101
|
Hora S, Pahwa P, Siddiqui H, Saxena A, Kashyap M, Sevak JK, Singh R, Javed M, Yadav P, Kale P, Ramakrishna G, Bajpai M, Rathore A, Maras JS, Tyagi S, Sarin SK, Trehanpati N. Metabolic alterations unravel the maternofetal immune responses with disease severity in pregnant women infected with SARS-CoV-2. J Med Virol 2023; 95:e29257. [PMID: 38054548 DOI: 10.1002/jmv.29257] [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: 05/10/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
Pregnancy being an immune compromised state, coronavirus disease of 2019 (COVID-19) disease poses high risk of premature delivery and threat to fetus. Plasma metabolome regulates immune cellular responses, therefore we aimed to analyze the change in plasma secretome, metabolome, and immune cells with disease severity in COVID-19 positive pregnant females and their cord blood. COVID-19 reverse transcriptase-polymerase chain reaction positive pregnant females (n = 112) with asymptomatic (Asy) (n = 82), mild (n = 21), or moderate (n = 9) disease, healthy pregnant (n = 18), COVID-19 positive nonpregnant females (n = 7) were included. Eighty-two cord blood from COVID-19 positive and seven healthy cord blood were also analyzed. Mother's peripheral blood and cord blood were analyzed for untargeted metabolome profiling and cytokines by using high-resolution mass spectrometry and cytokine bead array. Immune scan was performed only in mothers' blood by flow cytometry. In Asy severe acute respiratory syndrome coronavirus 2 infection, the amino acid metabolic pathways such as glycine, serine, l-lactate, and threonine metabolism were upregulated with downregulation of riboflavin and tyrosine metabolism. However, with mild-to-moderate disease, the pyruvate and nicotinamide adenine dinucleotide (NAD+ ) metabolism were mostly altered. Cord blood mimicked the mother's metabolomic profiles by showing altered valine, leucine, isoleucine, glycine, serine, threonine in Asy and NAD+ , riboflavin metabolism in mild and moderate. Additionally, with disease severity tumor necrosis factor-α, interferon (IFN)-α, IFN-γ, interleukin (IL)-6 cytokine storm, IL-9 was raised in both mothers and neonates. Pyruvate, NAD metabolism and increase in IL-9 and IFN-γ had an impact on nonclassical monocytes, exhausted T and B cells. Our results demonstrated that immune-metabolic interplay in mother and fetus is influenced with increase in IL-9 and IFN-γ regulated pyruvate, lactate tricarboxylic acid, and riboflavin metabolism with context to disease severity.
Collapse
Affiliation(s)
- Sandhya Hora
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Prabhjyoti Pahwa
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Hamda Siddiqui
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anoushka Saxena
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Minal Kashyap
- Department of Gynecology and Obstetrics, Lok Nayak Jai Prakash Hospital, New Delhi, India
| | - Jayesh K Sevak
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Ravinder Singh
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Maryam Javed
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Pushpa Yadav
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Pratibha Kale
- Department of Microbiology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Gayatri Ramakrishna
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Meenu Bajpai
- Department of Transfusion Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Asmita Rathore
- Department of Gynecology and Obstetrics, Lok Nayak Jai Prakash Hospital, New Delhi, India
| | - Jaswinder S Maras
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shakun Tyagi
- Department of Gynecology and Obstetrics, Lok Nayak Jai Prakash Hospital, New Delhi, India
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nirupama Trehanpati
- Department of Molecular and Cellular Medicine, Laboratory of Molecular Immunology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
102
|
Chen ZJ, Xiao J, Chen HH. Identification of Key Genes Related to Immune Cells in Patients with COVID-19 Via Integrated Bioinformatics-Based Analysis. Biochem Genet 2023; 61:2650-2671. [PMID: 37222960 PMCID: PMC10206360 DOI: 10.1007/s10528-023-10400-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/08/2023] [Indexed: 05/25/2023]
Abstract
COVID-19 has spread all over the world which poses a serious threat to social economic development and public health. Despite enormous progress has been made in the prevention and treatment of COVID-19, the specific mechanism and biomarker related to disease severity or prognosis have not been clarified yet. Our study intended to further explore the diagnostic markers of COVID-19 and their relationship with serum immunology by bioinformatics analysis. The datasets about COVID-19 were downloaded from the Gene Expression Omnibus (GEO) dataset. The differentially expressed genes (DEGs) were selected via the limma package. Then, weighted gene co-expression network analysis (WGCNA) was conducted to identify the critical module associated with the clinic status. The intersection DEGs were processed for further enrichment analysis. The final diagnostic genes for COVID-19 were selected and verified through special bioinformatics algorithms. There were significant DEGs between the normal and COVID-19 patients. These genes were mainly enriched in cell cycle, complement and coagulation cascade, extracellular matrix (ECM) receptor interaction, and the P53 signaling pathway. As much as 357 common intersected DEGs were selected in the end. These DEGs were enriched in organelle fission, mitotic cell cycle phase transition, DNA helicase activity, cell cycle, cellular senescence, and P53 signaling pathway. Our study also identified CDC25A, PDCD6, and YWAHE were potential diagnostic markers of COVID-19 with the AUC (area under curve), 0.958 (95% CI 0.920-0.988), 0.941(95% CI 0.892-0.980), and 0.929 (95% CI 0.880-0.971). Moreover, CDC25A, PDCD6, and YWAHE were correlated with plasma cells, macrophages M0, T cells CD4 memory resting, T cells CD8, dendritic cells, and NK cells. Our study discovered that CDC25A, PDCD6, and YWAHE can be used as diagnostic markers for COVID-19. Moreover, these biomarkers were also closely associated with immune cell infiltration, which plays a pivotal role in the diagnosis and progression of COVID-19.
Collapse
Affiliation(s)
- Zhao-Jun Chen
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Xiao
- Department of Cardiology, Wuhan Asia Heart General Hospital, Wuhan, China
| | - Hai-Hua Chen
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
103
|
Park HR, Yoo MG, Kim JM, Bae SJ, Lee H, Kim J. Effectiveness of Molnupiravir Treatment in Patients with COVID-19 in Korea: A Propensity Score Matched Study. Infect Chemother 2023; 55:490-499. [PMID: 38014730 PMCID: PMC10771951 DOI: 10.3947/ic.2023.0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The MOVe-OUT (efficacy and safety of molnupiravir [MK-4482] in non-hospitalized adult participants with COVID-19 [MK-4482-002]) trial reported that the administration of molnupiravir in unvaccinated patients with coronavirus disease 2019 (COVID-19) before the Omicron epidemic showed a preventive effect of 31% against hospitalization and death. However, studies on the preventive effect of molnupiravir against progression to severe disease and death in patients with COVID-19 during the Omicron epidemic are limited. This study aimed to evaluate the preventive effect of molnupiravir against severe/critical illness or death and death in Korean patients with COVID-19 who were vaccinated mostly during the Omicron epidemic. MATERIALS AND METHODS This study used large-scale retrospective cohort data to select patients with COVID-19 who were either treated or not treated with molnupiravir, between August 2022 and March 2023, at a ratio of 1 : 4 using the propensity score matching method. In total, 762,768 patients comprised the non- administered group, and 190,692 patients comprised the molnupiravir-administered group. The preventive effect of molnupiravir against severe/critical illness or death and death was analyzed using logistic regression analysis. RESULTS The preventive effect of molnupiravir against severe/critical illness or death and death, represented by the odds ratio (OR) and 95% confidence interval (CI), in the molnupiravir-administered and non-administered group was (OR: 0.714; CI: 0.667 - 0.764) and (OR: 0.749; CI: 0.682 - 0.823), respectively. As age increased, the preventive effect against severe/critical illness or death and death increased. The preventive effect against severe/critical illness or death at ≥60 years was (OR: 0.669; CI: 0.624 - 0.717), at ≥70 years was (OR: 0.614; CI: 0.570 - 0.661), and at ≥80 years was (OR: 0.563; CI: 0.515 - 0.615). The preventive effect against death at ≥60 years was (OR: 0.729; CI: 0.663 - 0.802), at ≥70 years was (OR: 0.676; CI: 0.612 - 0.747), and at ≥80 years was (OR: 0.622; CI: 0.554 - 0.698). CONCLUSION Although molnupiravir showed a relatively weak preventive effect against severe/critical illness or death (29%) and death (25%) among patients with COVID-19, it exhibited a stronger protective effect in older patients than in younger patients. In particular, the preventive effect against severe/critical illness or death (44%) and death (38%) in those aged ≥80 years was pronounced. This study strongly suggests that molnupiravir administration can alleviate the burden on the medical system, and treat patients with COVID-19 effectively by reducing its progression to severe disease and death.
Collapse
Affiliation(s)
- Hye Rim Park
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Min-Gyu Yoo
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Public Health Emergency Response Research, Bureau of Public Health Emergency Preparedness, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Jong Mu Kim
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Soon Jong Bae
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Hyungmin Lee
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Emerging Infectious Disease, Bureau of Infectious Disease Risk Response, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Vaccine-Prevetable Diseases Control and National Immunization Program, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Jungyeon Kim
- Patient Management Team, Central Disease Control Headquarters for COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Emerging Infectious Disease, Bureau of Infectious Disease Risk Response, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Clinical Research, Center for Emerging Virus Research, National Institute of Infectious Disease, Korea Disease Control and Prevention Agency, Cheongju, Korea.
| |
Collapse
|
104
|
Hwang JH, You YS, Yeom SW, Lee MG, Lee JH, Kim MG, Kim JS. Influenza viral infection is a risk factor for severe illness in COVID-19 patients: a nationwide population-based cohort study. Emerg Microbes Infect 2023; 12:2164215. [PMID: 36580041 PMCID: PMC9858545 DOI: 10.1080/22221751.2022.2164215] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In order to prepare for the twindemic of influenza and SARS-CoV-2 infection, we investigated the association between influenza infection and subsequent severity of SARS-CoV-2 infection. A population-based nationwide cohort study was performed using data from the National Health Insurance Service (NHIS) in the Republic of Korea. This study included 274,126 individuals who underwent SARS-CoV-2 PCR testing between 20 January 2020 and 1 October 2020. Among these patients, 28,338 tested positive for SARS-CoV-2, and 4,003 of these individuals had a history of influenza. The control group was selected through 1:1 propensity score matching. In the group of 4,003 COVID-19-positive individuals with no history of influenza, 192 (4.8%) experienced severe illness from COVID-19 infection. In the group of 4,003 COVID-19-positive individuals with a history of influenza, 260 (6.5%) had severe illness from COVID-19, and the overall adjusted odds ratio (aOR) was 1.29 (95% confidence interval 1.04-1.59). Among the 4,003 COVID-19-positive individuals with a history of influenza, severe COVID-19 infection was experienced by 143 of 1,760 (8.1%) with an influenza history within 1 year before the onset of COVID-19, 48 of 1,129 (4.3%) between 1 and 2 years, and 69 of 1,114 (6.2%) between 2 and 3 years before COVID-19 onset, and the aORs were 1.54 (1.20-1.98), 1.19 (0.84-1.70), and 1.00 (0.73-1.37), respectively. In conclusion, individuals who had an influenza infection less than 1 year before COVID-19 infection were at an increased risk of experiencing severe illness from the SARS-CoV-2 infection. To control the public health burden, it is essential that effective public health control measures, which include influenza vaccination, hand washing, cough etiquette, and mask use are in place.
Collapse
Affiliation(s)
- Jeong-Hwan Hwang
- Department of Internal Medicine, Division of Infectious Diseases, Jeonbuk National University Medical School, Jeonju, South Korea,Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Yeon Seok You
- Department of Medical Informatics, Jeonbuk National University, Jeonju, South Korea,Department of Otorhinolaryngology, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Sang Woo Yeom
- Department of Medical Informatics, Jeonbuk National University, Jeonju, South Korea
| | - Min Gyu Lee
- Department of Medical Informatics, Jeonbuk National University, Jeonju, South Korea
| | - Jong-hwan Lee
- Department of Otorhinolaryngology, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Min Gul Kim
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea,Department of Pharmacology, Jeonbuk National University Medical School, Jeonju, South Korea, Min Gul Kim Department of Pharmacology, Jeonbuk National University Medical School, Jeonju54907, South Korea
| | - Jong Seung Kim
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea,Department of Medical Informatics, Jeonbuk National University, Jeonju, South Korea,Department of Otorhinolaryngology, Jeonbuk National University Medical School, Jeonju, South Korea,Jong Seung Kim Department of Medical Informatics, Jeonbuk National University, Jeonju54907, South Korea; Department of Otorhinolaryngology, Jeonbuk National University Medical School, Jeonju54907, South Korea
| |
Collapse
|
105
|
Bertuccio P, Degli Antoni M, Minisci D, Amadasi S, Castelli F, Odone A, Quiros-Roldan E. The impact of early therapies for COVID-19 on death, hospitalization and persisting symptoms: a retrospective study. Infection 2023; 51:1633-1644. [PMID: 37024626 PMCID: PMC10079146 DOI: 10.1007/s15010-023-02028-5] [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: 01/12/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023]
Abstract
PURPOSE Oral antivirals (nirmatrelvir/ritonavir and molnupiravir), intravenous short treatment of remdesivir and anti-SARS-CoV-2 monoclonal antibodies (mAbs) have been used for early COVID-19 treatments in high risk of disease progression patients. The term long COVID has been used to refer to a range of new, returning, or ongoing symptoms after SARS-CoV-2 infection. Little is known about the impact of such therapies on long COVID. METHODS This is a retrospective observational study, including all outpatients evaluated from April 2021 to March 2022 in Brescia, Lombardy, northern Italy. Patients were stratified in three groups: (a) treated with mAbs, (b) treated with antivirals drugs and (c) controls (patients eligible for a or b who refused treatment). Data were collected at baseline and at month 1 and 3 (data on self-reported symptoms were collected using a telephone-administered questionnaire). We assessed early COVID-19 therapies effectiveness in preventing hospitalization, death at 1 or 3 months and persisting symptoms at 3 months after the onset of SARS-CoV-2 infection. RESULTS A total of 649 patients were included in the study, of which 242 (37.3%) were treated with mAbs, 197 (30.3%) with antiviral drugs and 210 (32.4%) were not treated. Patients most frequently reported cerebro-cardiovascular diseases (36.7%) followed by obesity (22%). Overall, 29 patients (4.5%) died or were hospitalized at 1 or 3-month follow-up. Death or hospitalization was positively associated with older ages, with a significant linear trend (OR 3.05; 95% CI 1.16-8.06, for patients aged 80 or more years compared to those aged less than 65). Data on long COVID at 3 months were available for 323 (49.8%) patients. A positive association emerged for females compared to men, with an OR of 2.14 (95% CI 1.30-3.53) for any symptoms. Conversely, inverse associations were found for treatment groups as compared to the control one, with significant estimates among patients treated with antiviral drugs for any symptoms (OR 0.43, 95% CI 0.21-0.87) and patients treated with mAbs for any neuro-behavioral symptoms (OR 0.48, 95% CI 0.25-0.92). CONCLUSIONS We report beneficial effect of early use of anti-SARS-CoV-2 antivirals and mAbs on long COVID.
Collapse
Affiliation(s)
- Paola Bertuccio
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Melania Degli Antoni
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali Civili Di Brescia and University of Brescia, Brescia, Italy
| | - Davide Minisci
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali Civili Di Brescia and University of Brescia, Brescia, Italy
| | - Silvia Amadasi
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali Civili Di Brescia and University of Brescia, Brescia, Italy
| | - Francesco Castelli
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali Civili Di Brescia and University of Brescia, Brescia, Italy
| | - Anna Odone
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Eugenia Quiros-Roldan
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali Civili Di Brescia and University of Brescia, Brescia, Italy.
| |
Collapse
|
106
|
Jeong K, Kim Y, Jeon J, Kim K. Subtyping of COVID-19 samples based on cell-cell interaction in single cell transcriptomes. Sci Rep 2023; 13:19629. [PMID: 37949890 PMCID: PMC10638268 DOI: 10.1038/s41598-023-46350-2] [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: 08/11/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
In single-cell transcriptome analysis, numerous biomarkers related to COVID-19 severity, including cell subtypes, genes, and pathways, have been identified. Nevertheless, most studies have focused on severity groups based on clinical features, neglecting immunological heterogeneity within the same severity level. In this study, we employed sample-level clustering using cell-cell interaction scores to investigate patient heterogeneity and uncover novel subtypes. The clustering results were validated using external datasets, demonstrating superior reproducibility and purity compared to gene expression- or gene set enrichment-based clustering. Furthermore, the cell-cell interaction score-based clusters exhibited a strong correlation with the WHO ordinal severity score based on clinical characteristics. By characterizing the identified subtypes through known COVID-19 severity-associated biomarkers, we discovered a "Severe-like moderate" subtype. This subtype displayed clinical features akin to moderate cases; however, molecular features, such as gene expression and cell-cell interactions, resembled those of severe cases. Notably, all patients who progressed from moderate to severe belonged to this subtype, underscoring the significance of cell-cell interactions in COVID-19 patient heterogeneity and severity.
