISTRATION TCTR20230403007, Registered 3 April 2023 - Retrospectively registered,https://trialsearch.who.int/Trial2.aspx?TrialID=TCTR20230403007.
Collapse
Affiliation(s)
- Taweegrit Siripongboonsitti
- Division of Infectious Diseases, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand; Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand.
| | - Kriangkrai Tawinprai
- Division of Infectious Diseases, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand; Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Panisadee Avirutnan
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kunlakanya Jitobaom
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Prasert Auewarakul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
38
|
Hurwitz SJ, De R, LeCher JC, Downs-Bowen JA, Goh SL, Zandi K, McBrayer T, Amblard F, Patel D, Kohler JJ, Bhasin M, Dobosh BS, Sukhatme V, Tirouvanziam RM, Schinazi RF. Why Certain Repurposed Drugs Are Unlikely to Be Effective Antivirals to Treat SARS-CoV-2 Infections. Viruses 2024; 16:651. [PMID: 38675992 PMCID: PMC11053489 DOI: 10.3390/v16040651] [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: 03/08/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Most repurposed drugs have proved ineffective for treating COVID-19. We evaluated median effective and toxic concentrations (EC50, CC50) of 49 drugs, mostly from previous clinical trials, in Vero cells. Ratios of reported unbound peak plasma concentrations, (Cmax)/EC50, were used to predict the potential in vivo efficacy. The 20 drugs with the highest ratios were retested in human Calu-3 and Caco-2 cells, and their CC50 was determined in an expanded panel of cell lines. Many of the 20 drugs with the highest ratios were inactive in human Calu-3 and Caco-2 cells. Antivirals effective in controlled clinical trials had unbound Cmax/EC50 ≥ 6.8 in Calu-3 or Caco-2 cells. EC50 of nucleoside analogs were cell dependent. This approach and earlier availability of more relevant cultures could have reduced the number of unwarranted clinical trials.
Collapse
Affiliation(s)
- Selwyn J. Hurwitz
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Ramyani De
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Julia C. LeCher
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Jessica A. Downs-Bowen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Shu Ling Goh
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Keivan Zandi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Tamara McBrayer
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Dharmeshkumar Patel
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - James J. Kohler
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Manoj Bhasin
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Brian S. Dobosh
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Vikas Sukhatme
- Morningside Center for Innovative and Affordable Medicine, Departments of Medicine and Hematology and Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Rabindra M. Tirouvanziam
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Raymond F. Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| |
Collapse
|
39
|
Meyerowitz EA, Li Y. Review: The Landscape of Antiviral Therapy for COVID-19 in the Era of Widespread Population Immunity and Omicron-Lineage Viruses. Clin Infect Dis 2024; 78:908-917. [PMID: 37949817 PMCID: PMC11487108 DOI: 10.1093/cid/ciad685] [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: 09/11/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
The goals of coronavirus disease 2019 (COVID-19) antiviral therapy early in the pandemic were to prevent severe disease, hospitalization, and death. As these outcomes have become infrequent in the age of widespread population immunity, the objectives have shifted. For the general population, COVID-19-directed antiviral therapy should decrease symptom severity and duration and minimize infectiousness, and for immunocompromised individuals, antiviral therapy should reduce severe outcomes and persistent infection. The increased recognition of virologic rebound following ritonavir-boosted nirmatrelvir (NMV/r) and the lack of randomized controlled trial data showing benefit of antiviral therapy for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection for standard-risk, vaccinated individuals remain major knowledge gaps. Here, we review data for selected antiviral agents and immunomodulators currently available or in late-stage clinical trials for use in outpatients. We do not review antibody products, convalescent plasma, systemic corticosteroids, IL-6 inhibitors, Janus kinase inhibitors, or agents that lack Food and Drug Administration approval or emergency use authorization or are not appropriate for outpatients.
