Other Sites:
Robert J. Robbins is a biologist, an educator, a science administrator, a publisher, an information technologist, and an IT leader and manager who specializes in advancing biomedical knowledge and supporting education through the application of information technology. More About: RJR | OUR TEAM | OUR SERVICES | THIS WEBSITE
RJR: Recommended Bibliography 19 Jan 2026 at 01:41 Created:
covid-19
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS coronavirus 2, or SARS-CoV-2), a virus closely related to the SARS virus. The disease was discovered and named during the 2019-20 coronavirus outbreak. Those affected may develop a fever, dry cough, fatigue, and shortness of breath. A sore throat, runny nose or sneezing is less common. While the majority of cases result in mild symptoms, some can progress to pneumonia and multi-organ failure. The infection is spread from one person to others via respiratory droplets produced from the airways, often during coughing or sneezing. Time from exposure to onset of symptoms is generally between 2 and 14 days, with an average of 5 days. The standard method of diagnosis is by reverse transcription polymerase chain reaction (rRT-PCR) from a nasopharyngeal swab or sputum sample, with results within a few hours to 2 days. Antibody assays can also be used, using a blood serum sample, with results within a few days. The infection can also be diagnosed from a combination of symptoms, risk factors and a chest CT scan showing features of pneumonia. Correct handwashing technique, maintaining distance from people who are coughing and not touching one's face with unwashed hands are measures recommended to prevent the disease. It is also recommended to cover one's nose and mouth with a tissue or a bent elbow when coughing. Those who suspect they carry the virus are recommended to wear a surgical face mask and seek medical advice by calling a doctor rather than visiting a clinic in person. Masks are also recommended for those who are taking care of someone with a suspected infection but not for the general public. There is no vaccine or specific antiviral treatment, with management involving treatment of symptoms, supportive care and experimental measures. The case fatality rate is estimated at between 1% and 3%. The World Health Organization (WHO) has declared the 2019-20 coronavirus outbreak a Public Health Emergency of International Concern (PHEIC). As of 29 February 2020, China, Hong Kong, Iran, Italy, Japan, Singapore, South Korea and the United States are areas having evidence of community transmission of the disease.
Created with PubMed® Query: ( SARS-CoV-2 OR COVID-19 OR (wuhan AND coronavirus) AND review[SB] )NOT 40982904[pmid] NOT 40982965[pmid] NOT pmcbook NOT ispreviousversion
Citations The Papers (from PubMed®)
RevDate: 2025-08-13
CmpDate: 2023-10-27
Cell-intrinsic and -extrinsic effects of SARS-CoV-2 RNA on pathogenesis: single-cell meta-analysis.
mSphere, 8(5):e0037523.
Single-cell RNA-seq has been used to characterize human COVID-19. To determine if preclinical models successfully mimic the cell-intrinsic and -extrinsic effects of severe disease, we conducted a meta-analysis of single-cell data across five model species. To assess whether dissemination of viral RNA in lung cells tracks pathology and results in cell-intrinsic and -extrinsic transcriptomic changes in COVID-19. We conducted a meta-analysis by analyzing six publicly available, scRNA-seq data sets. We used dual mapping (host and virus) and differential gene expression analyses to compare viral[+] and viral[-] cell populations. We conducted a principal component analysis to identify successful models of human COVID-19. We found expression of viral RNA in many non-epithelial cell types. Fibroblasts, macrophages, and endothelial cells exhibit clear evidence of viral-intrinsic and -extrinsic effects on host gene expression. Using viral RNA expression, we found that K18-hACE2 mice most closely modeled severe human COVID-19, followed by hamsters. Ferrets and macaques are poor models of human disease due to the low presence of viral RNA. Moreover, we found that increased transcripts of certain key inflammatory genes such as IL1B, IL18, and CXCL10 are not restricted to virally infected cells, suggesting these genes are regulated in a paracrine or autocrine fashion. These data affirm widespread dissemination of viral RNA in the lung, which may be key in the pathogenesis of severe COVID-19 and demonstrate ferrets and Rhesus macaques are poor models of human COVID-19. IMPORTANCE We conducted a high-resolution meta-analysis of scRNA-seq data from humans and five animal models of COVID-19. This study reports viral RNA dissemination in several cell types in human data as well as in some of the pre-clinical models. Using this metric, the K18-hACE2 mouse model, followed by the hamster model, most closely resembled human COVID-19. We observed clear evidence of viral-intrinsic effects within cells (e.g., IRF5 expression) as well as viral-extrinsic cytokine modulation (e.g., IL1B, IL18, CXCL10). We observed proinflammatory chemokine expression in cells devoid of viral RNA expression, suggesting autocrine/paracrine interferon regulation. This report serves as a resource-synthesizing data from COVID-19 humans and animal models and suggesting improvements for relevant pre-clinical models that may aid future diagnostic and therapeutic development projects.