Collapse
Affiliation(s)
- Kyeonghun Jeong
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yooeun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaemin Jeon
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
- Department of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
107
|
Noh HJ, Song JH, Ham SY, Park Y, Won HK, Kim SJ, Chung KB, Kim CK, Ahn YM, Lee BJ, Kang HR. Clinical outcomes of mild to moderate coronavirus disease 2019 patients treated with Regdanvimab in delta-variant outbreak: Retrospective cohort study. Medicine (Baltimore) 2023; 102:e35987. [PMID: 37960790 PMCID: PMC10637437 DOI: 10.1097/md.0000000000035987] [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: 08/02/2022] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
Regdanvimab is a novel neutralizing antibody agent used for the treatment of coronavirus disease 2019 (COVID-19). However, the effectiveness of regdanvimab in delta-variant patients has rarely been investigated. We examined the clinical outcomes and adverse events in COVID 19 patients treated with regdanvimab in the delta-variant era. Data were collected from laboratory-confirmed COVID-19 hospitalized patients who received regdanvimab in 2021 and categorized into pre-delta and delta variant groups. The primary outcome was the need for oxygen therapy. Rescue therapy, clinical improvement, and adverse events were analyzed. Among 101 patients treated with regdanvimab, 31 (30.7%) were delta patients and 49 (48.5) were pre-delta patients. 64.4% were male, the mean age was 60.3 years, and 70 patients (69%) had at least one underlying disease. The median interval from symptom onset to injection was 4 days. Twenty-three patients (23%) needed oxygen therapy, including 9 (29%) in the delta and 8 (16.3%) in the pre-delta group. (P = .176) The risk of early oxygen supplement was higher in the delta group (adjusted hazard ratio (aHR), 6.75; 95% confidence interval(CI), 1.53-29.8). The in-hospital survival rate was 100%, and no patients were admitted to the intensive care unit. Adverse events occurred in 43% of patients:13 (42%) delta patients and 23 (47%) pre-delta patients had any adverse events (P = .661). Patients treated with regdanvimab 4 days after symptom onset showed a favorable prognosis (aHR, 0.26; 95% CI, 0.26-0.91). We found that the high-risk mild to moderate COVID-19 patients treated with regdanvimab showed similar disease progression in delta-variant patients and pre-delta variants; however, we need to be more closely observed delta-variant patients than those in the pre-delta group despite regdanvimab treatment due to rapid disease aggravation.
Collapse
Affiliation(s)
- Hyeong-Jun Noh
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Jin Hwa Song
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Sin Young Ham
- Division of Infectious Disease, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Yeonkyung Park
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Ha-Kyeong Won
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Soo Jung Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Republic of Korea
| | - Keun Bum Chung
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Choon Kwan Kim
- Division of Infectious Disease, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Young Mee Ahn
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Byoung-Jun Lee
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Hye-Rin Kang
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
108
|
Kang SW, Kim JW, Kim JY, Lim SY, Jang CY, Chang E, Yang JS, Kim KC, Jang HC, Kim D, Shin Y, Lee JY, Kim SH. Virological characteristics and the rapid antigen test as deisolation criteria in immunocompromised patients with COVID-19: A prospective cohort study. J Med Virol 2023; 95:e29228. [PMID: 38009999 DOI: 10.1002/jmv.29228] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
There are limited data supporting current Centers for Disease Control and Prevention guidelines for the isolation period in moderate to severely immunocompromised patients with coronavirus disease 2019 (COVID-19). Adult COVID-19 patients who underwent solid organ transplantation (SOT) or received active chemotherapy against hematologic malignancy were enrolled and weekly respiratory samples were collected. Samples with positive genomic real-time polymerase chain reaction results underwent virus culture and rapid antigen testing (RAT). A total of 65 patients (40 with hematologic malignancy and 25 SOT) were enrolled. The median duration of viable virus shedding was 4 weeks (interquartile range: 3-7). Multivariable analysis revealed that B-cell depletion (hazard ratio [HR]: 4.76) was associated with prolonged viral shedding, and COVID-19 vaccination (≥3 doses) was negatively associated with prolonged viral shedding (HR: 0.22). The sensitivity, specificity, positive predictive value, and negative predictive value of RAT for viable virus shedding were 79%, 76%, 74%, and 81%, respectively. The negative predictive value of RAT was only 48% (95% confidence interval [CI]: 33-65) in the samples from those with symptom onset ≤20 days, but it was as high as 92% (95% CI: 85-96) in the samples from those with symptom onset >20 days. About half of immunocompromised COVID-19 patients shed viable virus for ≥4 weeks from the diagnosis, and virus shedding was prolonged especially in unvaccinated patients with B-cell-depleting therapy treatment. RAT beyond 20 days in immunocompromised patients had a relatively high negative predictive value for viable virus shedding.
Collapse
Affiliation(s)
- Sung-Woon Kang
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun-Won Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Ji Yeun Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - So Yun Lim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Choi-Young Jang
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Euijin Chang
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sun Yang
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Kyung-Chang Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hee-Chang Jang
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Dasol Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Younmin Shin
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
109
|
Mahato RK, Karna AK, Thakur N, Bajracharya B, Pyakurel UR, Hayman DTS, Talvani A, Alam J, Pandey KR, Pathak N, Singh UN, Upadhyaya MK, Shrestha MP, Paudel KP, Morita K, Pandey BD. An Early Epidemiological Investigation of COVID-19 in Parsa, Nepal. Asia Pac J Public Health 2023; 35:564-567. [PMID: 37737120 DOI: 10.1177/10105395231201937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Affiliation(s)
- Ram Kumar Mahato
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - Ajit Kumar Karna
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
- USAID's Strengthening Systems for Better Health Activity, Kathmandu, Nepal
| | - Nishant Thakur
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - Bijay Bajracharya
- Epidemiology and Disease Control Division/Malaria Program Management Unit, Kathmandu, Nepal
| | - Uttam Raj Pyakurel
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - David T S Hayman
- Molecular Epidemiology and Public Health Laboratory, Infectious Disease Research Centre, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Andre Talvani
- Laboratory of Immunobiology of Inflammation, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Jahir Alam
- Birgunj Metropolitan City Office, Birgunj, Nepal
| | - Krishna Raj Pandey
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | | | | | | | | | - Krishna Prasad Paudel
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
| | - Kouichi Morita
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki, Japan
| | - Basu Dev Pandey
- Epidemiology and Disease Control Division, Department of Health Services, Ministry of Health and Population, Kathmandu, Nepal
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
110
|
Gómez-López ME, Aldrete-Cortez V, González-Carpinteiro A, Mendizábal-Espinosa R, Bobadilla L. The Emotional Experience of Mexican Women with SARS-CoV-2 during Pregnancy-A Qualitative Study. Healthcare (Basel) 2023; 11:2785. [PMID: 37893859 PMCID: PMC10606424 DOI: 10.3390/healthcare11202785] [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: 09/01/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Pregnant women have been considered a high-risk group for SARS-CoV-2 infection; the impact of the disease on the health of a mother and her child is still being studied. The emotional impact of the pandemic on pregnant women has been extensively studied. Emotional distress is proposed as a perspective to explain the emotional manifestations in women during this stage as something common rather than pathological. The objective of this study was to explore the emotional experience of women who tested positive for SARS-CoV-2 towards the end of their pregnancy, during the first and second waves of COVID-19 in Mexico. A qualitative study was carried out: 18 pregnant women with COVID-19 were interviewed. A thematic analysis of the data was performed, resulting in 3 main themes and 14 subthemes. The COVID-19-infected mothers-to-be experienced mild to moderate emotional distress. It was more intense for those with comorbidities. This distress was aggravated during obstetrical complications and comorbidities, as well as during COVID-19 and postpartum. The emotional distress was appeased by both the perception of medical care and social support. The emotional distress of pregnant women with COVID-19 requires emotional support to reduce its impact.
Collapse
Affiliation(s)
| | - Vania Aldrete-Cortez
- Laboratory of Neuroscience and Cognitive Development, School of Psychology, Universidad Panamericana, Mexico City 03920, Mexico; (V.A.-C.); (A.G.-C.)
| | - Aline González-Carpinteiro
- Laboratory of Neuroscience and Cognitive Development, School of Psychology, Universidad Panamericana, Mexico City 03920, Mexico; (V.A.-C.); (A.G.-C.)
| | - Rosa Mendizábal-Espinosa
- Social Research Institute of London, University College London, London WC1E 6BT, UK;
- Nacer Temprano, Vivir en Grande, Civil Association, Tlalnepantla de Baz 54080, Mexico
| | - Liliana Bobadilla
- Hospital of Gynecology and Obstetrics “Dr. Luis Castelazo Ayala”, Instituto Mexicano del Seguro Social, Mexico City 01090, Mexico;
| |
Collapse
|
111
|
Pérez Catalán I, Roig Martí C, Fabra Juana S, Domínguez Bajo E, Herrero Rodríguez G, Segura Fábrega A, Varea Villanueva M, Folgado Escudero S, Esteve Gimeno MJ, Palomo de la Sota D, Cardenal Álvarez A, Mateu Campos ML, Usó Blasco J, Ramos Rincón JM. One-year quality of life among post-hospitalization COVID-19 patients. Front Public Health 2023; 11:1236527. [PMID: 37869178 PMCID: PMC10588695 DOI: 10.3389/fpubh.2023.1236527] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction The long-term effects of SARS-CoV-2 are unclear, as are the factors influencing the evolution. Objective: to assess health-related quality of life 1 year after a hospital admission due to COVID-19 and to identify factors that may influence it. Materials and methods Retrospective observational study in a tertiary hospital from March 2021 to February 2022. Inclusion criteria: ≥18 years old and admitted for SARS-CoV-2 infection. Exclusion criteria: death, not located, refusal to participate, cognitive impairment, and language barrier. Variables: demographic data, medical history, clinical and analytical outcomes during hospital admission, treatment received, and vaccination against SARS-CoV-2 following admission. Participants were interviewed by phone 1 year after admission, using the SF-36 quality of life questionnaire. Results There were 486 included patients. The domains yielding the lowest scores were general health (median 65%, interquartile range [IQR] 45-80), vitality (median 65%, IQR 45-80), and mental health (median 73.5%, IQR 60-100). Multivariable analysis showed that female sex and fibromyalgia/fatigue had a negative influence on all domains. Obesity was associated with worse outcomes in physical functioning, physical role, bodily pain, and vitality. Other factors associated with worse scores were an older age in physical functioning and high age-adjusted Charslon comorbidity in physical functioning and general health. Age was associated with better results in emotional role and High C-reactive protein at admission on vitality. Conclusion One year after admission for COVID-19, quality of life remains affected, especially the domains of general health, vitality, and mental health. Factors associated with worse outcomes are female sex, fibromyalgia/chronic fatigue, and obesity.
Collapse
Affiliation(s)
- Ignacio Pérez Catalán
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Celia Roig Martí
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Sergio Fabra Juana
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Elena Domínguez Bajo
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Germán Herrero Rodríguez
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Ana Segura Fábrega
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - María Varea Villanueva
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - Sofía Folgado Escudero
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - María José Esteve Gimeno
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | | | | | | | - Jorge Usó Blasco
- Internal Medicine Service, University General Hospital of Castellon, Castellón de la Plana, Spain
| | - José Manuel Ramos Rincón
- Internal Medicine Service, University General Hospital of Alicante, Alicante, Spain
- Department of Clinical Medicine, Miguel Hernández University of Elche, Elche, Spain
| |
Collapse
|
112
|
Sanhueza S, Vidal MA, Hernandez MA, Henriquez-Beltran ME, Cabrera C, Quiroga R, Antilef BE, Aguilar KP, Castillo DA, Llerena FJ, Fraga Figueroa M, Nazal M, Castro E, Lagos P, Moreno A, Lastra JJ, Gajardo J, Garcés P, Riffo B, Buchert J, Sanhueza R, Ormazába V, Saldivia P, Vargas C, Nourdin G, Koch E, Zuñiga FA, Lamperti L, Bustos P, Guzmán-Gutiérrez E, Tapia CA, Ferrada L, Cerda G, Woehlbier U, Riquelme E, Yuseff MI, Muñoz Ramirez BA, Lombardi G, De Gonzalo-Calvo D, Salomon C, Verdugo RA, Quiñones LA, Colombo A, Barría MI, Labarca G, Nova-Lamperti E. Clinical and pulmonary function analysis in long-COVID revealed that long-term pulmonary dysfunction is associated with vascular inflammation pathways and metabolic syndrome. Front Med (Lausanne) 2023; 10:1271863. [PMID: 37869162 PMCID: PMC10590130 DOI: 10.3389/fmed.2023.1271863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Long-term pulmonary dysfunction (L-TPD) is one of the most critical manifestations of long-COVID. This lung affection has been associated with disease severity during the acute phase and the presence of previous comorbidities, however, the clinical manifestations, the concomitant consequences and the molecular pathways supporting this clinical condition remain unknown. The aim of this study was to identify and characterize L-TPD in patients with long-COVID and elucidate the main pathways and long-term consequences attributed to this condition by analyzing clinical parameters and functional tests supported by machine learning and serum proteome profiling. Methods Patients with L-TPD were classified according to the results of their computer-tomography (CT) scan and diffusing capacity of the lungs for carbon monoxide adjusted for hemoglobin (DLCOc) tests at 4 and 12-months post-infection. Results Regarding the acute phase, our data showed that L-TPD was favored in elderly patients with hypertension or insulin resistance, supported by pathways associated with vascular inflammation and chemotaxis of phagocytes, according to computer proteomics. Then, at 4-months post-infection, clinical and functional tests revealed that L-TPD patients exhibited a restrictive lung condition, impaired aerobic capacity and reduced muscular strength. At this time point, high circulating levels of platelets and CXCL9, and an inhibited FCgamma-receptor-mediated-phagocytosis due to reduced FcγRIII (CD16) expression in CD14+ monocytes was observed in patients with L-TPD. Finally, 1-year post infection, patients with L-TPD worsened metabolic syndrome and augmented body mass index in comparison with other patient groups. Discussion Overall, our data demonstrated that CT scan and DLCOc identified patients with L-TPD after COVID-19. This condition was associated with vascular inflammation and impair phagocytosis of virus-antibody immune complexes by reduced FcγRIII expression. In addition, we conclude that COVID-19 survivors required a personalized follow-up and adequate intervention to reduce long-term sequelae and the appearance of further metabolic diseases.
Collapse
Affiliation(s)
- Sergio Sanhueza
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Mabel A. Vidal
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
- Facultad de Ingeniería, Diseño y Arquitectura, Universidad San Sebastián, Concepción, Chile
| | | | - Mario E. Henriquez-Beltran
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
- Núcleo de Investigación en Ciencias de la Salud, Universidad Adventista de Chile, Chillán, Chile
- Kinesiology School, Escuela de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
| | - Camilo Cabrera
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Romina Quiroga
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Bárbara E. Antilef
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Kevin P. Aguilar
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Daniela A. Castillo
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Faryd J. Llerena
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Marco Fraga Figueroa
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Mauricio Nazal
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Eritson Castro
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Paola Lagos
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Alexa Moreno
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Jaime J. Lastra
- Internal Medicine Department, Hospital Guillermo Grant Benavente and Medicine Faculty, University of Concepción, Concepción, Chile
| | - Jorge Gajardo
- Internal Medicine Department, Hospital Guillermo Grant Benavente and Medicine Faculty, University of Concepción, Concepción, Chile
| | - Pamela Garcés
- Internal Medicine Department, Hospital Guillermo Grant Benavente and Medicine Faculty, University of Concepción, Concepción, Chile
| | | | | | - Rocío Sanhueza
- Kinesiology School, Escuela de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile
| | - Valeska Ormazába
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Pablo Saldivia
- Division of Biotechnology, MELISA Institute, San Pedro de la Paz, Chile
| | - Cristian Vargas
- Division of Biotechnology, MELISA Institute, San Pedro de la Paz, Chile
| | - Guillermo Nourdin
- Division of Biotechnology, MELISA Institute, San Pedro de la Paz, Chile
| | - Elard Koch
- Division of Biotechnology, MELISA Institute, San Pedro de la Paz, Chile
| | - Felipe A. Zuñiga
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Liliana Lamperti
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Paula Bustos
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Enrique Guzmán-Gutiérrez
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Claudio A. Tapia
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| | - Luciano Ferrada
- CMA Bío-Bío - Advanced Microscopy Center, University of Concepción, Concepción, Chile
| | - Gustavo Cerda
- CMA Bío-Bío - Advanced Microscopy Center, University of Concepción, Concepción, Chile
| | - Ute Woehlbier
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Erick Riquelme
- Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Maria-Isabel Yuseff
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Braulio A. Muñoz Ramirez
- Department of Pharmacology and Toxicology, School of Medicine, Indiana University Bloomington, Bloomington, IN, United States
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - David De Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Medicine and Biomedical Science Faculty, The University of Queensland, Brisbane, QLD, Australia
| | - Ricardo A. Verdugo
- Instituto de Investigación Interdisciplinaria y Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Luis A. Quiñones
- Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Pharmaceutical Sciences and Technology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | - Alicia Colombo
- Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
- Servicio de Anatomía Patológica, Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Maria I. Barría
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
- Internal Medicine, Complejo Asistencial Dr. Víctor Ríos Ruiz, Los Ángeles, Chile
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, University of Concepción, Concepción, Chile
| |
Collapse
|
113
|
Todsen T, Tolsgaard MG, Benfield T, Folke F, Jakobsen KK, Gredal NT, Ersbøll AK, von Buchwald C, Kirkby N. Higher SARS-CoV-2 detection of oropharyngeal compared with nasopharyngeal or saliva specimen for molecular testing: a multicentre randomised comparative accuracy study. Thorax 2023; 78:1028-1034. [PMID: 37208187 PMCID: PMC10511974 DOI: 10.1136/thorax-2022-219599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 04/22/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Testing is critical for detecting SARS-CoV-2 infection, but the best sampling method remains unclear. OBJECTIVES To determine whether nasopharyngeal swab (NPS), oropharyngeal swab (OPS) or saliva specimen collection has the highest detection rate for SARS-CoV-2 molecular testing. METHODS We conducted a randomised clinical trial at two COVID-19 outpatient test centres where NPS, OPS and saliva specimens were collected by healthcare workers in different orders for reverse transcriptase PCR testing. The SARS-CoV-2 detection rate was calculated as the number positive by a specific sampling method divided by the number in which any of the three sampling methods was positive. As secondary outcomes, test-related discomfort was measured with an 11-point numeric scale and cost-effectiveness was calculated. RESULTS Among 23 102 adults completing the trial, 381 (1.65%) were SARS-CoV-2 positive. The SARS-CoV-2 detection rate was higher for OPSs, 78.7% (95% CI 74.3 to 82.7), compared with NPSs, 72.7% (95% CI 67.9 to 77.1) (p=0.049) and compared with saliva sampling, 61.9% (95% CI 56.9 to 66.8) (p<0.001). The discomfort score was highest for NPSs, at 5.76 (SD, 2.52), followed by OPSs, at 3.16 (SD 3.16) and saliva samples, at 1.03 (SD 18.8), p<0.001 between all measurements. Saliva specimens were associated with the lowest cost, and the incremental costs per detected SARS-CoV-2 infection for NPSs and OPSs were US$3258 and US$1832, respectively. CONCLUSIONS OPSs were associated with higher SARS-CoV-2 detection and lower test-related discomfort than NPSs for SARS-CoV-2 testing. Saliva sampling had the lowest SARS-CoV-2 detection but was the least costly strategy for mass testing. TRIAL REGISTRATION NUMBER NCT04715607.