Collapse
Affiliation(s)
- Eric A Meyerowitz
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Yijia Li
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
40
|
Massoumi LE, Rosenbaum A. Case Report on High Dose Lithium Treatment for Post-COVID Depression, Recurrent Fevers, and Skin Lesions. PSYCHOPHARMACOLOGY BULLETIN 2024; 54:39-45. [PMID: 38601833 PMCID: PMC11003256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
This is a case of a 35-year-old woman who presented with an 18-month history of post (long)-COVID depression and exhaustion along with recurrent fevers and treatment-resistant skin boils, all of which abated with lithium treatment at a serum level of 1.14 mmol/L, and all of which worsened when the lithium serum level was lowered to 0.8. This paper illustrates Lithium's effectiveness in the treatment of post (long)-COVID syndrome, though a higher serum concentration may be required.
Collapse
Affiliation(s)
- Lila Elizabeth Massoumi
- Massoumi, MD, Michigan State University, Integrative Psychiatry Services, PC, Bingham Farms, MI
| | - Alan Rosenbaum
- Rosenbaum, MD, Wayne State University, West Bloomfield, MI
| |
Collapse
|
41
|
Hayward G, Yu LM, Little P, Gbinigie O, Shanyinde M, Harris V, Dorward J, Saville BR, Berry N, Evans PH, Thomas NPB, Patel MG, Richards D, Hecke OV, Detry MA, Saunders C, Fitzgerald M, Robinson J, Latimer-Bell C, Allen J, Ogburn E, Grabey J, de Lusignan S, Hobbs FR, Butler CC. Ivermectin for COVID-19 in adults in the community (PRINCIPLE): An open, randomised, controlled, adaptive platform trial of short- and longer-term outcomes. J Infect 2024; 88:106130. [PMID: 38431155 PMCID: PMC10981761 DOI: 10.1016/j.jinf.2024.106130] [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: 10/26/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND The evidence for whether ivermectin impacts recovery, hospital admissions, and longer-term outcomes in COVID-19 is contested. The WHO recommends its use only in the context of clinical trials. METHODS In this multicentre, open-label, multi-arm, adaptive platform randomised controlled trial, we included participants aged ≥18 years in the community, with a positive SARS-CoV-2 test, and symptoms lasting ≤14 days. Participants were randomised to usual care, usual care plus ivermectin tablets (target 300-400 μg/kg per dose, once daily for 3 days), or usual care plus other interventions. Co-primary endpoints were time to first self-reported recovery, and COVID-19 related hospitalisation/death within 28 days, analysed using Bayesian models. Recovery at 6 months was the primary, longer term outcome. TRIAL REGISTRATION ISRCTN86534580. FINDINGS The primary analysis included 8811 SARS-CoV-2 positive participants (median symptom duration 5 days), randomised to ivermectin (n = 2157), usual care (n = 3256), and other treatments (n = 3398) from June 23, 2021 to July 1, 2022. Time to self-reported recovery was shorter in the ivermectin group compared with usual care (hazard ratio 1·15 [95% Bayesian credible interval, 1·07 to 1·23], median decrease 2.06 days [1·00 to 3·06]), probability of meaningful effect (pre-specified hazard ratio ≥1.2) 0·192). COVID-19-related hospitalisations/deaths (odds ratio 1·02 [0·63 to 1·62]; estimated percentage difference 0% [-1% to 0·6%]), serious adverse events (three and five respectively), and the proportion feeling fully recovered were similar in both groups at 6 months (74·3% and 71·2% respectively (RR = 1·05, [1·02 to 1·08]) and also at 3 and 12 months. INTERPRETATION Ivermectin for COVID-19 is unlikely to provide clinically meaningful improvement in recovery, hospital admissions, or longer-term outcomes. Further trials of ivermectin for SARS-Cov-2 infection in vaccinated community populations appear unwarranted. FUNDING UKRI/National Institute of Health Research (MC_PC_19079).