Additional Links: PMID-37737611
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37737611,
year = {2023},
author = {Khatun, MS and Remcho, TP and Qin, X and Kolls, JK},
title = {Cell-intrinsic and -extrinsic effects of SARS-CoV-2 RNA on pathogenesis: single-cell meta-analysis.},
journal = {mSphere},
volume = {8},
number = {5},
pages = {e0037523},
pmid = {37737611},
issn = {2379-5042},
support = {R35 HL139930/HL/NHLBI NIH HHS/United States ; R01 DK129881/DK/NIDDK NIH HHS/United States ; AHA962950/AHA/American Heart Association-American Stroke Association/United States ; P51 OD011104/OD/NIH HHS/United States ; R01 HL165265/HL/NHLBI NIH HHS/United States ; R35HL139930/HL/NHLBI NIH HHS/United States ; 962950/AHA/American Heart Association-American Stroke Association/United States ; HL165265/HL/NHLBI NIH HHS/United States ; },
mesh = {Cricetinae ; Humans ; Animals ; Mice ; *RNA, Viral/genetics ; SARS-CoV-2/genetics ; Endothelial Cells ; Ferrets ; Interleukin-18 ; Macaca mulatta ; *COVID-19 ; },
abstract = {Single-cell RNA-seq has been used to characterize human COVID-19. To determine if preclinical models successfully mimic the cell-intrinsic and -extrinsic effects of severe disease, we conducted a meta-analysis of single-cell data across five model species. To assess whether dissemination of viral RNA in lung cells tracks pathology and results in cell-intrinsic and -extrinsic transcriptomic changes in COVID-19. We conducted a meta-analysis by analyzing six publicly available, scRNA-seq data sets. We used dual mapping (host and virus) and differential gene expression analyses to compare viral[+] and viral[-] cell populations. We conducted a principal component analysis to identify successful models of human COVID-19. We found expression of viral RNA in many non-epithelial cell types. Fibroblasts, macrophages, and endothelial cells exhibit clear evidence of viral-intrinsic and -extrinsic effects on host gene expression. Using viral RNA expression, we found that K18-hACE2 mice most closely modeled severe human COVID-19, followed by hamsters. Ferrets and macaques are poor models of human disease due to the low presence of viral RNA. Moreover, we found that increased transcripts of certain key inflammatory genes such as IL1B, IL18, and CXCL10 are not restricted to virally infected cells, suggesting these genes are regulated in a paracrine or autocrine fashion. These data affirm widespread dissemination of viral RNA in the lung, which may be key in the pathogenesis of severe COVID-19 and demonstrate ferrets and Rhesus macaques are poor models of human COVID-19. IMPORTANCE We conducted a high-resolution meta-analysis of scRNA-seq data from humans and five animal models of COVID-19. This study reports viral RNA dissemination in several cell types in human data as well as in some of the pre-clinical models. Using this metric, the K18-hACE2 mouse model, followed by the hamster model, most closely resembled human COVID-19. We observed clear evidence of viral-intrinsic effects within cells (e.g., IRF5 expression) as well as viral-extrinsic cytokine modulation (e.g., IL1B, IL18, CXCL10). We observed proinflammatory chemokine expression in cells devoid of viral RNA expression, suggesting autocrine/paracrine interferon regulation. This report serves as a resource-synthesizing data from COVID-19 humans and animal models and suggesting improvements for relevant pre-clinical models that may aid future diagnostic and therapeutic development projects.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Cricetinae
Humans
Animals
Mice
*RNA, Viral/genetics
SARS-CoV-2/genetics
Endothelial Cells
Ferrets
Interleukin-18
Macaca mulatta
*COVID-19
RevDate: 2025-12-11
CmpDate: 2024-03-07
Loneliness and depression: bidirectional mendelian randomization analyses using data from three large genome-wide association studies.