Collapse
Affiliation(s)
- Tobias Todsen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Otorhinolaryngology and Maxillofacial Surgery, Zealand University Hospital, Koege, Denmark
| | - Martin G Tolsgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Academy for Medical Education and Simulation, Rigshospitalet, Copenhagen, Denmark
- Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Benfield
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark
| | - Fredrik Folke
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Copenhagen Emergency Medical Services, Capital region of Denmark, Copenhagen, Denmark
| | - Kathrine K Jakobsen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Niels Tobias Gredal
- Copenhagen Emergency Medical Services, Capital region of Denmark, Copenhagen, Denmark
| | - Annette K Ersbøll
- Copenhagen Emergency Medical Services, Capital region of Denmark, Copenhagen, Denmark
- University of Southern Denmark National Institute of Public Health, Copenhagen, Hovedstaden, Denmark
| | - Christian von Buchwald
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nikolai Kirkby
- Department of Clinical Microbiology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
114
|
Wareham NE, Hamm SR, Liebermann RH, Møller DL, Laursen-Keldorff LB, Poulsen AR, Lund TK, Jensen K, Schultz HHL, Perch M, Nielsen SD. Incidence and severity of SARS-CoV-2 infection in lung transplant recipients in the Omicron era. JHLT OPEN 2023; 1:100004. [PMID: 40144583 PMCID: PMC11935381 DOI: 10.1016/j.jhlto.2023.100004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may cause serious illness in lung transplant recipients. We aimed to investigate incidence and severity of SARS-CoV-2 infection in lung transplant recipients in the Omicron era. We conducted a retrospective study investigating COVID-19 incidence and outcomes among lung transplant recipients between December 27, 2021, and October 31, 2022, in Denmark. We performed COX regression analysis of potential risk factors with hospitalization as an endpoint. Among 236 included patients, 108 had a first positive SARS-CoV-2 polymerase chain reaction during a total of 133 person-years of follow-up, resulting in an incidence rate of 813 per 1000 person-years (95% confidence intervals (CI) 670-977). The cumulative incidence of hospitalization was 24.1% (95% CI 26-32.1) and admission to the intensive care unit was 3.7% (95% CI 0.1-6.3). The 30-day mortality of recipients with a SARS-CoV-2 infection was 0.9% (95% CI 0-2.7). We found that the incidence rate of patients with SARS-CoV-2 infection was markedly higher, whereas the mortality rate was lower in the omicron era compared to earlier reports for lung transplant recipients conducted in the delta era. On the other hand, a substantial proportion of patients were hospitalized, suggesting a continuous impact on this patient population.
Collapse
Affiliation(s)
- Neval Ete Wareham
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sebastian Rask Hamm
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Regitze Hertz Liebermann
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Dina Leth Møller
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Laurids Brandt Laursen-Keldorff
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Andreas Runge Poulsen
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Kromann Lund
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kristine Jensen
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Henrik L. Schultz
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
115
|
Arons E, Henry K, Haas C, Gould M, Tsintolas J, Mauter J, Zhou H, Burbelo PD, Cohen JI, Kreitman RJ. Characterization of B-cell receptor clonality and immunoglobulin gene usage at multiple time points during active SARS-CoV-2 infection. J Med Virol 2023; 95:e29179. [PMID: 37877800 PMCID: PMC11323229 DOI: 10.1002/jmv.29179] [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: 08/08/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023]
Abstract
Although monoclonal antibodies to the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are known, B-cell receptor repertoire and its change in patients during coronavirus disease-2019 (COVID-19) progression is underreported. We aimed to study this molecularly. We used immunoglobulin heavy chain (IGH) variable region (IGHV) spectratyping and next-generation sequencing of peripheral blood B-cell genomic DNA collected at multiple time points during disease evolution to study B-cell response to SARS-CoV-2 infection in 14 individuals with acute COVID-19. We found a broad distribution of responding B-cell clones. The IGH gene usage was not significantly skewed but frequencies of individual IGH genes changed repeatedly. We found predominant usage of unmutated and low mutation-loaded IGHV rearrangements characterizing naïve and extrafollicular B cells among the majority of expanded peripheral B-cell clonal lineages at most tested time points in most patients. IGH rearrangement usage showed no apparent relation to anti-SARS-CoV-2 antibody titers. Some patients demonstrated mono/oligoclonal populations carrying highly mutated IGHV rearrangements indicating antigen experience at some of the time points tested, including even before anti-SARS-CoV-2 antibodies were detected. We present evidence demonstrating that the B-cell response to SARS-CoV-2 is individual and includes different lineages of B cells at various time points during COVID-19 progression.
Collapse
Affiliation(s)
- Evgeny Arons
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| | - Kiersten Henry
- Medstar Montgomery Medical Center, 18101 Prince Philip Drive, Olney, MD 20832, United States
| | - Christopher Haas
- Medstar Franklin Square Medical Center, 9000 Franklin Square Drive, Baltimore, MD 21237, United States
| | - Mory Gould
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| | - Jack Tsintolas
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| | - Jack Mauter
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| | - Hong Zhou
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| | - Peter D. Burbelo
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892, United States
| | - Jeffrey I. Cohen
- Laboratory of Infectious Disease, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, United States
| | - Robert J. Kreitman
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, 20892, United States
| |
Collapse
|
116
|
Martínez-Gómez LE, Martinez-Armenta C, Medina-Luna D, Ordoñez-Sánchez ML, Tusie-Luna T, Ortega-Peña S, Herrera-López B, Suarez-Ahedo C, Jimenez-Gutierrez GE, Hidalgo-Bravo A, Vázquez-Cárdenas P, Vidal-Vázquez RP, Ramírez-Hinojosa JP, Martinez Matsumoto PM, Vargas-Alarcón G, Posadas-Sánchez R, Fragoso JM, Martínez-Ruiz FDJ, Zayago-Angeles DM, Mata-Miranda MM, Vázquez-Zapién GJ, Martínez-Cuazitl A, Andrade-Alvarado J, Granados J, Ramos-Tavera L, Camacho-Rea MDC, Segura-Kato Y, Rodríguez-Pérez JM, Coronado-Zarco R, Franco-Cendejas R, López-Jácome LE, Magaña JJ, Vela-Amieva M, Pineda C, Martínez-Nava GA, López-Reyes A. Implication of myddosome complex genetic variants in outcome severity of COVID-19 patients. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:939-950. [PMID: 37365052 PMCID: PMC10273757 DOI: 10.1016/j.jmii.2023.06.002] [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: 09/06/2022] [Revised: 03/31/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND/PURPOSE(S) During a viral infection, the immune response is mediated by the toll-like receptors and myeloid differentiation Factor 88 (MyD88) that play an important role sensing infections such as SARS-CoV-2 which has claimed the lives of more than 6.8 million people around the world. METHODS We carried out a cross-sectional with a population of 618 SARS-CoV-2-positive unvaccinated subjects and further classified based on severity: 22% were mild, 34% were severe, 26% were critical, and 18% were deceased. Toll Like Receptor 7 (TLR7) single-nucleotide polymorphisms (rs3853839, rs179008, rs179009, and rs2302267) and MyD88 (rs7744) were genotyped using TaqMan OpenArray. The association of polymorphisms with disease outcomes was performed by logistic regression analysis adjusted by covariates. RESULTS A significant association of rs3853839 and rs7744 of the TLR7 and MyD88 genes, respectively, was found with COVID-19 severity. The G/G genotype of the rs3853839 TLR7 was associated with the critical outcome showing an Odd Ratio = 1.98 (95% IC = 1.04-3.77). The results highlighted an association of the G allele of MyD88 gene with severe, critical and deceased outcomes. Furthermore, in the dominant model (AG + GG vs. AA), we observed an Odd Ratio = 1.70 (95% CI = 1.02-2.86) with severe, Odd Ratio = 1.82 (95% CI = 1.04-3.21) with critical, and Odd Ratio = 2.44 (95% CI = 1.21-4.9) with deceased outcomes. CONCLUSION To our knowledge this work represents an innovative report that highlights the significant association of TLR7 and MyD88 gene polymorphisms with COVID-19 outcomes and the possible implication of the MyD88 variant with D-dimer and IFN-α concentrations.
Collapse
Affiliation(s)
- Laura E Martínez-Gómez
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Carlos Martinez-Armenta
- Graduate Program in Experimental Biology, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, Mexico.
| | - Daniel Medina-Luna
- Microbiology & Immunology Department, Dalhousie University, Halifax, B3H4R2, Nova Scotia, Canada.
| | - María Luisa Ordoñez-Sánchez
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico.
| | - Tere Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico; Instituto de Investigaciones Biomédicas Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Silvestre Ortega-Peña
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Brígida Herrera-López
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Carlos Suarez-Ahedo
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Guadalupe Elizabeth Jimenez-Gutierrez
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Alberto Hidalgo-Bravo
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | - Rosa P Vidal-Vázquez
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | - Juan P Ramírez-Hinojosa
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | | | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - Rosalinda Posadas-Sánchez
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - José-Manuel Fragoso
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | | | | | - Mónica Maribel Mata-Miranda
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Gustavo Jesús Vázquez-Zapién
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Adriana Martínez-Cuazitl
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Javier Andrade-Alvarado
- Servicio de Cirugía General, Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico.
| | - Julio Granados
- Departamento de Inmunogenética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico.
| | - Luis Ramos-Tavera
- Departamento de Inmunogenética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico.
| | - María Del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico
| | - Yayoi Segura-Kato
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico.
| | - José Manuel Rodríguez-Pérez
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - Roberto Coronado-Zarco
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Rafael Franco-Cendejas
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Luis Esau López-Jácome
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Jonathan J Magaña
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaria de Salud, Ciudad de México, Mexico.
| | - Carlos Pineda
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| |
Collapse
|
117
|
Bayuo J, Wong FKY. Transitional and aftercare needs of persons recovering from COVID-19 using the Omaha System: A Scoping Review. J Clin Nurs 2023; 32:6894-6916. [PMID: 37353967 DOI: 10.1111/jocn.16798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 08/09/2022] [Accepted: 06/01/2023] [Indexed: 06/25/2023]
Abstract
AIM To identify and classify the transitional and aftercare needs of persons hospitalised with and recovering from COVID-19. BACKGROUND Several studies exist that describe the patient needs at the acute phase of COVID-19. The transitional and aftercare needs that emerge during recovery, however, remain vague. METHODS A scoping review was conducted and reported according to the PRISMA extension guidelines for scoping reviews (PRISMA-ScR). Primary studies were identified from database search. Narrative synthesis was undertaken, with the Omaha System as a framework. RESULTS Forty studies were included. Persons recovering from the infection may have several needs in all domains of the Omaha System. Although the severity and persistence of the needs may be unrelated to the severity of the initial infection, they may vary based on factors such as age and pre-morbid factors. CONCLUSION Recovering from COVID-19 is associated with varied biopsychosocial-environmental needs which can adversely affect the quality-of-life experience. The review findings represent an inventory of needs that can guide the development of multi-disciplinary post-acute or aftercare programmes. RELEVANCE TO CLINICAL PRACTICE Recovering from COVID-19 can be a protracted process requiring ongoing professional support after discharge. Policies are required to support the development and implementation of post-acute programmes of care. Comprehensive transitional and aftercare rehabilitative programmes are needed to support the recovery process.
Collapse
Affiliation(s)
- Jonathan Bayuo
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, China
| | | |
Collapse
|
118
|
Abstract
COVID-19, the illness caused by SARS-CoV-2, became a worldwide pandemic in 2020. Initial clinical manifestations range from asymptomatic infection to mild upper respiratory illness but may progress to pulmonary involvement with hypoxemia and, in some cases, multiorgan involvement, shock, and death. Older adults, pregnant persons, those with common comorbidities, and those with immunosuppression are at greatest risk for progression. Vaccination is effective in preventing symptomatic infection and reducing risk for severe disease, hospitalization, and death. Antiviral treatment and immunomodulators have been shown to benefit certain patients. This article summarizes current recommendations on prevention, diagnosis, management, and treatment of COVID-19.
Collapse
Affiliation(s)
| | - Roy M Gulick
- Weill Cornell Medicine, New York, New York (K.M.M., R.M.G.)
| |
Collapse
|
119
|
Guo BC, Wu KH, Chen CY, Lin WY, Chang YJ, Lee TA, Lin MJ, Wu HP. Mesenchymal Stem Cells in the Treatment of COVID-19. Int J Mol Sci 2023; 24:14800. [PMID: 37834246 PMCID: PMC10573267 DOI: 10.3390/ijms241914800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Since the emergence of the coronavirus disease 2019 (COVID-19) pandemic, many lives have been tragically lost to severe infections. The COVID-19 impact extends beyond the respiratory system, affecting various organs and functions. In severe cases, it can progress to acute respiratory distress syndrome (ARDS) and multi-organ failure, often fueled by an excessive immune response known as a cytokine storm. Mesenchymal stem cells (MSCs) have considerable potential because they can mitigate inflammation, modulate immune responses, and promote tissue regeneration. Accumulating evidence underscores the efficacy and safety of MSCs in treating severe COVID-19 and ARDS. Nonetheless, critical aspects, such as optimal routes of MSC administration, appropriate dosage, treatment intervals, management of extrapulmonary complications, and potential pediatric applications, warrant further exploration. These research avenues hold promise for enriching our understanding and refining the application of MSCs in confronting the multifaceted challenges posed by COVID-19.
Collapse
Affiliation(s)
- Bei-Cyuan Guo
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chun-Yu Chen
- Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 43503, Taiwan;
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Wen-Ya Lin
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung 43503, Taiwan
| | - Yu-Jun Chang
- Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Tai-An Lee
- Department of Emergency Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan;
| | - Mao-Jen Lin
- Division of Cardiology, Department of Medicine, Taichung Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97002, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
120
|
Gedefie A, Birara T, Misganaw S, Bambo GM, Kebede SS, Tilahun M, Mohammed O, Kassa Y, Bisetegn H, Alemayehu E. Clinical profiles, epidemiological characteristics and treatment outcomes of COVID-19 patients in North-eastern Ethiopia: A retrospective cohort study. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0002285. [PMID: 37729157 PMCID: PMC10511068 DOI: 10.1371/journal.pgph.0002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND COVID-19 is a rapidly emerging global health threat and economic disaster. The epidemiology and outcomes of COVID-19 patients in Ethiopia are scarce. Thus, the present study aimed to assess clinical profiles, epidemiological characteristics, and treatment outcomes of patients with COVID-19 and to identify determinants of the disease outcome among COVID-19 patients in North-eastern Ethiopia. METHODS A retrospective observational cohort study was conducted in North-eastern Ethiopia, from May 2020 to Jan 2022 on a total of 364 SARS-COV-2 infected patients. Demographic and clinical data were abstracted from the medical records of patients. Bivariable and multivariable analyses were conducted to determine the factors associated with the mortality of COVID-19 patients and variables with a P-value < 0.05 were considered statistically significant. RESULT Among 364 COVID-19 patients included in this study, two-thirds (68.1%) were males with a median age of 34 years. The majority; 42.9% & 33.0% respectively cases were detected at the health facility and community level surveillance. Furthermore, 6.6% of patients had pre-existing comorbidities of which diabetes mellitus (23.1%) and hypertension (15.3%) had the highest frequency. The symptomatic rate of COVID-19 patients was 30.5%. The most common clinical presentations were cough (26.9%), fever (26.1%), and shortness of breath (15.2%). Moreover, the mortality rate of COVID-19 patients was 4.1% which was independently predicted by a history of underlining co-morbidity (AHR:6.09; 95%CI:1.299-28.56; P = 0.022) and a history of severe or critical conditions (AHR 11.8; 95%CI:4.89-28.83; P = 0.003). CONCLUSION Severe or critical acute COVID-19 and underlining comorbidities are associated with higher mortality. Therefore, critical follow-up and management should be given to patients with underlying diseases is required.