Collapse
Affiliation(s)
- Gail Hayward
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Ly-Mee Yu
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Paul Little
- Primary Care Research Centre, University of Southampton, Southampton, UK
| | - Oghenekome Gbinigie
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Milensu Shanyinde
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Victoria Harris
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Jienchi Dorward
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Benjamin R Saville
- Berry Consultants, TX, USA; Department of Biostatistics, Vanderbilt University School of Medicine, TN, USA
| | | | - Philip H Evans
- College of Medicine and Health, University of Exeter, Exeter, UK; National Institute for Health Research (NIHR) Clinical Research Network, National Institute for Health Research, London, UK
| | - Nicholas P B Thomas
- National Institute for Health Research (NIHR) Clinical Research Network, National Institute for Health Research, London, UK
| | - Mahendra G Patel
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Duncan Richards
- Royal College of General Practitioners, London, UK; Oxford Clinical Trials Research Unit, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Oliver V Hecke
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | | | | | - Jared Robinson
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | - Julie Allen
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Emma Ogburn
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Jenna Grabey
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Fd Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK.
| | - Christopher C Butler
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Papic I, Bistrovic P, Krecak I, Ortner Hadziabdic M, Lucijanic M. Specific adverse outcomes associated with selective serotonin reuptake inhibitors use in COVID-19 patients might be potentiated by remdesivir use. J Psychopharmacol 2024; 38:395-403. [PMID: 38481078 DOI: 10.1177/02698811241237868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
BACKGROUND Due to non-consistent reports in the literature, there are uncertainties about the potential benefits and harms of selective serotonin reuptake inhibitors (SSRIs) in patients with Coronavirus disease 2019 (COVID-19). AIM To investigate associations of SSRIs with clinical characteristics and unwanted outcomes among real-life severe and critical COVID-19 patients and their relationship with remdesivir (RDV) use. METHODS This retrospective cohort study evaluated a total of 1558 COVID-19 patients of the white race treated in a tertiary center institution, among them 779 patients treated with RDV and 779 1:1 case-matched patients. RESULTS A total of 78 (5%) patients were exposed to SSRIs during hospitalization, similarly distributed among patients treated with RDV and matched patients (5.1 and 4.9%). No significant associations of SSRI use with age, sex, comorbidity burden, and COVID-19 severity were present in either of the two cohorts (p > 0.05 for all analyses). In multivariate analyses adjusted for clinically meaningful variables, SSRI use was significantly associated with higher mortality among RDV (adjusted odds ratio (aOR) 2.0, p = 0.049) and matched patients (aOR 2.22, p = 0.044) and with higher risk for mechanical-ventilation (aOR 2.57, p = 0.006), venous-thromboembolism (aOR 3.69, p = 0.007), and bacteremia (aOR 2.22, p = 0.049) among RDV treated patients. CONCLUSIONS Adverse outcomes associated with SSRI use in COVID-19 patients might be potentiated by RDV use, and clinically significant interactions between these two drug classes might exist. Although our findings raise important considerations for clinical practice, they are limited by retrospective nature of the study, lack of ethnic diversity, and the potential for unmeasured confounding factors. Future studies exploring underlying biological mechanisms are needed.
Collapse
Affiliation(s)
- Ivan Papic
- Department of Pharmacy, University hospital Dubrava, Zagreb, Croatia
| | - Petra Bistrovic
- Department of Cardiology, University hospital Dubrava, Zagreb, Croatia
| | - Ivan Krecak
- Department of Internal Medicine, General hospital of Sibenik-Knin county, Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- University of Applied Sciences, Sibenik, Croatia
| | - Maja Ortner Hadziabdic
- Centre for Applied Pharmacy, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Marko Lucijanic
- Department of Hematology, University hospital Dubrava, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
43
|
Song Z, Shi S, Zhang Y. Ivermectin for treatment of COVID-19: A systematic review and meta-analysis. Heliyon 2024; 10:e27647. [PMID: 38510038 PMCID: PMC10950893 DOI: 10.1016/j.heliyon.2024.e27647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
The effect of ivermectin (IVM) in treating coronavirus disease 2019 (COVID-19) is still controversial, yet the drug has been widely used in the world. The aim of this review was to systematically evaluate the clinical outcomes of IVM in patients with COVID-19. From inception to June 22, 2023, the PubMed, EMBASE, Web of Science (WOS), and scopus databases were searched for relevant observational studies on the risk of RA in migraineurs. We searched PubMed/Medline, EMBASE, the Cochrane Library, Web of Science, medRxiv, and bioRxiv to collect all relevant publications from inception to June 22, 2023. Primary outcomes were all-cause mortality rate, mechanical ventilation (MV) requirement, PCR negative conversion, and adverse events (AEs). Revman 5.4 was used to assess the risk of bias (RoB) and quality of evidence. Thirty-three RCTs (n = 10,489) were included. No significant difference in all-cause mortality rates or PCR negative conversion between IVM and controls. There were significant differences in MV requirement (RR 0.67, 95% CI 0.47-0.96) and AEs (RR 0.87, 95% CI 0.80-0.95) between the two groups. Ivermectin could reduce the risk of MV requirement and AEs in patients with COVID-19, without increasing other risks. In the absence of a better alternative, clinicians could use it with caution.