Collapse
Affiliation(s)
- Alemu Gedefie
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Tadesse Birara
- Department of Information System, College of Informatics, Wollo University, Kombolcha, Ethiopia
| | - Sisay Misganaw
- Department of Information System, College of Informatics, Wollo University, Kombolcha, Ethiopia
| | - Getachew Mesfin Bambo
- Department of Medical Laboratory Sciences, College of Health Sciences, Mizan-Tepi University, Mizan, Ethiopia
| | - Samuel Sahile Kebede
- Department of Medical Laboratory Sciences, College of Health Sciences, Mizan-Tepi University, Mizan, Ethiopia
| | - Mihret Tilahun
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Ousman Mohammed
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Yeshimebet Kassa
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Habtye Bisetegn
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Ermiyas Alemayehu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
121
|
Cheng Z, Cai Y, Zhang K, Zhang J, Gui H, Luo YS, Zhou J, DeVeale B. MAP3K19 regulatory variation in populations with African ancestry may increase COVID-19 severity. iScience 2023; 26:107555. [PMID: 37649700 PMCID: PMC10462844 DOI: 10.1016/j.isci.2023.107555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/20/2023] [Accepted: 08/03/2023] [Indexed: 09/01/2023] Open
Abstract
To identify ancestry-linked genetic risk variants associated with COVID-19 hospitalization, we performed an integrative analysis of two genome-wide association studies and resolved four single nucleotide polymorphisms more frequent in COVID-19-hospitalized patients with non-European ancestry. Among them, the COVID-19 risk SNP rs16831827 shows the largest difference in minor allele frequency (MAF) between populations with African and European ancestry and also shows higher MAF in hospitalized COVID-19 patients among cohorts of mixed ancestry (odds ratio [OR] = 1.20, 95% CI: 1.10-1.30) and entirely African ancestry (OR = 1.30, 95% CI: 1.02-1.67). rs16831827 is an expression quantitative trait locus of MAP3K19. MAP3K19 expression is induced during ciliogenesis and most abundant in ciliated tissues including lungs. Single-cell RNA sequencing analyses revealed that MAP3K19 is highly expressed in multiple ciliated cell types. As rs16831827∗T is associated with reduced MAP3K19 expression, it may increase the risk of severe COVID-19 by reducing MAP3K19 expression.
Collapse
Affiliation(s)
- Zhongshan Cheng
- Center for Applied Bioinformatics, St Jude Children’s Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen 518000, China
| | - Ke Zhang
- The Key and Characteristic Laboratory of Modern Pathogenicity Biology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, Guiyang 550025, China
| | - Jingxuan Zhang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Hongsheng Gui
- Behavioral Health Services and Psychiatry Research, Henry Ford Health, Detroit, MI 48202, USA
| | - Yu-Si Luo
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang 550004, China
| | - Jie Zhou
- Department of Microbiology, The University of Hong Kong, Hong Kong 999077, China
| | - Brian DeVeale
- The Department of Biomedical Sciences, University of Windsor, Windsor, ON N9B 3P4, Canada
| |
Collapse
|
122
|
Gonzaga A, Andreu E, Hernández-Blasco LM, Meseguer R, Al-Akioui-Sanz K, Soria-Juan B, Sanjuan-Gimenez JC, Ferreras C, Tejedo JR, Lopez-Lluch G, Goterris R, Maciá L, Sempere-Ortells JM, Hmadcha A, Borobia A, Vicario JL, Bonora A, Aguilar-Gallardo C, Poveda JL, Arbona C, Alenda C, Tarín F, Marco FM, Merino E, Jaime F, Ferreres J, Figueira JC, Cañada-Illana C, Querol S, Guerreiro M, Eguizabal C, Martín-Quirós A, Robles-Marhuenda Á, Pérez-Martínez A, Solano C, Soria B. Rationale for combined therapies in severe-to-critical COVID-19 patients. Front Immunol 2023; 14:1232472. [PMID: 37767093 PMCID: PMC10520558 DOI: 10.3389/fimmu.2023.1232472] [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: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
An unprecedented global social and economic impact as well as a significant number of fatalities have been brought on by the coronavirus disease 2019 (COVID-19), produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Acute SARS-CoV-2 infection can, in certain situations, cause immunological abnormalities, leading to an anomalous innate and adaptive immune response. While most patients only experience mild symptoms and recover without the need for mechanical ventilation, a substantial percentage of those who are affected develop severe respiratory illness, which can be fatal. The absence of effective therapies when disease progresses to a very severe condition coupled with the incomplete understanding of COVID-19's pathogenesis triggers the need to develop innovative therapeutic approaches for patients at high risk of mortality. As a result, we investigate the potential contribution of promising combinatorial cell therapy to prevent death in critical patients.
Collapse
Affiliation(s)
- Aitor Gonzaga
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute of Bioengineering, Miguel Hernández University, Elche, Spain
| | - Etelvina Andreu
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Applied Physics Department, Miguel Hernández University, Elche, Spain
| | | | - Rut Meseguer
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinic University Hospital, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA) Health Research Institute, Valencia, Spain
| | - Karima Al-Akioui-Sanz
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Bárbara Soria-Juan
- Réseau Hospitalier Neuchâtelois, Hôpital Pourtalès, Neuchâtel, Switzerland
| | | | - Cristina Ferreras
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, University Pablo de Olavide, Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Guillermo Lopez-Lluch
- University Pablo de Olavide, Centro Andaluz de Biología del Desarrollo - Consejo Superior de Investigaciones Científicas (CABD-CSIC), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Sevilla, Spain
| | - Rosa Goterris
- Clinic University Hospital, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA) Health Research Institute, Valencia, Spain
| | - Loreto Maciá
- Nursing Department, University of Alicante, Alicante, Spain
| | - Jose M. Sempere-Ortells
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Biotechnology Department, University of Alicante, Alicante, Spain
| | - Abdelkrim Hmadcha
- Department of Molecular Biology and Biochemical Engineering, University Pablo de Olavide, Seville, Spain
- Biosanitary Research Institute (IIB-VIU), Valencian International University (VIU), Valencia, Spain
| | - Alberto Borobia
- Clinical Pharmacology Department, La Paz University Hospital, School of Medicine, Universidad Autónoma de Madrid, IdiPAz, Madrid, Spain
| | - Jose L. Vicario
- Transfusion Center of the Autonomous Community of Madrid, Madrid, Spain
| | - Ana Bonora
- Health Research Institute Hospital La Fe, Valencia, Spain
| | | | - Jose L. Poveda
- Health Research Institute Hospital La Fe, Valencia, Spain
| | - Cristina Arbona
- Valencian Community Blood Transfusion Center, Valencia, Spain
| | - Cristina Alenda
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Fabian Tarín
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Francisco M. Marco
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Immunology Department, Dr. Balmis General University Hospital, Alicante, Spain
| | - Esperanza Merino
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Department of Clinical Medicine, Miguel Hernández University, Elche, Spain
- Infectious Diseases Unit, Dr. Balmis General University Hospital, Alicante, Spain
| | - Francisco Jaime
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - José Ferreres
- Intensive Care Service, Hospital Clinico Universitario, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA), Valencia, Spain
| | | | | | | | - Manuel Guerreiro
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Cristina Eguizabal
- Research Unit, Basque Center for Blood Transfusion and Human Tissues, Galdakao, Spain
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | | | - Antonio Pérez-Martínez
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Solano
- Hematology Service, Hospital Clínico Universitario, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA), Valencia, Spain
| | - Bernat Soria
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute of Bioengineering, Miguel Hernández University, Elche, Spain
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| |
Collapse
|
123
|
Siripongboonsitti T, Muadchimkaew M, Tawinprai K, Issaranon O, Meepholkij W, Arttawejkul P, Vararungzarit A, Thavornwattana K, Mahanonda N. Favipiravir treatment in non-severe COVID-19: promising results from multicenter propensity score-matched study (FAVICOV). Sci Rep 2023; 13:14884. [PMID: 37689754 PMCID: PMC10492810 DOI: 10.1038/s41598-023-42195-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
This study aimed to evaluate the efficacy of favipiravir (FPV) in preventing the development of severe COVID-19 in patients with mild-to-moderate symptoms. The study evaluated 1037 COVID-19 patients treated with FPV or standard treatment between April and September 2021, analyzed by propensity score matching. 149 patients were included in each arm after propensity score matching. The clinical outcomes showed no deterioration of the WHO clinical progression scale in the FPV group compared to the standard treatment group on day 5 (83.2% vs. 69.1%, p < 0.001). The WHO clinical progression scale also showed improvements on day 14 in the FPV group compared to the standard treatment group (66.4% vs. 46.3%, p < 0.001). The rates of oxygen supplementation and hospitalization were significantly lower in the FPV group compared to the standard treatment group (0% vs. 12.1% and 0.7% vs. 17.4%, respectively, p < 0.001 for both). There were no differences in adverse events between the two groups. The study highlights the effectiveness of FPV in preventing severe COVID-19 and hospitalization in patients with mild-to-moderate symptoms. The findings emphasize the importance of personalized treatment plans for COVID-19 patients, starting FPV treatment early, and adjusting dosages based on ethnicity and body weight.
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.
- Infection Control Unit, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand.
| | - Marisa Muadchimkaew
- Infection Control Unit, Chulabhorn Hospital, 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
- Infection Control Unit, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | | | - Pureepat Arttawejkul
- Department of Medicine, Faculty of Medicine, Burapha University Hospital, Chonburi, Thailand
| | - Apiradee Vararungzarit
- Department of Medicine, Faculty of Medicine, Burapha University Hospital, Chonburi, Thailand
| | - Kaewklao Thavornwattana
- Health Data Science Unit, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Nithi Mahanonda
- Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
124
|
Herman JD, Atyeo C, Zur Y, Cook CE, Patel NJ, Vanni KM, Kowalski EN, Qian G, Srivatsan S, Shadick NA, Rao DA, Kellman B, Mann CJ, Lauffenburger D, Wallace ZS, Sparks JA, Alter G. Humoral immunity to an endemic coronavirus is associated with postacute sequelae of COVID-19 in individuals with rheumatic diseases. Sci Transl Med 2023; 15:eadf6598. [PMID: 37672567 PMCID: PMC10764151 DOI: 10.1126/scitranslmed.adf6598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 08/05/2023] [Indexed: 09/08/2023]
Abstract
Beyond the acute illness caused by severe acute respiratory coronavirus 2 (SARS-CoV-2) infection, about one-fifth of infections result in long-term persistence of symptoms despite the apparent clearance of infection. Insights into the mechanisms that underlie postacute sequelae of COVID-19 (PASC) will be critical for the prevention and clinical management of long-term complications of COVID-19. Several hypotheses have been proposed that may account for the development of PASC, including persistence of virus and dysregulation of immune responses. Among the immunological changes noted in PASC, alterations in humoral immunity have been observed in some patient subsets. To begin to determine whether SARS-CoV-2- or other pathogen-specific humoral immune responses evolve uniquely in PASC, we performed comprehensive antibody profiling against SARS-CoV-2, a panel of endemic pathogens, and a panel of routine vaccine antigens using systems serology in two cohorts of patients with preexisting systemic autoimmune rheumatic disease (SARD) who either developed or did not develop PASC. A distinct qualitative shift observed in Fcγ receptor (FcγR) binding was observed in individuals with PASC. Specifically, individuals with PASC harbored weaker FcγR-binding anti-SARS-CoV-2 antibodies and stronger FcγR-binding antibody responses against the endemic coronavirus OC43. Individuals with PASC developed an OC43 S2-specific antibody response with stronger FcγR binding, linked to cross-reactivity across SARS-CoV-2 and common coronaviruses. These findings identify previous coronavirus imprinting as a potential marker for the development of PASC in individuals with SARDs.
Collapse
Affiliation(s)
- Jonathan D Herman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Yonatan Zur
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Claire E Cook
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Naomi J Patel
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kathleen M Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Emily N Kowalski
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Grace Qian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Shruthi Srivatsan
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nancy A Shadick
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Benjamin Kellman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Colin J Mann
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zachary S Wallace
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
125
|
Han C, Jang H, Oh J. Excess mortality during the Coronavirus disease pandemic in Korea. BMC Public Health 2023; 23:1698. [PMID: 37660007 PMCID: PMC10474701 DOI: 10.1186/s12889-023-16546-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/17/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Although the ongoing epidemics of Coronavirus disease 2019 (COVID-19) may have affected the mortality trend of the nation, the national level assessment of excess mortality (changes in overall mortality in the entire population) is still scarce in Korea. Therefore, this study evaluated the excess mortality during the COVID-19 pandemic in Korea using the certified mortality data. METHODS Monthly mortality and population data from January 2013 to June 2022 was obtained from the National Health Insurance Service database and Statistics Korea. A quasi-Poisson interrupted time-series model adjusted for age structure, population, seasonality, and long-term trends was used to estimate the counterfactual projections (expected) of mortality during the COVID-19 pandemic (March 2020 to June 2022). The absolute difference (observed-expected) and ratio (observed / expected) of mortality were calculated. Stratified analysis based on pandemic years (years 2020, 2021, and 2022), sex, and age groups (aged 0-4, 5-19, 20-64, and ≥ 65 years) were conducted. RESULTS An 8.7% increase in mortality was observed during the COVID-19 pandemic [absolute difference: 61,277 persons; ratio (95% confidence interval (CI)): 1.087 (1.066, 1.107)]. The gap between observed and estimated mortality became wider with continuation of the pandemic [ratio (95% CI), year 2020: 1.021 (1.003, 1.040); year 2021: 1.060 (1.039, 1.080), year 2022: 1.244 (1.219, 1.270)]. Although excess mortality across sex was similar, the adult [aged 20-64, ratio (95% CI): 1.059 (1.043, 1.076)] and elderly [aged 65-, ratio (95% CI): 1.098 (1.062, 1.135)] population showed increased excess mortality during the pandemic. CONCLUSIONS Despite Korea's successful quarantine policy response, the continued epidemic has led to an excess mortality. The estimated mortality exceeded the number of deaths from COVID-19 infection. Excess mortality should be monitored to estimate the overall impact of the pandemic on a nation.
Collapse
Affiliation(s)
- Changwoo Han
- Department of Preventive Medicine, Chungnam National University College of Medicine, 266, Munhwa-Ro, Jung-Gu, Daejeon, 35015, Korea.
| | - Hoyeon Jang
- Department of Big Data Strategy, National Health Insurance Service, Wonju, Korea
| | - Juhwan Oh
- Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
126
|
Luxenburger H, Thimme R. SARS-CoV-2 and the liver: clinical and immunological features in chronic liver disease. Gut 2023; 72:1783-1794. [PMID: 37316169 PMCID: PMC10423489 DOI: 10.1136/gutjnl-2023-329623] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/24/2023] [Indexed: 06/16/2023]
Abstract
SARS-CoV-2 infection may affect the liver in healthy individuals but also influences the course of COVID-19 in patients with chronic liver disease (CLD). As described in healthy individuals, a strong SARS-CoV-2-specific adaptive immune response is important for the outcome of COVID-19, however, knowledge on the adaptive immune response in CLD is limited.Here, we review the clinical and immunological features of SARS-CoV-2 infection in individuals with CLD. Acute liver injury occurs in many cases of SARS-CoV-2 infection and may be induced by multiple factors, such as cytokines, direct viral infection or toxic effects of COVID-19 drugs. In individuals with CLD, SARS-CoV-2 infection may have a more severe course and promote decompensation and particularly in patients with cirrhosis. Compared with healthy individuals, the SARS-CoV-2-specific adaptive immune responses is impaired in patients with CLD after both, natural infection and vaccination but improves at least partially after booster vaccination.Following SARS-CoV-2 vaccination, rare cases of acute vaccine-induced liver injury and the development of autoimmune-like hepatitis have been reported. However, the concomitant elevation of liver enzymes is reversible under steroid treatment.
Collapse
Affiliation(s)
- Hendrik Luxenburger
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
127
|
Ardellier FD, Baloglu S, Sokolska M, Noblet V, Lersy F, Collange O, Ferré JC, Maamar A, Carsin-Nicol B, Helms J, Schenck M, Khalil A, Gaudemer A, Caillard S, Pottecher J, Lefèbvre N, Zorn PE, Matthieu M, Brisset JC, Boulay C, Mutschler V, Hansmann Y, Mertes PM, Schneider F, Fafi-Kremer S, Ohana M, Meziani F, Meyer N, Yousry T, Anheim M, Cotton F, Jäger HR, Kremer S, SFNR-COVID group 2BonnevilleFabricezAdamGilleszMartin-BlondelGuillaumezParienteJérémiezGeeraertsThomaszOesterléHélèneaaBologniniFedericoaaMessieJulienaaHmeydiaGhaziabBenzakounJosephabOppenheimCatherineabConstansJean-MarcacMetanbouSergeacHeintzAdrienacBapstBlancheadMegdicheImenadJagerLaviniaaeNesserPatrickaeMbaYannick TallaaeTourdiasThomasafCoutureauJulietteafHemmertCélineagFeuersteinPhilippeagSebagNathanagCarreSophieahAllegManelahLecocqClaireahSchmittEmmanuelaiAnxionnatRenéaiZhuFrançoisaiForestierGéraudajRouchaudAymericajCombyPierre-OlivierakRicolfiFredericakThouantPierreakGrandSylviealKrainikAlexandrealde BeaurepaireIsaureamBornetGrégoireamLacalmAudreyanMiailhesPatrickanPiqueJulieanBoutetClaireaoFabreXavieraoClaiseBéatriceapMirafzalSoniaapCalvetLaureapDesalHubertaqBergeJéromeaqBoulouisGrégoireaqKazemiApollineaqPyatigorskayaNadyaaqLeclerAugustinaqSalemeSuzanaaqEdjlali-GoujonMyriamaqKerlerouxBasileaqBrissetJean-ChristopheaqChenafSamirarToulouse University Hospital, Toulouse, FranceLouis Pasteur Hospital, Colmar, FranceSainte-Anne Hospital, Paris, FranceAmiens University Hospital, Amiens, FranceHenri-Mondor University Hospital, Créteil, FranceCHIC Unisanté, Forbach, FranceBordeaux University Hospital, Bordeaux, FranceEmile Muller Hospital, Mulhouse, FranceHospital of Haguenau, Haguenau, FranceNancy University Hospital, Nancy, FranceLimoges University Hospital, Limoges, FranceDijon University Hospital, Dijon, FranceGrenoble Alpes University Hospital, Grenoble, FranceAntony Private Hospital, Antony, FranceHospices Civils de Lyon, Lyon, FranceSaint-Etienne University Hospital, Saint-Etienne, FranceClermont-Ferrand University Hospital, Clermont-Ferrand, FranceFrench Neuroradiological Society (SFNR) Board, FranceUCIEC, France, Adam G, Martin-Blondel G, Pariente J, Geeraerts T, Oesterlé H, Bolognini F, Messie J, Hmeydia G, Benzakoun J, Oppenheim C, Constans JM, Metanbou S, Heintz A, Bapst B, Megdiche I, Jager L, Nesser P, Mba YT, Tourdias T, Coutureau J, Hemmert C, Feuerstein P, Sebag N, Carre S, Alleg M, Lecocq C, Schmitt E, Anxionnat R, Zhu F, Forestier G, Rouchaud A, Comby PO, Ricolfi F, Thouant P, Grand S, Krainik A, de Beaurepaire I, Bornet G, Lacalm A, Miailhes P, Pique J, Boutet C, Fabre X, Claise B, Mirafzal S, Calvet L, Desal H, Berge J, Boulouis G, Kazemi A, Pyatigorskaya N, Lecler A, Saleme S, Edjlali-Goujon M, Kerleroux B, Brisset JC, Chenaf S. Cerebral perfusion using ASL in patients with COVID-19 and neurological manifestations: A retrospective multicenter observational study. J Neuroradiol 2023; 50:470-481. [PMID: 36657613 PMCID: PMC9842391 DOI: 10.1016/j.neurad.2023.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral hypoperfusion has been reported in patients with COVID-19 and neurological manifestations in small cohorts. We aimed to systematically assess changes in cerebral perfusion in a cohort of 59 of these patients, with or without abnormalities on morphological MRI sequences. METHODS Patients with biologically-confirmed COVID-19 and neurological manifestations undergoing a brain MRI with technically adequate arterial spin labeling (ASL) perfusion were included in this retrospective multicenter study. ASL maps were jointly reviewed by two readers blinded to clinical data. They assessed abnormal perfusion in four regions of interest in each brain hemisphere: frontal lobe, parietal lobe, posterior temporal lobe, and temporal pole extended to the amygdalo-hippocampal complex. RESULTS Fifty-nine patients (44 men (75%), mean age 61.2 years) were included. Most patients had a severe COVID-19, 57 (97%) needed oxygen therapy and 43 (73%) were hospitalized in intensive care unit at the time of MRI. Morphological brain MRI was abnormal in 44 (75%) patients. ASL perfusion was abnormal in 53 (90%) patients, and particularly in all patients with normal morphological MRI. Hypoperfusion occurred in 48 (81%) patients, mostly in temporal poles (52 (44%)) and frontal lobes (40 (34%)). Hyperperfusion occurred in 9 (15%) patients and was closely associated with post-contrast FLAIR leptomeningeal enhancement (100% [66.4%-100%] of hyperperfusion with enhancement versus 28.6% [16.6%-43.2%] without, p = 0.002). Studied clinical parameters (especially sedation) and other morphological MRI anomalies had no significant impact on perfusion anomalies. CONCLUSION Brain ASL perfusion showed hypoperfusion in more than 80% of patients with severe COVID-19, with or without visible lesion on conventional MRI abnormalities.
Collapse
Affiliation(s)
- François-Daniel Ardellier
- Service D'imagerie 2, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France,Engineering science, computer science and imaging laboratory (ICube), Integrative Multimodal Imaging in Healthcare, UMR 7357, University of Strasbourg-CNRS, Strasbourg, France,Corresponding author at: Service D'imagerie 2, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Seyyid Baloglu
- Service D'imagerie 2, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Magdalena Sokolska
- Department of Medical Physics and Biomedical Engineering, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London NW1 2BU, United Kingdom,Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Vincent Noblet
- Engineering science, computer science and imaging laboratory (ICube), Integrative Multimodal Imaging in Healthcare, UMR 7357, University of Strasbourg-CNRS, Strasbourg, France
| | - François Lersy
- Service D'imagerie 2, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Collange
- Service d'Anesthésie-Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | | | - Adel Maamar
- Medical Intensive Care Unit, CHU Rennes, Rennes, France
| | | | - Julie Helms
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France,Immuno-Rhumatologie Moléculaire, INSERM UMR S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg (UNISTRA), Strasbourg, France
| | - Maleka Schenck
- Service de Médecine Intensive Réanimation, Hôpitaux universitaires de Strasbourg, Hautepierre, Strasbourg, France
| | - Antoine Khalil
- Department of Radiology, Assistance Publique–Hôpitaux de Paris (APHP), Denis Diderot University and Medical School, Bichat University Hospital, Paris, France
| | - Augustin Gaudemer
- Neuroradiology Unit, Department of Radiology, Assistance Publique-Hôpitaux de Paris (APHP), Bichat University Hospital, Paris, France
| | - Sophie Caillard
- Immuno-Rhumatologie Moléculaire, INSERM UMR S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg (UNISTRA), Strasbourg, France,Nephrology and Transplantation department, Hôpitaux Universitaires de Strasbourg. Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Julien Pottecher
- Hôpital de Hautepierre, Service d'Anesthésie, Réanimation & Médecine Péri-Opératoire - Université de Strasbourg, Faculté de Médecine, FMTS, EA3072, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nicolas Lefèbvre
- Service de Maladies Infectieuses, NHC, CHU de Strasbourg, Strasbourg, France
| | - Pierre-Emmanuel Zorn
- Hôpitaux Universitaires de Strasbourg, UCIEC, Pôle d'Imagerie, Strasbourg, France
| | - Muriel Matthieu
- Hôpitaux Universitaires de Strasbourg, UCIEC, Pôle d'Imagerie, Strasbourg, France
| | | | - Clotilde Boulay
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Véronique Mutschler
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Yves Hansmann
- Service de Maladies Infectieuses, NHC, CHU de Strasbourg, Strasbourg, France
| | - Paul-Michel Mertes
- Service d'Anesthésie-Réanimation, Nouvel Hôpital Civil, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Francis Schneider
- Service de Médecine Intensive Réanimation, Hôpitaux universitaires de Strasbourg, Hautepierre, Strasbourg, France
| | - Samira Fafi-Kremer
- Laboratoire de Virologie Médicale, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Mickael Ohana
- Radiology Department, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Ferhat Meziani
- Service de Médecine Intensive Réanimation, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France,UMR 1260, Regenerative Nanomedicine (RNM), FMTS, INSERM (French National Institute of Health and Medical Research), Strasbourg, France
| | - Nicolas Meyer
- Service de Santé Publique, GMRC, CHU de Strasbourg, Strasbourg F-67091 , France
| | - Tarek Yousry
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Mathieu Anheim
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France,INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | - François Cotton
- MRI center, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France,CREATIS-LRMN, CNRS/UMR/5220-INSERM U630, Université Lyon 1, Villeurbanne, France
| | - Hans Rolf Jäger
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Stéphane Kremer
- Service D'imagerie 2, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France,Engineering science, computer science and imaging laboratory (ICube), Integrative Multimodal Imaging in Healthcare, UMR 7357, University of Strasbourg-CNRS, Strasbourg, France
| | - SFNR-COVID group2BonnevilleFabricezAdamGilleszMartin-BlondelGuillaumezParienteJérémiezGeeraertsThomaszOesterléHélèneaaBologniniFedericoaaMessieJulienaaHmeydiaGhaziabBenzakounJosephabOppenheimCatherineabConstansJean-MarcacMetanbouSergeacHeintzAdrienacBapstBlancheadMegdicheImenadJagerLaviniaaeNesserPatrickaeMbaYannick TallaaeTourdiasThomasafCoutureauJulietteafHemmertCélineagFeuersteinPhilippeagSebagNathanagCarreSophieahAllegManelahLecocqClaireahSchmittEmmanuelaiAnxionnatRenéaiZhuFrançoisaiForestierGéraudajRouchaudAymericajCombyPierre-OlivierakRicolfiFredericakThouantPierreakGrandSylviealKrainikAlexandrealde BeaurepaireIsaureamBornetGrégoireamLacalmAudreyanMiailhesPatrickanPiqueJulieanBoutetClaireaoFabreXavieraoClaiseBéatriceapMirafzalSoniaapCalvetLaureapDesalHubertaqBergeJéromeaqBoulouisGrégoireaqKazemiApollineaqPyatigorskayaNadyaaqLeclerAugustinaqSalemeSuzanaaqEdjlali-GoujonMyriamaqKerlerouxBasileaqBrissetJean-ChristopheaqChenafSamirarToulouse University Hospital, Toulouse, FranceLouis Pasteur Hospital, Colmar, FranceSainte-Anne Hospital, Paris, FranceAmiens University Hospital, Amiens, FranceHenri-Mondor University Hospital, Créteil, FranceCHIC Unisanté, Forbach, FranceBordeaux University Hospital, Bordeaux, FranceEmile Muller Hospital, Mulhouse, FranceHospital of Haguenau, Haguenau, FranceNancy University Hospital, Nancy, FranceLimoges University Hospital, Limoges, FranceDijon University Hospital, Dijon, FranceGrenoble Alpes University Hospital, Grenoble, FranceAntony Private Hospital, Antony, FranceHospices Civils de Lyon, Lyon, FranceSaint-Etienne University Hospital, Saint-Etienne, FranceClermont-Ferrand University Hospital, Clermont-Ferrand, FranceFrench Neuroradiological Society (SFNR) Board, FranceUCIEC, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Roquencourt C, Salvator H, Bardin E, Lamy E, Farfour E, Naline E, Devillier P, Grassin-Delyle S. Enhanced real-time mass spectrometry breath analysis for the diagnosis of COVID-19. ERJ Open Res 2023; 9:00206-2023. [PMID: 37727677 PMCID: PMC10505950 DOI: 10.1183/23120541.00206-2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/21/2023] [Indexed: 09/21/2023] Open
Abstract
Background Although rapid screening for and diagnosis of coronavirus disease 2019 (COVID-19) are still urgently needed, most current testing methods are long, costly or poorly specific. The objective of the present study was to determine whether or not artificial-intelligence-enhanced real-time mass spectrometry breath analysis is a reliable, safe, rapid means of screening ambulatory patients for COVID-19. Methods In two prospective, open, interventional studies in a single university hospital, we used real-time, proton transfer reaction time-of-flight mass spectrometry to perform a metabolomic analysis of exhaled breath from adults requiring screening for COVID-19. Artificial intelligence and machine learning techniques were used to build mathematical models based on breath analysis data either alone or combined with patient metadata. Results We obtained breath samples from 173 participants, of whom 67 had proven COVID-19. After using machine learning algorithms to process breath analysis data and further enhancing the model using patient metadata, our method was able to differentiate between COVID-19-positive and -negative participants with a sensitivity of 98%, a specificity of 74%, a negative predictive value of 98%, a positive predictive value of 72% and an area under the receiver operating characteristic curve of 0.961. The predictive performance was similar for asymptomatic, weakly symptomatic and symptomatic participants and was not biased by COVID-19 vaccination status. Conclusions Real-time, noninvasive, artificial-intelligence-enhanced mass spectrometry breath analysis might be a reliable, safe, rapid, cost-effective, high-throughput method for COVID-19 screening.
Collapse
Affiliation(s)
| | - Hélène Salvator
- Exhalomics, Hôpital Foch, Suresnes, France
- Service de Pneumologie, Hôpital Foch, Suresnes, France
- Laboratoire de Recherche en Pharmacologie Respiratoire – VIM Suresnes, UMR 0892, Université Paris-Saclay, Suresnes, France
| | - Emmanuelle Bardin
- Exhalomics, Hôpital Foch, Suresnes, France
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation (2I), U1173, Département de Biotechnologie de la Santé, Montigny le Bretonneux, France
- Institut Necker Enfants Malades, U1151, Paris, France
| | - Elodie Lamy
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation (2I), U1173, Département de Biotechnologie de la Santé, Montigny le Bretonneux, France
| | - Eric Farfour
- Service de Biologie Clinique, Hôpital Foch, Suresnes, France
| | | | - Philippe Devillier
- Exhalomics, Hôpital Foch, Suresnes, France
- Laboratoire de Recherche en Pharmacologie Respiratoire – VIM Suresnes, UMR 0892, Université Paris-Saclay, Suresnes, France
| | - Stanislas Grassin-Delyle
- Exhalomics, Hôpital Foch, Suresnes, France
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation (2I), U1173, Département de Biotechnologie de la Santé, Montigny le Bretonneux, France
| |
Collapse
|
129
|
Mann JK, Reddy T, van der Stok M, Ngubane A, Mulaudzi T, Mchunu N, Nevhungoni P, Manickchund N, Manickchund P, Louise Cairns CH, Govender V, Ndung'u T, Suleman Moosa MY, Gosnell BI. Hen egg white bovine colostrum supplement reduces symptoms of mild/moderate COVID-19: a randomized control trial. Future Sci OA 2023; 9:FSO882. [PMID: 37621850 PMCID: PMC10445555 DOI: 10.2144/fsoa-2023-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/03/2023] [Indexed: 08/26/2023] Open
Abstract
Aim The ability of a hen egg white bovine colostrum supplement to prevent severe COVID-19 was tested in a double-blind randomized control study. Methods Adults with mild/moderate COVID-19, risk factors for severe disease, and within 5 days of symptom onset were assigned to the intervention (n = 77) or placebo (n = 79) arms. Symptoms were documented until day 42 post-enrollment and viral clearance was assessed at 11-13 days post-symptom onset. Results One participant developed severe COVID-19. The severe-type symptom score was lower in the active arm at 11-13 days post-symptom onset (p = 0.049). Chest pain, fever/chills, joint pain/malaise, and sore throat were significantly less frequent in the active arm. No differences in viral clearance were observed. Conclusion The intervention reduced symptoms of mild/moderate COVID-19. Clinical Trial Registration DOH-27-062021-9191 (South African National Clinical Trials Register).