Collapse
Affiliation(s)
- Zhilong Song
- School of Public Health, Xiamen University, Fujian, China
| | - Senyuan Shi
- School of Medicine, Southeast University, Jiangsu, China
| | - Yongli Zhang
- School of Medicine, Xiamen University, Fujian, China
| |
Collapse
|
44
|
Silverii GA, Fumagalli C, Rozzini R, Milani M, Mannucci E, Marchionni N. Is Metformin Use Associated with a More Favorable COVID-19 Course in People with Diabetes? J Clin Med 2024; 13:1874. [PMID: 38610639 PMCID: PMC11012895 DOI: 10.3390/jcm13071874] [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: 02/18/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Diabetes Mellitus (DM) has been associated with a higher Coronavirus disease-19 (COVID-19) mortality, both in hospitalized patients and in the general population. A possible beneficial effect of metformin on the prognosis of COVID-19 has been reported in some observational studies, whereas other studies disagree. Methods: To investigate the possible effect of metformin on COVID-19 in-hospital mortality, we performed a retrospective study that included all SARS-CoV-2-positive patients with DM who were admitted to two Italian hospitals. In order to adjust for possible confounders accounting for the observed reduction of mortality in metformin users, we adopted the COVID-19 Mortality Risk Score (COVID-19 MRS) as a covariate. Results: Out of the 524 included patients, 33.4% died. A binomial logistic regression showed that metformin use was associated with a significant reduction in case fatality (OR 0.67 [0.45-0.98], p = 0.039), with no significant effect on the need for ventilation (OR 0.75 [0.5-1.11], p = 0.146). After adjusting for COVID-19 MRS, metformin did not retain a significant association with in-hospital mortality [OR 0.795 (0.495-1.277), p = 0.342]. Conclusions: A beneficial effect of metformin on COVID-19 was not proven after adjusting for confounding factors. The use of validated tools to stratify the risk for COVID-19 severe disease and death, such as COVID-19 MRS, may be useful to better explore the potential association of medications and comorbidities with COVID-19 prognosis.
Collapse
Affiliation(s)
- Giovanni Antonio Silverii
- Experimental and Clinical Biomedical Sciences “Mario Serio” Department, University of Florence, 50134 Florence, Italy (G.A.S.)
| | - Carlo Fumagalli
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Renzo Rozzini
- Department of Internal Medicine and Geriatrics, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Marta Milani
- Experimental and Clinical Biomedical Sciences “Mario Serio” Department, University of Florence, 50134 Florence, Italy (G.A.S.)
| | - Edoardo Mannucci
- Experimental and Clinical Biomedical Sciences “Mario Serio” Department, University of Florence, 50134 Florence, Italy (G.A.S.)