Collapse
Affiliation(s)
- Jaclyn Kelly Mann
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Tarylee Reddy
- Biostatistics Research Unit, South African Medical Research Council, Durban, 4091, South Africa
| | - Mary van der Stok
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Ayanda Ngubane
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Takalani Mulaudzi
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Nobuhle Mchunu
- Biostatistics Research Unit, South African Medical Research Council, Durban, 4091, South Africa
| | - Portia Nevhungoni
- Biostatistics Research Unit, South African Medical Research Council, Durban, 4091, South Africa
| | - Nithendra Manickchund
- Department of Infectious Diseases, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Pariva Manickchund
- Department of Infectious Diseases, University of KwaZulu-Natal, Durban, 4001, South Africa
| | | | | | - Thumbi Ndung'u
- HIV Pathogenesis Programme, University of KwaZulu-Natal, Durban, 4001, South Africa
- Africa Health Research Institute, Durban, 4001, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology & Harvard University, Cambridge, MA 02139, USA
- Division of Infection & Immunity, University College London, London, WC1E 6BT, UK
| | | | | |
Collapse
|
130
|
Mastboim NS, Angel A, Shaham O, Ber TI, Navon R, Simon E, Rosenberg M, Israeli Y, Hainrichson M, Avni N, Reiner E, Feigin P, Oved K, Tadmor B, Singer P, Kagan I, Lev S, Diker D, Jarjou'I A, Kurd R, Ben-Chetrit E, Danziger G, Tegethoff SA, Papan C, Motov S, Shapira M, Stein M, Gottlieb TM, Eden E, Klein A. An immune-protein score combining TRAIL, IP-10 and CRP for predicting severe COVID-19 disease. Cytokine 2023; 169:156246. [PMID: 37327532 PMCID: PMC10235675 DOI: 10.1016/j.cyto.2023.156246] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/11/2023] [Accepted: 05/21/2023] [Indexed: 06/18/2023]
Abstract
COVID-19 patients are oftentimes over- or under-treated due to a deficit in predictive management tools. This study reports derivation of an algorithm that integrates the host levels of TRAIL, IP-10, and CRP into a single numeric score that is an early indicator of severe outcome for COVID-19 patients and can identify patients at-risk to deteriorate. 394 COVID-19 patients were eligible; 29% meeting a severe outcome (intensive care unit admission/non-invasive or invasive ventilation/death). The score's area under the receiver operating characteristic curve (AUC) was 0.86, superior to IL-6 (AUC 0.77; p = 0.033) and CRP (AUC 0.78; p < 0.001). Likelihood of severe outcome increased significantly (p < 0.001) with higher scores. The score differentiated severe patients who further deteriorated from those who improved (p = 0.004) and projected 14-day survival probabilities (p < 0.001). The score accurately predicted COVID-19 patients at-risk for severe outcome, and therefore has potential to facilitate timely care escalation and de-escalation and appropriate resource allocation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Paul Feigin
- Technion-Israel Institute of Technology, Israel
| | - Kfir Oved
- MeMed, Tirat Carmel, Israel; Canopy Immuno-therapeutics, Israel
| | | | | | | | | | | | - Amir Jarjou'I
- Shaare Zedek Medical Center, Hebrew University School of Medicine, Israel
| | - Ramzi Kurd
- Shaare Zedek Medical Center, Hebrew University School of Medicine, Israel
| | - Eli Ben-Chetrit
- Shaare Zedek Medical Center, Hebrew University School of Medicine, Israel
| | | | | | | | | | - Ma'anit Shapira
- Technion-Israel Institute of Technology, Israel; Hillel Yaffe Medical Center, Israel
| | - Michal Stein
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Israel; Sackler School of Medicine,Tel Aviv University, Tel Aviv, Israel
| | | | | | - Adi Klein
- Technion-Israel Institute of Technology, Israel; Hillel Yaffe Medical Center, Israel
| |
Collapse
|
131
|
Soto J, Linsley C, Song Y, Chen B, Fang J, Neyyan J, Davila R, Lee B, Wu B, Li S. Engineering Materials and Devices for the Prevention, Diagnosis, and Treatment of COVID-19 and Infectious Diseases. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2455. [PMID: 37686965 PMCID: PMC10490511 DOI: 10.3390/nano13172455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023]
Abstract
Following the global spread of COVID-19, scientists and engineers have adapted technologies and developed new tools to aid in the fight against COVID-19. This review discusses various approaches to engineering biomaterials, devices, and therapeutics, especially at micro and nano levels, for the prevention, diagnosis, and treatment of infectious diseases, such as COVID-19, serving as a resource for scientists to identify specific tools that can be applicable for infectious-disease-related research, technology development, and treatment. From the design and production of equipment critical to first responders and patients using three-dimensional (3D) printing technology to point-of-care devices for rapid diagnosis, these technologies and tools have been essential to address current global needs for the prevention and detection of diseases. Moreover, advancements in organ-on-a-chip platforms provide a valuable platform to not only study infections and disease development in humans but also allow for the screening of more effective therapeutics. In addition, vaccines, the repurposing of approved drugs, biomaterials, drug delivery, and cell therapy are promising approaches for the prevention and treatment of infectious diseases. Following a comprehensive review of all these topics, we discuss unsolved problems and future directions.
Collapse
Affiliation(s)
- Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Chase Linsley
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Binru Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jun Fang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Josephine Neyyan
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Raul Davila
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brandon Lee
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
132
|
Manzoor F, Redelmeier DA. COVID-19 deaths on weekends. BMC Public Health 2023; 23:1596. [PMID: 37608262 PMCID: PMC10464124 DOI: 10.1186/s12889-023-16451-8] [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: 12/16/2022] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Mortality statistics about daily deaths might change on weekends due to delays in reporting, uneven staffing, a different mix of personnel, or decreased efficiency. We hypothesized that reported deaths for COVID-19 might increase on weekends compared to weekdays. METHODS We collected data from the World Health Organization COVID-19 database. All deaths from March 7, 2020 to March 7, 2022 were included (two years). The primary analysis evaluated mean daily deaths on weekends compared to the preceding five workdays. Analyses were replicated in ten individual countries: United States, United Kingdom, France, Germany, Italy, Spain, Russia, India, Brazil, and Canada. RESULTS The mean COVID-19 daily deaths was higher on weekends compared to weekdays (8,532 vs. 8,083 p < 0.001), equal to a 6% relative increase (95% confidence interval 3% to 8%). The highest absolute increase was in the United States (1,483 vs. 1,220 deaths, p < 0.001). The second highest absolute increase was in Brazil (1,061 vs. 823 deaths, p < 0.001). The increase in deaths on weekends remained significant during the earlier and later months of the pandemic, as well as during the greater and lesser weeks of the pandemic. CONCLUSIONS The apparent increased COVID-19 deaths reported on weekends might potentially reflect patient care, confound community trends, and affect the public perception of risk.
Collapse
Affiliation(s)
- Fizza Manzoor
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Donald A Redelmeier
- Department of Medicine, University of Toronto, Toronto, Canada.
- Institute for Clinical Evaluative Sciences, Toronto, Canada.
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Sunnybrook Hospital, G-151, 2075 Bayview Ave, ON, M4N 3M5, Toronto, Canada.
| |
Collapse
|
133
|
Mar KB, Wells AI, Caballero Van Dyke MC, Lopez AH, Eitson JL, Fan W, Hanners NW, Evers BM, Shelton JM, Schoggins JW. LY6E is a pan-coronavirus restriction factor in the respiratory tract. Nat Microbiol 2023; 8:1587-1599. [PMID: 37443277 PMCID: PMC11234902 DOI: 10.1038/s41564-023-01431-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
LY6E is an antiviral restriction factor that inhibits coronavirus spike-mediated fusion, but the cell types in vivo that require LY6E for protection from respiratory coronavirus infection are unknown. Here we used a panel of seven conditional Ly6e knockout mice to define which Ly6e-expressing cells confer control of airway infection by murine coronavirus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Loss of Ly6e in Lyz2-expressing cells, radioresistant Vav1-expressing cells and non-haematopoietic cells increased susceptibility to murine coronavirus. Global conditional loss of Ly6e expression resulted in clinical disease and higher viral burden after SARS-CoV-2 infection, but little evidence of immunopathology. We show that Ly6e expression protected secretory club and ciliated cells from SARS-CoV-2 infection and prevented virus-induced loss of an epithelial cell transcriptomic signature in the lung. Our study demonstrates that lineage confined rather than broad expression of Ly6e sufficiently confers resistance to disease caused by murine and human coronaviruses.
Collapse
Affiliation(s)
- Katrina B Mar
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexandra I Wells
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Alexandra H Lopez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jennifer L Eitson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wenchun Fan
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Natasha W Hanners
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Departments of Pathology and Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John W Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
134
|
Knopp BW, Weiss HZ, Fahmy S, Goldstein E, Parmar J. A Comparison of the Adverse Effects and Utility of Different Monoclonal Antibodies for SARS-CoV-2: A Retrospective Cohort Study. Cureus 2023; 15:e43094. [PMID: 37680398 PMCID: PMC10482545 DOI: 10.7759/cureus.43094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/06/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Multiple monoclonal antibody (mAb) treatments have been developed to combat the growing number of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strains. These treatments have been shown to be effective in reducing the risk of hospitalization and death from SARS-CoV-2 infection with a low risk of adverse effects; however, more data is required to evaluate the comparative efficacy of mAbs. The primary objective of this study is to describe the hospitalization rate, length of stay (LOS), and mortality rate in SARS-CoV-2 patients treated with four different mAb treatments, including bamlanivimab plus etesevimab, casirivimab plus imdevimab, sotrovimab, and bebtelovimab. Methods A retrospective chart review and prospective phone surveys of SARS-CoV-2 patients treated with mAbs in a 400-bed tertiary, suburban medical center were conducted between June 2020 and April 2022. Eligibility criteria for mAbs included non-hospitalized patients over the age of 18 with less than 10 days of SARS-CoV-2 symptoms and no oxygen requirement on emergency department (ED) admission. Data were collected from the retrospective chart review and subjective patient surveys. A chi-squared test was used. Significance was assessed at p < 0.05. Results The study population included 3249 patients, with 1537 males and 1712 females and an average age of 62.48 ± 17.54 years. Five hundred forty-two patients received bamlanivimab plus etesevimab; 849 received bebtelovimab; 1577 received casirivimab plus imdevimab; and 281 received sotrovimab. The overall hospitalization rate was 1.0%, and the mortality rate was 0.2% following mAb treatment. The hospitalization rate was greatest among patients administered Sotrovimab (2.1%) and least among patients administered Bebtelovimab (0.1%) (p = 0.010). 2.4% of patients who were discharged from the ED after receiving one of the four mAbs returned within 30 days with SARS-CoV-2 symptoms. The average length of stay was 4.75 ± 4.56 days, with no significant differences between the mAbs. The provider-reported adverse event rate was 2.2%, with significant differences in adverse event rates between mAbs. Bamlanivimab-etesevimab was associated with the highest adverse event rate (4.6%), and sotrovimab was associated with the lowest adverse event rate (1.4%) (p < 0.001). Conclusion This study shows a low hospitalization and mortality rate following mAb infusion in patients with mild and moderate COVID-19. However, there were significant differences in hospitalization and mortality among patients receiving each of the four mAb treatments. There was a high degree of patient-reported symptom improvement, and adverse reactions were reported in only 2.2% of patients with no severe reactions. Multiple monoclonal antibody treatments are not effective as monotherapy; however, this study shows the potential benefits of including a mAb infusion as part of a SARS-CoV-2 treatment plan.
Collapse
Affiliation(s)
- Brandon W Knopp
- Department of Emergency Medicine, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, USA
| | - Hannah Z Weiss
- Department of Emergency Medicine, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, USA
| | - Samer Fahmy
- Department of Emergency Medicine, Boca Raton Regional Hospital, Boca Raton, USA
| | - Evan Goldstein
- Department of Emergency Medicine, Boca Raton Regional Hospital, Boca Raton, USA
| | - Jeniel Parmar
- Department of Emergency Medicine, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, USA
| |
Collapse
|
135
|
Lee J, Seo GH, Song K. Beyond Acute COVID-19: Investigating the Incidence of Subacute Thyroiditis in Long COVID-19 in Korea. Endocrinol Metab (Seoul) 2023; 38:455-461. [PMID: 37550861 PMCID: PMC10475963 DOI: 10.3803/enm.2023.1711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGRUOUND The correlation between acute coronavirus disease 2019 (COVID-19) and subacute thyroiditis (SAT) has not been clearly investigated in "long COVID" patients. We aimed to investigate the incidence of SAT during convalescence and after the acute phase of COVID-19, comparing with that of the general population. METHODS Data from a total of 422,779 COVID-19 patients and a control group of 2,113,895 individuals were analyzed. The index date was defined as the date 3 months after confirmation of COVID-19. The incidence rate (IR) of SAT and hazard ratios (HRs) were calculated per 100,000 persons. Subgroup analysis included analysis of HRs 90-179 and 180 days post-COVID-19 diagnosis; and additional analysis was conducted according to hospitalization status, sex, and age group. RESULTS The IR of SAT was 17.28 per 100,000 persons (95% confidence interval [CI], 12.56 to 23.20) in the COVID-19 group and 8.63 (95% CI, 6.37 to 11.45) in the control group. The HR of COVID-19 patients was 1.76 (95% CI, 1.01 to 3.06; P=0.045). The HR of SAT was 1.39 (95% CI, 0.82 to 2.34; P=0.220) up to 6 months after the index date and 2.30 (95% CI, 1.60 to 3.30; P<0.001) beyond 6 months. The HR for SAT among COVID-19 patients was 2.00 (95% CI, 1.41 to 2.83) in hospitalized patients and 1.76 (95% CI, 1.01 to 3.06) in non-hospitalized patients compared to the control group. The IR of SAT was 27.09 (95% CI, 20.04 to 35.82) for females and 6.47 (95% CI, 3.34 to 11.30) for males. In the 19 to 64 age group, the IR of SAT was 18.19 (95% CI, 13.70 to 23.67), while the IR was 9.18 (95% CI, 7.72 to 10.84) in the 65 to 69 age group. CONCLUSION SAT could be a potential long-term complication of COVID-19. Long-term surveillance for thyroid dysfunction is needed especially in hospitalized, female and young-aged subjects.
Collapse
Affiliation(s)
- Jeongmin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi Hyeon Seo
- Health Insurance Review and Assessment Service, Wonju, Korea
| | - Keeho Song
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Konkuk University Hospital, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
136
|
Samiei N, Rahnamoun Z, Kamali M, Asadian S, Rezaei Y, ghadrdoost B, Shirkhanloo N. Paradoxical increase in left atrial strains early after Covid-19 infection, a result of comprehensive recovery phase four-chamber strains study. Int J Cardiovasc Imaging 2023; 39:1437-1447. [PMID: 37162708 PMCID: PMC10171154 DOI: 10.1007/s10554-023-02865-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023]
Abstract
Cardiac consequences of Covid-19 infection have been mentioned in various studies as a serious risk factor for in-hospital mortality. However, the existence of residual cardiac dysfunction after the acute phase is seldom investigated especially in people without a history of specific medical disease. One hundred health care workers with positive reverse transcription-polymerase chain reaction test underwent comprehensive 2D and 3D echocardiography six to eight weeks after infection. Patients were classified into Mild, Moderate, and Severe groups based on their clinical characteristics of covid-19 infection, and all echocardiographic parameters were compared between the three groups. Left ventricular (LV) stroke volume index was reduced in all groups compared to normal ranges and was more prominent in the severe group (P-value < 0.05). 3D-derived LV global longitudinal strain (GLS) was significantly lower in the severe group in comparison to the mild group (- 19.3 ± 1 Vs. - 22.2 ± 2, P-value < 0.001) and correlated with highly sensitive CRP level at the acute phase. Left atrial (LA) strains, including LA peak strain, LA contraction strain, and LA reservoir strain, were considerably higher and LA volume index was significantly lower in the clinically severe covid patients. Analysis based on the extent of lung involvement showed significantly increased 3D-derived right ventricular volumes in patients who experienced severe pneumonia despite normalized strains. Conclusion: subclinical LV dysfunction as reduced stroke volume index and GLS exists in the early recovery phase of normal individuals with severe course of covid-19. LA function indicated by LA strains paradoxically increases in severe covid-19 infection in this phase.
Collapse
Affiliation(s)
- Niloufar Samiei
- Echocardiography Department, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Rahnamoun
- Tehran Heart Center, Tehran University of Medical Science, Tehran, Iran
| | - Monireh Kamali
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sanaz Asadian
- Radiology Department, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Yousef Rezaei
- Heart Valve Disease Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Behshid ghadrdoost
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Shirkhanloo
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
137
|
Silaghi-Dumitrescu R, Patrascu I, Lehene M, Bercea I. Comorbidities of COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1393. [PMID: 37629683 PMCID: PMC10456773 DOI: 10.3390/medicina59081393] [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: 07/11/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023]
Abstract
The novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) responsible for the coronavirus disease outbreak initiated in 2019 (COVID-19) has been shown to affect the health of infected patients in a manner at times dependent on pre-existing comorbidities. Reported here is an overview of the correlation between comorbidities and the exacerbation of the disease in patients with COVID-19, which may lead to poor clinical outcomes or mortality. General medical issues are also reviewed, such as the types of symptoms present in people infected with SARS-CoV-2, the long-term effects of COVID-19 disease, and the types of treatment that are currently used.
Collapse
Affiliation(s)
- Radu Silaghi-Dumitrescu
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Str., 400028 Cluj-Napoca, Romania (M.L.)
| | - Iulia Patrascu
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Str., 400028 Cluj-Napoca, Romania (M.L.)