| | - Niccolò Marchionni
- Experimental and Clinical Medicine Department, University of Florence, 50134 Florence, Italy;
| |
Collapse
|
45
|
Akyol BA, Gokbulut C. The effect of intravenous lipid emulsion (ILE) on the pharmacokinetic/toxicokinetic dispositions of ivermectin and carprofen in rabbits. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1841-1852. [PMID: 37768375 DOI: 10.1007/s00210-023-02738-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Intravenous lipid emulsion (ILE) has been widely used as an effective antidote in both veterinary and human medicine for the treatment of acute intoxications caused by drugs and pesticides with high lipid solubility. This study was conducted to investigate the effect of ILE co-administration on the kinetic dispositions of ivermectin (IVM) and carprofen (CRP) following intravenous bolus administration at subtoxic doses in rabbits.Twenty-four male New Zealand rabbits weighing 2.78 ± 0.2 kg were used in this study. Rabbits were divided into four groups (Group 1: IVM and Group 2: IVM + ILE or Group 3: CRP and Group 4: CRP + ILE), each group consisting of 6 animals. In the IVM study, Group 1 received IVM (0.6 mg/kg) alone while Group 2 received IVM (0.6 mg/kg) and ILE (2.5 ml/kg). In the CRP study, Group 3 received CRP (12 mg/kg) alone while Group 4 received CRP (12 mg/kg) and ILE (2.5 ml/kg). In both drug groups, ILE was administered 3 times as an i.v. bolus at the 10th min and repeated 4th and 8th h after the drug administration. Blood samples were collected from the auricular vein at various times after drug administration. The drug concentrations in plasma samples were determined by high-pressure liquid chromatography. Kinetic parameters were calculated using a non-compartmental model for both CRP and IVM.The C0 and area under the concentration-time curve from zero up to ∞ (AUC0-∞) values were significantly greater with ILE co-administration (2136 ng/ml and 360.84 ng.d/ml) compared to the IVM alone (1340.63 ng/ml and 206 ng.d/ml), respectively. Moreover, the volume of distribution (Vdss) and clearance (Cl) of IVM were reduced by approximately 42% and 46% with ILE co-administration compared to IVM alone resulting in a reduction of the distribution and slower elimination, respectively. Similar differences in C0, and Vdss values were also observed in CRP with ILE co-administration compared to CRP alone. ILE co-administration changed significantly the kinetic profile of both IVM and CRP in rabbits, supporting the lipid sink theory in which highly lipid-soluble compounds are absorbed into the lipid phase of plasma from peripheral organs such as the heart and brain affected by the acute toxicity of the compounds.
Collapse
Affiliation(s)
- Busra Aslan Akyol
- Department of Veterinary Pharmacology and Toxicology, Institute of Health Sciences, Balikesir University, CoHE 100/2000 Scholarship Holder, University Rectorate Çağış Campus 17. Km, Bigadiç Caddesi, 10145, Balikesir, Turkey
| | - Cengiz Gokbulut
- Department of Medical Pharmacology, Faculty of Medicine, Balikesir University, University Rectorate Çağış Campus 17. Km, Bigadiç Caddesi, 10145, Balikesir, Turkey.
- Department of Veterinary Pharmacology and Toxicology, Institute of Health Sciences, Balikesir University, University Rectorate Çağış Campus 17. Km, Bigadiç Caddesi, 10145, Balikesir, Turkey.
| |
Collapse
|
46
|
Malar DS, Verma K, Prasanth MI, Tencomnao T, Brimson JM. Network analysis-guided drug repurposing strategies targeting LPAR receptor in the interplay of COVID, Alzheimer's, and diabetes. Sci Rep 2024; 14:4328. [PMID: 38383841 PMCID: PMC10882047 DOI: 10.1038/s41598-024-55013-9] [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: 09/15/2023] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has greatly affected global health. Emerging evidence suggests a complex interplay between Alzheimer's disease (AD), diabetes (DM), and COVID-19. Given COVID-19's involvement in the increased risk of other diseases, there is an urgent need to identify novel targets and drugs to combat these interconnected health challenges. Lysophosphatidic acid receptors (LPARs), belonging to the G protein-coupled receptor family, have been implicated in various pathological conditions, including inflammation. In this regard, the study aimed to investigate the involvement of LPARs (specifically LPAR1, 3, 6) in the tri-directional relationship between AD, DM, and COVID-19 through network analysis, as well as explore the therapeutic potential of selected anti-AD, anti-DM drugs as LPAR, SPIKE antagonists. We used the Coremine Medical database to identify genes related to DM, AD, and COVID-19. Furthermore, STRING analysis was used to identify the interacting partners of LPAR1, LPAR3, and LPAR6. Additionally, a literature search revealed 78 drugs on the market or in clinical studies that were used for treating either AD or DM. We carried out docking analysis of these drugs against the LPAR1, LPAR3, and LPAR6. Furthermore, we modeled the LPAR1, LPAR3, and LPAR6 in a complex with the COVID-19 spike protein and performed a docking study of selected drugs with the LPAR-Spike complex. The analysis revealed 177 common genes implicated in AD, DM, and COVID-19. Protein-protein docking analysis demonstrated that LPAR (1,3 & 6) efficiently binds with the viral SPIKE protein, suggesting them as targets for viral infection. Furthermore, docking analysis of the anti-AD and anti-DM drugs against LPARs, SPIKE protein, and the LPARs-SPIKE complex revealed promising candidates, including lupron, neflamapimod, and nilotinib, stating the importance of drug repurposing in the drug discovery process. These drugs exhibited the ability to bind and inhibit the LPAR receptor activity and the SPIKE protein and interfere with LPAR-SPIKE protein interaction. Through a combined network and targeted-based therapeutic intervention approach, this study has identified several drugs that could be repurposed for treating COVID-19 due to their expected interference with LPAR(1, 3, and 6) and spike protein complexes. In addition, it can also be hypothesized that the co-administration of these identified drugs during COVID-19 infection may not only help mitigate the impact of the virus but also potentially contribute to the prevention or management of post-COVID complications related to AD and DM.
Collapse
Affiliation(s)
- Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kanika Verma
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
- Department of Molecular Epidemiology, ICMR- National Institute of Malaria Research (NIMR), New Delhi, India.
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research Unit for Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
47
|
Bonnet U, Juckel G, Kuhn J. Antidepressants for prevention of severe COVID-19, Long COVID and outlook for other viral diseases. Front Med (Lausanne) 2024; 11:1305184. [PMID: 38444417 PMCID: PMC10912328 DOI: 10.3389/fmed.2024.1305184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Affiliation(s)
- Udo Bonnet
- Department of Mental Health, Evangelisches Krankenhaus Castrop-Rauxel, Academic Teaching Hospital of the University of Duisburg/Essen, Castrop-Rauxel, Germany
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, Landschaftsverband Rheinland-Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Georg Juckel
- Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr University Bochum, Bochum, Germany
| | - Jens Kuhn
- Department of Psychiatry and Psychotherapy, University Hospital Cologne, Cologne, Germany
- Alexianer Hospital Cologne, Cologne, Germany
| |
Collapse
|
48
|
Liu TH, Wu JY, Huang PY, Tsai YW, Hsu WH, Chuang MH, Tang HJ, Lai CC. Clinical efficacy of N-acetylcysteine for COVID-19: A systematic review and meta-analysis of randomized controlled trials. Heliyon 2024; 10:e25179. [PMID: 38318025 PMCID: PMC10839595 DOI: 10.1016/j.heliyon.2024.e25179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Background The association between N-acetylcysteine (NAC) and COVID-19 remains undetermined; therefore, this meta-analysis assessed the clinical efficacy of NAC in the treatment of patients with COVID-19. Methods This study searched PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov for studies published from their inception to December 17, 2022. Only randomized controlled trials (RCTs) that assessed the clinical efficacy of NAC for patients with COVID-19 were included. Results Five RCTs involving 651 patients were included. There was no significant difference in mortality between the study group receiving NAC and the control group (15.6 % [50/320] vs. 32.3 %, [107/331]; risk ratio [RR]: 0.58; 95 % confidence interval [CI]: 0.24-1.40). In addition, the two groups did not differ with respect to the incidence of invasive mechanical ventilation (RR: 0.93; 95 % CI: 0.65-1.33), the risk of intensive care unit (ICU) admission (RR: 0.86; 95 % CI: 0.62-1.21), the length of hospital stay (mean difference [MD]: 0.17 days; 95 % CI: -0.67-1.01), and the length of ICU stay (MD: -0.77 days; 95 % CI: -2.97-1.42). Conclusions The administration of NAC did not improve the clinical outcomes of patients with COVID-19; its routine use is not recommended for patients with SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Ting-Hui Liu
- Department of Psychiatry, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan City, Taiwan
| | - Po-Yu Huang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ya-Wen Tsai
- Center for Integrative Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Wan-Hsuan Hsu
- Department of Internal Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| | - Min-Hsiang Chuang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| | - Hung-Jen Tang
- Department of Nutrition, Chi Mei Medical Center, Tainan City, Taiwan
| | - Chih-Cheng Lai
- Division of Hospital Medicine, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
49
|