- Bistrita County Emergency Clinical Hospital, 42 General Grigore Bălan, Bld., 420094 Bistrita, Romania
| | - Maria Lehene
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Str., 400028 Cluj-Napoca, Romania (M.L.)
| | - Iulia Bercea
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Str., 400028 Cluj-Napoca, Romania (M.L.)
| |
Collapse
|
138
|
Toro DM, da Silva-Neto PV, de Carvalho JCS, Fuzo CA, Pérez MM, Pimentel VE, Fraga-Silva TFC, Oliveira CNS, Caruso GR, Vilela AFL, Nobre-Azevedo P, Defelippo-Felippe TV, Argolo JGM, Degiovani AM, Ostini FM, Feitosa MR, Parra RS, Vilar FC, Gaspar GG, da Rocha JJR, Feres O, Costa GP, Maruyama SRC, Russo EMS, Fernandes APM, Santos IKFM, Malheiro A, Sadikot RT, Bonato VLD, Cardoso CRB, Dias-Baruffi M, Trapé ÁA, Faccioli LH, Sorgi CA, ImmunoCovid Consortium Group. Plasma Sphingomyelin Disturbances: Unveiling Its Dual Role as a Crucial Immunopathological Factor and a Severity Prognostic Biomarker in COVID-19. Cells 2023; 12:1938. [PMID: 37566018 PMCID: PMC10417089 DOI: 10.3390/cells12151938] [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: 06/03/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
SARS-CoV-2 infection triggers distinct patterns of disease development characterized by significant alterations in host regulatory responses. Severe cases exhibit profound lung inflammation and systemic repercussions. Remarkably, critically ill patients display a "lipid storm", influencing the inflammatory process and tissue damage. Sphingolipids (SLs) play pivotal roles in various cellular and tissue processes, including inflammation, metabolic disorders, and cancer. In this study, we employed high-resolution mass spectrometry to investigate SL metabolism in plasma samples obtained from control subjects (n = 55), COVID-19 patients (n = 204), and convalescent individuals (n = 77). These data were correlated with inflammatory parameters associated with the clinical severity of COVID-19. Additionally, we utilized RNAseq analysis to examine the gene expression of enzymes involved in the SL pathway. Our analysis revealed the presence of thirty-eight SL species from seven families in the plasma of study participants. The most profound alterations in the SL species profile were observed in patients with severe disease. Notably, a predominant sphingomyelin (SM d18:1) species emerged as a potential biomarker for COVID-19 severity, showing decreased levels in the plasma of convalescent individuals. Elevated SM levels were positively correlated with age, hospitalization duration, clinical score, and neutrophil count, as well as the production of IL-6 and IL-8. Intriguingly, we identified a putative protective effect against disease severity mediated by SM (d18:1/24:0), while ceramide (Cer) species (d18:1/24:1) and (d18:1/24:0)were associated with increased risk. Moreover, we observed the enhanced expression of key enzymes involved in the SL pathway in blood cells from severe COVID-19 patients, suggesting a primary flow towards Cer generation in tandem with SM synthesis. These findings underscore the potential of SM as a prognostic biomarker for COVID-19 and highlight promising pharmacological targets. By targeting sphingolipid pathways, novel therapeutic strategies may emerge to mitigate the severity of COVID-19 and improve patient outcomes.
Collapse
Affiliation(s)
- Diana Mota Toro
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Pedro V. da Silva-Neto
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Jonatan C. S. de Carvalho
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Carlos A. Fuzo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Malena M. Pérez
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Vinícius E. Pimentel
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thais F. C. Fraga-Silva
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Camilla N. S. Oliveira
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Glaucia R. Caruso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Adriana F. L. Vilela
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Pedro Nobre-Azevedo
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thiago V. Defelippo-Felippe
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Jamille G. M. Argolo
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Augusto M. Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Fátima M. Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Marley R. Feitosa
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Rogerio S. Parra
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Fernando C. Vilar
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - Gilberto G. Gaspar
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - José J. R. da Rocha
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
| | - Omar Feres
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Gabriel P. Costa
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Sandra R. C. Maruyama
- Department of Genetics and Evolution, Center for Biological and Health Sciences, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil;
| | - Elisa M. S. Russo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Ana Paula M. Fernandes
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Isabel K. F. M. Santos
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Adriana Malheiro
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Ruxana T. Sadikot
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Vânia L. D. Bonato
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Cristina R. B. Cardoso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Marcelo Dias-Baruffi
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Átila A. Trapé
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Lúcia H. Faccioli
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Carlos A. Sorgi
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | | |
Collapse
|
139
|
Sharifi-Kia A, Nahvijou A, Sheikhtaheri A. Machine learning-based mortality prediction models for smoker COVID-19 patients. BMC Med Inform Decis Mak 2023; 23:129. [PMID: 37479990 PMCID: PMC10360290 DOI: 10.1186/s12911-023-02237-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND The large number of SARS-Cov-2 cases during the COVID-19 global pandemic has burdened healthcare systems and created a shortage of resources and services. In recent years, mortality prediction models have shown a potential in alleviating this issue; however, these models are susceptible to biases in specific subpopulations with different risks of mortality, such as patients with prior history of smoking. The current study aims to develop a machine learning-based mortality prediction model for COVID-19 patients that have a history of smoking in the Iranian population. METHODS A retrospective study was conducted across six medical centers between 18 and 2020 and 15 March 2022, comprised of 678 CT scans and laboratory-confirmed COVID-19 patients that had a history of smoking. Multiple machine learning models were developed using 10-fold cross-validation. The target variable was in-hospital mortality and input features included patient demographics, levels of care, vital signs, medications, and comorbidities. Two sets of models were developed for at-admission and post-admission predictions. Subsequently, the top five prediction models were selected from at-admission models and post-admission models and their probabilities were calibrated. RESULTS The in-hospital mortality rate for smoker COVID-19 patients was 20.1%. For "at admission" models, the best-calibrated model was XGBoost which yielded an accuracy of 87.5% and F1 score of 86.2%. For the "post-admission" models, XGBoost also outperformed the rest with an accuracy of 90.5% and F1 score of 89.9%. Active smoking was among the most important features in patients' mortality prediction. CONCLUSION Our machine learning-based mortality prediction models have the potential to be adapted for improving the management of smoker COVID-19 patients and predicting patients' chance of survival.
Collapse
Affiliation(s)
- Ali Sharifi-Kia
- Department of Health Information Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Azin Nahvijou
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Sheikhtaheri
- Department of Health Information Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
140
|
Woodruff MC, Bonham KS, Anam FA, Walker TA, Faliti CE, Ishii Y, Kaminski CY, Ruunstrom MC, Cooper KR, Truong AD, Dixit AN, Han JE, Ramonell RP, Haddad NS, Rudolph ME, Yalavarthi S, Betin V, Natoli T, Navaz S, Jenks SA, Zuo Y, Knight JS, Khosroshahi A, Lee FEH, Sanz I. Chronic inflammation, neutrophil activity, and autoreactivity splits long COVID. Nat Commun 2023; 14:4201. [PMID: 37452024 PMCID: PMC10349085 DOI: 10.1038/s41467-023-40012-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
While immunologic correlates of COVID-19 have been widely reported, their associations with post-acute sequelae of COVID-19 (PASC) remain less clear. Due to the wide array of PASC presentations, understanding if specific disease features associate with discrete immune processes and therapeutic opportunities is important. Here we profile patients in the recovery phase of COVID-19 via proteomics screening and machine learning to find signatures of ongoing antiviral B cell development, immune-mediated fibrosis, and markers of cell death in PASC patients but not in controls with uncomplicated recovery. Plasma and immune cell profiling further allow the stratification of PASC into inflammatory and non-inflammatory types. Inflammatory PASC, identifiable through a refined set of 12 blood markers, displays evidence of ongoing neutrophil activity, B cell memory alterations, and building autoreactivity more than a year post COVID-19. Our work thus helps refine PASC categorization to aid in both therapeutic targeting and epidemiological investigation of PASC.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Kevin S Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Fabliha A Anam
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Tiffany A Walker
- Department of Medicine, Division of General Internal Medicine, Emory University, Atlanta, GA, USA
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Yusho Ishii
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | | | - Martin C Ruunstrom
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Kelly Rose Cooper
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Alexander D Truong
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Adviteeya N Dixit
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Jenny E Han
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Richard P Ramonell
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | - Sherwin Navaz
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Scott A Jenks
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Yu Zuo
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Arezou Khosroshahi
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| |
Collapse
|
141
|
Sixt T, Moretto F, Esteve C, Duong M, Buisson M, Mahy S, Blot M, Piroth L. Healing Treatments in COVID-19 Patients: A Narrative Review. J Clin Med 2023; 12:4672. [PMID: 37510786 PMCID: PMC10380607 DOI: 10.3390/jcm12144672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Since December 2019, many drugs have been evaluated or advocated as potential treatments of SARS-CoV-2 induced disease (COVID-19), including many repositioned drugs and some others specifically developed for these diseases. They can be roughly classified into three categories according to their main mechanism of action (passive immunization, direct antivirals, and anti-inflammatory treatments), and their use depends on the stage of the disease. Despite often promising preclinical data, most of the treatments evaluated failed to show a significant clinical benefit. In addition, a few others have seen their effectiveness affected by the occurrence of SARS-CoV-2 variants and sub-variants. Herein, the aim of this article is to take stock of the data available as of the 14th of July 2022, concerning the specific healing options evaluated for patients suffering from COVID-19. We focus particularly on healing treatments of COVID-19 and do not deal with preventive treatments such as vaccine. Associated therapies such as venous thromboembolism prophylaxis are not detailed since they are covered in a specific chapter of this issue. Passive immunization, especially through monoclonal antibodies, showed a positive impact on the clinical evolution, whether in outpatients or inpatients without oxygen supply. However, their effectiveness strongly depends on the type of SARS-CoV-2 variant, and often decreases or even vanishes with the most recent variants. Among direct antiviral treatments, ritonavir-boosted nirmatrelvir appears to currently be the cornerstone in the management of early infections, but its use may be limited by drug interactions. Remdesivir remains as an alternative in this situation, even though it is potentially less convenient. Anti-inflammatory treatments have often been shown to be the most effective in inpatients with oxygen supply. Dexamethasone is now a cornerstone of management of these patients. Added tocilizumab seems beneficial in the case of hyper inflammation. JAK inhibitors and anakinra have also gained an interest in some studies. As a conclusion of this narrative review, the best treatment strategy has yet to be defined and is likely to evolve in the future, not only because many other drugs are still under development and evaluation, but also because of the viral epidemics and epidemiology evolution.
Collapse
Affiliation(s)
- Thibault Sixt
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Florian Moretto
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Clementine Esteve
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Michel Duong
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Marielle Buisson
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Sophie Mahy
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Mathieu Blot
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
- CHU Dijon-Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Épidémiologie Clinique, 21000 Dijon, France
- Lipness Team, INSERM Research Centre LNC-UMR1231 and LabEx LipSTIC, University of Burgundy, 21078 Dijon, France
| | - Lionel Piroth
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
- CHU Dijon-Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Épidémiologie Clinique, 21000 Dijon, France
| |
Collapse
|
142
|
Tian L, He M, Fan H, Zhang H, Dong X, Qiao M, Tang C, Yu Y, Chen T, Zhou N. COVID-19 of differing severity: from bulk to single-cell expression data analysis. Cell Cycle 2023; 22:1777-1797. [PMID: 37486005 PMCID: PMC10446813 DOI: 10.1080/15384101.2023.2239620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/03/2023] [Accepted: 06/24/2023] [Indexed: 07/25/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is raging worldwide and causes an immense disease burden. Despite this, the biomarkers and targeting drugs of COVID-19 of differing severity remain largely unknown. Based on the GSE164805 dataset, we identified modules most critical for mild COVID-19 (mCOVID-19) and severe COVID-19 (sCOVID-19) through WGCNA, respectively. We subsequently constructed a protein-protein interaction network, and detected 16 hub genes for mCOVID-19 and 10 hub genes for sCOVID-19, followed by the prediction of upstream transcription factors (TFs) and kinases. The enrichment analysis then showed downregulation of TNFA signaling via NFKB for mCOVID-19, as well as downregulation of MYC targets V1 for sCOVID-19. Infiltration degrees of many immune cells, such as macrophages, were also sharply different between mCOVID-19 and sCOVID-19 samples. Predicted protein targeting drugs with the highest scores nearly all belong to naturally derived or synthetic glucocorticoids. For the two single-cell RNA-seq datasets, we explored the expression distribution of hub genes for mCOVID-19/sCOVID-19 in each cell type. The expression levels of PRKCA, MCM5, TYMS, RBBP4, BCL6, FLOT1, KDM6B, and TLR2 were found to be cell-type-specific. Furthermore, the expression levels of 10 hub genes for mCOVID-19 were significantly upregulated in PBMCs between eight healthy controls and eight mCOVID-19 patients at our institution. Collectively, we detected critical modules, pathways, TFs, kinases, immune cells, targeting drugs, hub genes, and their expression distributions in different cell types that may involve the pathogenesis of COVID-19 of differing severity, which may propel earlier diagnosis and more effective treatment of this intractable disease in the future.
Collapse
Affiliation(s)
- Linlin Tian
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Min He
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Huafeng Fan
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Hongying Zhang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Xiaoxiao Dong
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Mengkai Qiao
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Chenyu Tang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Yan Yu
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Tong Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Nan Zhou
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
143
|
Cantarella G, Aldè M, Consonni D, Zuccotti G, Berardino FD, Barozzi S, Bertoli S, Battezzati A, Zanetti D, Pignataro L. Prevalence of Dysphonia in Non hospitalized Patients with COVID-19 in Lombardy, the Italian Epicenter of the Pandemic. J Voice 2023; 37:605-609. [PMID: 33766419 PMCID: PMC7955941 DOI: 10.1016/j.jvoice.2021.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Dysphonia has been described in patients affected by coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the prevalence of dysphonia, and its severity and extent, of voice fatigue and of dysphagia in non hospitalized patients affected by COVID-19 in Lombardy, the Italian region most hit by the first explosive outbreak of COVID-19 in Europe. METHODS Demographic and clinical data of 160 consecutive patients, with COVID-19 diagnosis confirmed by nasal swabs processed by reverse transcription polymerase chain reaction, were gathered by means of telephone interviews performed by physicians in charge of daily follow-up. General and specific symptoms concerning voice and swallowing impairment were investigated. Dysphonia grade and duration were graded on 4-point scales, while voice fatigue was graded on a 5-point scale. RESULTS Dysphonia was reported by 70 (43.7%) patients and was positively associated with voice fatigue (P < 0.001), cough (P = 0.005), rhinitis (P = 0.01), and dyspnea (P = 0.06); it was mild and/or moderate in 69 patients, but its duration was > 2 weeks in 33/70 (47.1%) patients and >1 month in 11/70 (15.7%). Grade and duration of dysphonia were positively associated with cough and rhinitis (all P values < 0.01). Voice fatigue was reported by 43/160 patients (26.8%) and its severity was correlated with dysphonia (P < 0.0001), cough (P = 0.02), rhinitis (P = 0.02), dyspnea (P < 0.001), and loss of appetite (P = 0.01). Dysphagia was encountered in 27/160 patients (16.9%) and was associated with dysgeusia, cough, arthralgia, myalgia and loss of appetite but not with dysphonia. CONCLUSIONS Dysphonia was a highly prevalent and long-lasting symptom in this series; it has been underestimated to date. Further studies might shed light on the pathophysiology of voice disorders in COVID-19 patients.
Collapse
Affiliation(s)
- Giovanna Cantarella
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Otolaryngology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Mirko Aldè
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Audiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Consonni
- Epidemiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Children's Hospital "Vittore Buzzi", Azienda Socio Sanitaria Territoriale (ASST) Fatebenefratelli, Milan, Italy; "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Federica Di Berardino
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Audiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Barozzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Audiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simona Bertoli
- International Center for the Assessment of Nutritional Status (ICANS), Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy; Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS (Scientific Institute for Research, Hospitalization, and Healthcare) Italian Auxologic Institute (IAI), Milan, Italy
| | - Alberto Battezzati
- International Center for the Assessment of Nutritional Status (ICANS), Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy; Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS (Scientific Institute for Research, Hospitalization, and Healthcare) Italian Auxologic Institute (IAI), Milan, Italy
| | - Diego Zanetti
- Audiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Pignataro
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Otolaryngology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
144
|
Oliveira DCD, Spiri BS, Schluga YC, Justus JLP, Lopes Neto FDN, Azambuja APD. Evaluation of lymphocyte count, T-cell subsets and neutrophil-to-lymphocyte ratio as early predictors for severity and outcome of COVID-19 disease-a report from a highly complex hospital in Brazil. Hematol Transfus Cell Ther 2023; 45:330-337. [PMID: 35782909 PMCID: PMC9239916 DOI: 10.1016/j.htct.2022.05.007] [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: 11/30/2021] [Revised: 02/19/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Lymphopenia is a laboratory marker of poor prognosis and severity of disease in the SARS-CoV-2 infection. This study aims to describe the immune profile of a Brazilian population. METHODS A total of 121 consecutive patients with severe acute respiratory syndrome (SARS) were analyzed between April and June 2020. Routine peripheral blood counts and multiparametric flow cytometry were performed on admission to assess lymphocytes and subsets (CD3, CD4, CD8). Demographic, clinical and laboratory data were collected from hospital sources. RESULTS The total of 116 patients included 63 (54.3%) males; 76 (62.8%) COVID-19 patients were divided, based on clinical characteristics and mechanical ventilation (MV) use, into moderate (n = 41; no MV) and severe (n = 35; MV) groups. The control group (n = 40) was comprised of patients with SARS of different etiologies. All patients had lymphopenia, with overall lymphocyte counts and their subsets considerably lower in severe patients, when compared to the moderate and controls. Patients with a high neutrophil-to-lymphocyte ratio (> 15.2) and T-cell lymphopenia (CD3 < 593 cells/µL, CD4 < 326 cells/µL, CD8 < 121 cells/µL) had a higher risk of being intubated and progressing to death. A total of 39 patients (95.1%) in the moderate group and 54.3% (n = 19) in the severe group were discharged; 28 patients died. CONCLUSION Laboratory assessment of the neutrophil/lymphocyte ratio and T-cell subsets may be predictive of mortality and may be useful for stratifying COVID-19 patients.
Collapse
Affiliation(s)
| | - Beatriz Sanada Spiri
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| | - Yara Carolina Schluga
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| | | | | | - Ana Paula de Azambuja
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| |
Collapse
|
145
|
Müller S, Schultze JL. Systems analysis of human innate immunity in COVID-19. Semin Immunol 2023; 68:101778. [PMID: 37267758 PMCID: PMC10201327 DOI: 10.1016/j.smim.2023.101778] [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: 02/02/2023] [Revised: 05/13/2023] [Accepted: 05/13/2023] [Indexed: 06/04/2023]
Abstract
Recent developments in sequencing technologies, the computer and data sciences, as well as increasingly high-throughput immunological measurements have made it possible to derive holistic views on pathophysiological processes of disease and treatment effects directly in humans. We and others have illustrated that incredibly predictive data for immune cell function can be generated by single cell multi-omics (SCMO) technologies and that these technologies are perfectly suited to dissect pathophysiological processes in a new disease such as COVID-19, triggered by SARS-CoV-2 infection. Systems level interrogation not only revealed the different disease endotypes, highlighted the differential dynamics in context of disease severity, and pointed towards global immune deviation across the different arms of the immune system, but was already instrumental to better define long COVID phenotypes, suggest promising biomarkers for disease and therapy outcome predictions and explains treatment responses for the widely used corticosteroids. As we identified SCMO to be the most informative technologies in the vest to better understand COVID-19, we propose to routinely include such single cell level analysis in all future clinical trials and cohorts addressing diseases with an immunological component.
Collapse
Affiliation(s)
- Sophie Müller
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany; Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany.
| |
Collapse
|
146
|
Fuzo CA, Fraga-Silva TFC, Maruyama SR, Bastos VAF, Rogerio LA, Takamiya NT, da Silva-Neto PV, Pimentel VE, Toro DM, Pérez MM, de Carvalho JCS, Carmona-Garcia I, Oliveira CNS, Degiovani AM, Ostini FM, Constant LF, de Amorim AP, Vilar FC, Feitosa MR, Parra RS, da Rocha JJR, Feres O, Gaspar GG, Viana AL, Fernandes APM, Santos IKFM, Russo EMS, Cardoso CRB, Sorgi CA, Faccioli LH, Bonato VLD, Dias-Baruffi M. The turning point of COVID-19 severity is associated with a unique circulating neutrophil gene signature. Immunology 2023; 169:323-343. [PMID: 36740582 DOI: 10.1111/imm.13631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/01/2023] [Indexed: 02/07/2023] Open
Abstract
COVID-19 has a broad spectrum of clinical manifestations associated with the host immune response heterogeneity. Despite the advances in COVID-19 research, it is still crucial to seek a panel of molecular markers that enable accurate stratification of COVID-19 patients. Here, we performed a study that combined analysis of blood transcriptome, demographic data, clinical aspects and laboratory findings from 66 participants classified into different degrees of COVID-19 severity and healthy subjects. We identified a perturbation in blood-leukocyte transcriptional profile associated with COVID-19 aggravation, which was mainly related to processes that disfavoured lymphocyte activation and favoured neutrophil activation. This transcriptional profile stratified patients according to COVID-19 severity. Hence, it enabled identification of a turning point in transcriptional dynamics that distinguished disease outcomes and non-hospitalized from hospitalized moderate patients. Central genes of this unique neutrophil signature were S100A9, ANXA3, CEACAM6, VNN1, OLFM4, IL1R2, TCN1 and CD177. Our study indicates the molecular changes that are linked with the differing clinical aspects presented by humans when suffering from COVID-19, which involve neutrophil activation.
Collapse
Affiliation(s)
- Carlos A Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thais F C Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sandra R Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Víctor A F Bastos
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luana A Rogerio
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Nayore T Takamiya
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Vinícius E Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Malena M Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ingryd Carmona-Garcia
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Camilla N S Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Augusto M Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fátima M Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Leticia F Constant
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Alessandro P de Amorim
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fernando C Vilar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marley R Feitosa
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rogerio S Parra
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José J R da Rocha
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Omar Feres
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gilberto G Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Angelina L Viana
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana P M Fernandes
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Isabel K F M Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elisa M S Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cristina R B Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos A Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vânia L D Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
147
|
Ahmad AM, Nawar NM, Dabess HM, Gallab MA. Effect of diaphragm manual release versus conventional breathing exercises and prone positioning on physical functional performance in women with COVID-19: A randomized trial. J Bodyw Mov Ther 2023; 35:311-319. [PMID: 37330786 PMCID: PMC10121151 DOI: 10.1016/j.jbmt.2023.04.064] [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: 10/08/2022] [Revised: 02/22/2023] [Accepted: 04/12/2023] [Indexed: 06/19/2023]
Abstract
INTRODUCTION Manual therapy has recently gained much interest in managing COVID-19 patients. This study aimed to mainly compare the effect of diaphragm manual release to the effect of conventional breathing exercises and prone positioning on physical functional performance in women with COVID-19. METHODS Forty COVID-19 women patients completed this study. They were randomly assigned to two groups. Group A received diaphragm manual release, and group B received conventional breathing exercises and prone positioning. Both groups received pharmacological treatment. Inclusion criteria were moderate COVID-19 illness, women patients, and ages from 35 to 45 years. The outcome measures were 6-min walk distance (6MWD), chest expansion, Barthel index (BI), oxygen saturation, fatigue Assessment Scale (FAS), and Medical Research Council (MRC) dyspnea scale. RESULTS Both groups showed significant improvements in all outcome measures compared to the baseline (p < 0.001). Compared to group B, group A showed more significant improvements in the 6MWD (MD, 22.75 m; 95% CI, 15.21 to 30.29; p < 0.001), chest expansion (MD, 0.80 cm; 95% CI, 0.46 to 1.14; p < 0.001), BI (MD, 9.50; 95% CI, 5.69 to 13.31; p < 0.001), the O2 saturation (MD, 1.3%; 95% CI, 0.71 to 1.89; p < 0.001), the FAS (MD, -4.70; 95% CI, -6.69 to -2.71; p < 0.001), and dyspnea severity assessed by the MRC dyspnea scale (p = 0.013) post-intervention. CONCLUSION Combined with pharmacological treatment, diaphragm manual release could be superior to conventional breathing exercises and prone positioning in improving physical functional performance, chest expansion, daily living activities, O2 saturation, and measures of fatigue and dyspnea in middle-aged women with moderate COVID-19 illness. TRIAL REGISTRATION Pan African Clinical Trial Registry (PACTR), retrospective, PACTR202302877569441.
Collapse
Affiliation(s)
- Ahmad Mahdi Ahmad
- Department of Physical Therapy for Cardiovascular and Respiratory Disorders, Faculty of Physical Therapy, Cairo University, Giza, Egypt.
| | | | | | - Mona Abulraouf Gallab
- Department of Physical Therapy for Cardiovascular and Respiratory Disorders, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| |
Collapse
|
148
|
Jaulmes L, Yordanov Y, Descamps A, Durand-Zaleski I, Dinh A, Jourdain P, Dechartres A. Effectiveness and Medicoeconomic Evaluation of Home Monitoring of Patients With Mild COVID-19: Covidom Cohort Study. J Med Internet Res 2023; 25:e43980. [PMID: 37134021 PMCID: PMC10337320 DOI: 10.2196/43980] [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: 11/01/2022] [Revised: 03/03/2023] [Accepted: 03/20/2023] [Indexed: 03/22/2023] Open
Abstract
BACKGROUND Covidom was a telemonitoring solution for home monitoring of patients with mild to moderate COVID-19, deployed in March 2020 in the Greater Paris area in France to alleviate the burden on the health care system. The Covidom solution included a free mobile application with daily monitoring questionnaires and a regional control center to quickly handle patient alerts, including dispatching emergency medical services when necessary. OBJECTIVE This study aimed to provide an overall evaluation of the Covidom solution 18 months after its inception in terms of effectiveness, safety, and cost. METHODS Our primary outcome was to measure effectiveness using the number of handled alerts, response escalation, and patient-reported medical contacts outside of Covidom. Then, we analyzed the safety of Covidom by assessing its ability to detect clinical worsening, defined as hospitalization or death, and the number of patients with clinical worsening without any preceding alert. We evaluated the cost of Covidom and compared the cost of hospitalization for Covidom and non-Covidom patients with mild COVID-19 cases seen in the emergency departments of the largest network of hospitals in the Greater Paris area (Assistance Publique-Hôpitaux de Paris). Finally, we reported on user satisfaction. RESULTS Of the 60,073 patients monitored by Covidom, the regional control center handled 285,496 alerts and dispatched emergency medical services 518 times. Of the 13,204 respondents who responded to either of the follow-up questionnaires, 65.8% (n=8690) reported having sought medical care outside the Covidom solution during their monitoring period. Of the 947 patients who experienced clinical worsening while adhering to daily monitoring, only 35 (3.7%) did not previously trigger alerts (35 were hospitalized, including 1 who died). The average cost of Covidom was €54 (US $1=€0.8614) per patient, and the cost of hospitalization for COVID-19 worsening was significantly lower in Covidom than in non-Covidom patients with mild COVID-19 cases seen in the emergency departments of Assistance Publique-Hôpitaux de Paris. The patients who responded to the satisfaction questionnaire had a median rating of 9 (out of 10) for the likelihood of recommending Covidom. CONCLUSIONS Covidom may have contributed to alleviating the pressure on the health care system in the initial months of the pandemic, although its impact was lower than anticipated, with a substantial number of patients having consulted outside of Covidom. Covidom seems to be safe for home monitoring of patients with mild to moderate COVID-19.
Collapse
Affiliation(s)
- Luc Jaulmes
- Centre de pharmaco-épidémiologie de l'APHP, Dépt. de Santé Publique, Hôpital Pitié Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Youri Yordanov
- Sorbonne Université, AP-HP, Hôpital Saint Antoine, Service d'Accueil des Urgences, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, UMR-S 1136, Paris, France
| | - Alexandre Descamps
- CIC Cochin Pasteur, INSERM CIC 1417, Université Paris Cité, AP-HP, Paris, France
| | - Isabelle Durand-Zaleski
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Université Paris Est, AP-HP, Paris, France
- URC Eco, Hôpital de l'Hôtel Dieu, DRCI, AP-HP, Paris, France
| | - Aurélien Dinh
- Infectious Disease department, University Hospital R. Poincaré, UVSQ, AP-HP, Garches, France
| | - Patrick Jourdain
- INSERM U999, CHU Bicêtre AP-HP, Université Paris-Saclay, AP-HP, Gif-sur-Yvette, France
| | - Agnès Dechartres
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, AP-HP. Sorbonne Université, Hôpital Pitié Salpêtrière, Département de Santé Publique, centre de pharmaco-épidémiologie de l'APHP, F75013, Paris, France
| |
Collapse
|
149
|
Augustyniak A, Szymański T, Porzucek F, Mieloch AA, Semba JA, Hubert KA, Grajek D, Krela R, Rogalska Z, Zalc-Budziszewska E, Wysocki S, Sobczak K, Kuczyński L, Rybka JD. A cohort study reveals different dynamics of SARS-CoV-2-specific antibody formation after Comirnaty and Vaxzevria vaccination. Vaccine 2023:S0264-410X(23)00665-5. [PMID: 37407407 PMCID: PMC10284451 DOI: 10.1016/j.vaccine.2023.06.008] [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: 02/16/2023] [Revised: 05/23/2023] [Accepted: 06/02/2023] [Indexed: 07/07/2023]
Abstract
The Coronavirus (COVID-19) Disease Pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has affected millions of people worldwide, prompting a collective effort from the global scientific community to develop a vaccine against it. This study purports to investigate the influence of factors such as sex, age, type of vaccination (Comirnaty, BNT162b2, Pfizer Inc. or Vaxzevria, ChAdOx1-S, Oxford/AstraZeneca), and time since vaccine administration on the process of antibody production. Both of them are based on the introduction of SARS-CoV-2 spike protein (S protein) to the body using different mechanisms (mRNA and recombinant adenovirus, respectively). S protein is responsible for host cell attachment and penetration via its receptor-binding domain (RBD domain). The level of anti-RBD IgG antibodies was tested with an ELISA-based immunodiagnostic assay in serum samples from a total of 1395 patients at 3 time points: before vaccination, after the first dose, and after the second dose. Our novel statistical model, the Generalized Additive Model, revealed variability in antibody production dynamics for both vaccines. Interestingly, no discernible variation in antibody levels between men and women was found. A nonlinear relationship between age and antibody production was observed, characterized by decreased antibody levels for people up to 30 and over 60 years of age, with a lack of correlation in the middle age range. Collectively, our findings further the understanding of the mechanism driving vaccine-induced immunity. Additionally, we propose the Generalized Additive Model as a standardized way of presenting data in similar research.
Collapse
Affiliation(s)
- Adam Augustyniak
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Tomasz Szymański
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland; Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland; Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland
| | - Filip Porzucek
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Adam Aron Mieloch
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Julia Anna Semba
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland; Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland
| | - Katarzyna Anna Hubert
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland; Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland
| | - Dominika Grajek
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Rafał Krela
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Zuzanna Rogalska
- Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland; Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznan, Poland
| | - Ewa Zalc-Budziszewska
- Provincial Specialist Complex of Healthcare Institutions of Lung Diseases and Tuberculosis, Wolica 113, 62-872 Godziesze Małe, Poland
| | - Sławomir Wysocki
- Provincial Specialist Complex of Healthcare Institutions of Lung Diseases and Tuberculosis, Wolica 113, 62-872 Godziesze Małe, Poland
| | - Krzysztof Sobczak
- Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland; Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznan, Poland
| | - Lechosław Kuczyński
- Population Ecology Lab, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznan, Poland
| | - Jakub Dalibor Rybka
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland.
| |
Collapse
|
150
|
Silva Borborema T, Moreira Brito JC, Lima Batista EM, Siqueira Batista R. Case Fatality Rate and Severity of COVID-19 among Patients with Sickle Cell Disease: A Systematic Review and Meta-Analysis. Hemoglobin 2023:1-12. [PMID: 37325879 DOI: 10.1080/03630269.2023.2219847] [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: 09/09/2022] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/17/2023]
Abstract
The sickle cell disease (SCD) population has been considered particularly vulnerable to viral pandemics since the emergence of H1N1 in 2009. In this sense, the advance of the COVID-19 pandemic from 2020 has brought this group of patients to the center of concern. However, scientific knowledge about the susceptibility of patients with SCD to a severe COVID-19 pandemic is still insufficient, and efforts to establish a general profile of the disease in these patients, remain inadequate. The present study, therefore, sought to characterize the case fatality rate and severity of COVID-19 in patients with SCD throughout the world. A systematic review of Pubmed/MEDLINE, Scopus, Cochrane Library, and Virtual Health Library databases through December 2021 was then performed. Subsequently, the primary and secondary outcomes were used in the meta-analysis in RStudio® software. Seventy-two studies were included with 6,011 SCD patients confirmed to have SARS-CoV-2 infection between mid-2020 and early 2022. The mean age of patients was 27 years. During this period, 218 deaths caused by COVID-19 were reported in the studied population, corresponding to an overall case fatality rate of 3%. In addition, 10% of patients with SCD were admitted to the ICU after complications caused by COVID-19, and 4% of them required invasive ventilatory support. In conclusion, the high fatality rate, intensive care unit admission and need for mechanical ventilation due to COVID-19 in young patients with SCD indicate that this population is at high risk for severe disease progression.
Collapse
Affiliation(s)
- Tarcísio Silva Borborema
- Faculdade Dinâmica do Vale do Piranga, Ponte Nova, Minas Gerais, Brazil
- Hospital Infantil João Paulo II, Belo Horizonte, MG, Brazil
| | | | | | - Rodrigo Siqueira Batista
- Faculdade Dinâmica do Vale do Piranga, Ponte Nova, Minas Gerais, Brazil
- Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| |
Collapse
|