De Jesús-González LA, del Ángel RM, Palacios-Rápalo SN, Cordero-Rivera CD, Rodríguez-Carlos A, Trujillo-Paez JV, Farfan-Morales CN, Osuna-Ramos JF, Reyes-Ruiz JM, Rivas-Santiago B, León-Juárez M, García-Herrera AC, Ramos-Cortes AC, López-Gándara EA, Martínez-Rodríguez E. A Dual Pharmacological Strategy against COVID-19: The Therapeutic Potential of Metformin and Atorvastatin. Microorganisms 2024; 12:383. [PMID: 38399787 PMCID: PMC10893401 DOI: 10.3390/microorganisms12020383] [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: 12/29/2023] [Revised: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Metformin (MET) and atorvastatin (ATO) are promising treatments for COVID-19. This review explores the potential of MET and ATO, commonly prescribed for diabetes and dyslipidemia, respectively, as versatile medicines against SARS-CoV-2. Due to their immunomodulatory and antiviral capabilities, as well as their cost-effectiveness and ubiquitous availability, they are highly suitable options for treating the virus. MET's effect extends beyond managing blood sugar, impacting pathways that can potentially decrease the severity and fatality rates linked with COVID-19. It can partially block mitochondrial complex I and stimulate AMPK, which indicates that it can be used more widely in managing viral infections. ATO, however, impacts cholesterol metabolism, a crucial element of the viral replicative cycle, and demonstrates anti-inflammatory characteristics that could modulate intense immune reactions in individuals with COVID-19. Retrospective investigations and clinical trials show decreased hospitalizations, severity, and mortality rates in patients receiving these medications. Nevertheless, the journey from observing something to applying it in a therapeutic setting is intricate, and the inherent diversity of the data necessitates carefully executed, forward-looking clinical trials. This review highlights the requirement for efficacious, easily obtainable, and secure COVID-19 therapeutics and identifies MET and ATO as promising treatments in this worldwide health emergency.
Collapse
Affiliation(s)
- Luis Adrián De Jesús-González
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico; (S.N.P.-R.); (C.D.C.-R.)
| | - Rosa María del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico; (S.N.P.-R.); (C.D.C.-R.)
| | - Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico; (S.N.P.-R.); (C.D.C.-R.)
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico; (S.N.P.-R.); (C.D.C.-R.)
| | - Adrián Rodríguez-Carlos
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Juan Valentin Trujillo-Paez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Carlos Noe Farfan-Morales
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana (UAM), Unidad Cuajimalpa, Ciudad de México 05348, Mexico;
| | | | - José Manuel Reyes-Ruiz
- División de Investigación en Salud, Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS), Veracruz 91897, Mexico;
- Facultad de Medicina, Región Veracruz, Universidad Veracruzana (UV), Veracruz 91700, Mexico
| | - Bruno Rivas-Santiago
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Moisés León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México 11000, Mexico;
| | - Ana Cristina García-Herrera
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Adriana Clara Ramos-Cortes
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Erika Alejandra López-Gándara
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| | - Estefanía Martínez-Rodríguez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (A.R.-C.); (J.V.T.-P.); (B.R.-S.); (A.C.G.-H.); (A.C.R.-C.); (E.A.L.-G.); (E.M.-R.)
| |
Collapse
|
50
|
Servais T, Laurent F, Roland T, Rossi C, De Groote E, Godart V, Repetto E, Ponchon M, Chasseur P, Crenier L, Van Eeckhoudt S, Yango J, Oriot P, Morisca Gavriliu M, Rouhard S, Deketelaere B, Maiter D, Hermans MP, Yombi JC, Orioli L. Mortality-related risk factors of inpatients with diabetes and COVID-19: A multicenter retrospective study in Belgium. ANNALES D'ENDOCRINOLOGIE 2024; 85:36-43. [PMID: 37574109 DOI: 10.1016/j.ando.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/01/2023] [Accepted: 08/03/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND AND AIMS We describe mortality-related risk factors of inpatients with diabetes and coronavirus disease 2019 (COVID-19) in Belgium. METHODS We conducted a multicenter retrospective study from March to May, 2020, in 8 Belgian centers. Data on admission of patients with diabetes and COVID-19 were collected. Survivors were compared to non-survivors to identify prognostic risk factors for in-hospital death using multivariate analysis in both the total population and in the subgroup of patients admitted in the intensive care unit (ICU). RESULTS The study included 375 patients. The mortality rate was 26.4% (99/375) in the total population and 40% (27/67) in the ICU. Multivariate analysis identified older age (HR 1.05 [CI 1.03-1.07], P<0.0001) and male gender (HR 2.01 [1.31-3.07], P=0.0013) as main independent risk factors for in-hospital death in the total population. Metformin (HR 0.51 [0.34-0.78], P=0.0018) and renin-angiotensin-aldosterone system blockers (HR 0.56 [0.36-0.86], P=0.0088) use before admission were independent protective factors. In the ICU, chronic kidney disease (CKD) was identified as an independent risk factor for death (HR 4.96 [2.14-11.5], P<0.001). CONCLUSION In-hospital mortality due to the first wave of COVID-19 pandemic in Belgium was high in patients with diabetes. We found that advanced age and male gender were independent risk factors for in-hospital death. We also showed that metformin use before admission was associated with a significant reduction of COVID-19-related in-hospital mortality. Finally, we showed that CKD is a COVID-19-related mortality risk factor in patients with diabetes admitted in the ICU.
Collapse
Affiliation(s)
- Thomas Servais
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - France Laurent
- Department of Infectiology, Centre Hospitalier Universitaire Ambroise Paré, Boulevard John Fitzgerald Kennedy 2, 7000 Mons, Belgium
| | - Thomas Roland
- Department of Infectiology, Centre Hospitalier Universitaire Ambroise Paré, Boulevard John Fitzgerald Kennedy 2, 7000 Mons, Belgium
| | - Camelia Rossi
- Department of Infectiology, Centre Hospitalier Universitaire Ambroise Paré, Boulevard John Fitzgerald Kennedy 2, 7000 Mons, Belgium
| | - Elodie De Groote
- Department of Infectiology, Hôpital de Jolimont, Rue Ferrer 159, 7100 Haine-Saint-Paul, Belgium
| | - Valérie Godart
- Department of Endocrinology-Diabetology, Hôpital de Jolimont, Rue Ferrer 159, 7100 Haine-Saint-Paul, Belgium
| | - Ernestina Repetto
- Department of Infectiology, Clinique Saint-Jean, Boulevard du Jardin Botanique 32, 1000 Brussels, Belgium
| | - Michel Ponchon
- Department of Endocrinology-Diabetology, Clinique Saint-Jean, Boulevard du Jardin Botanique 32, 1000 Brussels, Belgium
| | - Pascale Chasseur
- Department of Endocrinology, Hôpital Erasme, Cliniques Universiraires de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Laurent Crenier
- Department of Endocrinology, Hôpital Erasme, Cliniques Universiraires de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Sandrine Van Eeckhoudt
- Department of Internal Medicine and Infectious Diseases, Clinique Saint-Luc Bouge, Rue Saint-Luc 8, 5004 Namur, Belgium
| | - John Yango
- Department of Endocrinology-Diabetology, Clinique Saint-Luc Bouge, Rue Saint-Luc 8, 5004 Namur, Belgium
| | - Philippe Oriot
- Department of Diabetology, Centre Hospitalier de Mouscron, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Mirela Morisca Gavriliu
- Department of Diabetology, Centre Hospitalier de Mouscron, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Stéphanie Rouhard
- Department of Endocrinology-Diabetology, Centre Hospitalier Régional de Huy, Rue Delloye Matthieu 2, 4500 Huy, Belgium
| | - Benjamin Deketelaere
- Institute of Statistics, Biostatistics and Actuarial Sciences, Université Catholique de Louvain, Rue des Wallons 6, 1348 Ottignies-Louvain-La-Neuve, Belgium
| | - Dominique Maiter
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Michel Paul Hermans
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Jean Cyr Yombi
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Laura Orioli
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Endocrinology, Diabetology and Nutrition, Institute of Clinical and Experimental Research, Université Catholique de Louvain, Avenue Hippocrate 55, 1200 Brussels, Belgium.
| |
Collapse
|