Molecular psychiatry, 28(11):4594-4601.
Major depression (MD) is a serious psychiatric illness afflicting nearly 5% of the world's population. A large correlational literature suggests that loneliness is a prospective risk factor for MD; correlational assocations of this nature may be confounded for a variety of reasons. This report uses Mendelian Randomization (MR) to examine potentially causal associations between loneliness and MD. We report on analyses using summary statistics from three large genome wide association studies (GWAS). MR analyses were conducted using three independent sources of GWAS summary statistics. In the first set of analyses, we used available summary statistics from an extant GWAS of loneliness to predict MD risk. We used two sources of outcome data: the Psychiatric Genomics Consortium (PGC) meta-analysis of MD (PGC-MD; N = 142,646) and the Million Veteran Program (MVP-MD; N = 250,215). Finally, we reversed analyses using data from the MVP and PGC samples to identify risk variants for MD and used loneliness outcome data from UK Biobank. We find robust evidence for a bidirectional causal relationship between loneliness and MD, including between loneliness, depression cases status, and a continuous measure of depressive symptoms. The estimates remained significant across several sensitivity analyses, including models that account for horizontal pleiotropy. This paper provides the first genetically-informed evidence that reducing loneliness may play a causal role in decreasing risk for depressive illness, and these findings support efforts to reduce loneliness in order to prevent or ameliorate MD. Discussion focuses on the public health significance of these findings, especially in light of the SARS-CoV-2 pandemic.
Additional Links: PMID-37735503
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37735503,
year = {2023},
author = {Sbarra, DA and Ramadan, FA and Choi, KW and Treur, JL and Levey, DF and Wootton, RE and Stein, MB and Gelernter, J and Klimentidis, YC},
title = {Loneliness and depression: bidirectional mendelian randomization analyses using data from three large genome-wide association studies.},
journal = {Molecular psychiatry},
volume = {28},
number = {11},
pages = {4594-4601},
pmid = {37735503},
issn = {1476-5578},
support = {K08 MH127413/MH/NIMH NIH HHS/United States ; R01 AG078361/AG/NIA NIH HHS/United States ; K08MH127413//U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)/ ; R01AG078361-01//U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)/ ; },
mesh = {Humans ; *Genome-Wide Association Study ; Depression/genetics ; Loneliness ; Mendelian Randomization Analysis ; Prospective Studies ; *Depressive Disorder, Major/genetics ; },
abstract = {Major depression (MD) is a serious psychiatric illness afflicting nearly 5% of the world's population. A large correlational literature suggests that loneliness is a prospective risk factor for MD; correlational assocations of this nature may be confounded for a variety of reasons. This report uses Mendelian Randomization (MR) to examine potentially causal associations between loneliness and MD. We report on analyses using summary statistics from three large genome wide association studies (GWAS). MR analyses were conducted using three independent sources of GWAS summary statistics. In the first set of analyses, we used available summary statistics from an extant GWAS of loneliness to predict MD risk. We used two sources of outcome data: the Psychiatric Genomics Consortium (PGC) meta-analysis of MD (PGC-MD; N = 142,646) and the Million Veteran Program (MVP-MD; N = 250,215). Finally, we reversed analyses using data from the MVP and PGC samples to identify risk variants for MD and used loneliness outcome data from UK Biobank. We find robust evidence for a bidirectional causal relationship between loneliness and MD, including between loneliness, depression cases status, and a continuous measure of depressive symptoms. The estimates remained significant across several sensitivity analyses, including models that account for horizontal pleiotropy. This paper provides the first genetically-informed evidence that reducing loneliness may play a causal role in decreasing risk for depressive illness, and these findings support efforts to reduce loneliness in order to prevent or ameliorate MD. Discussion focuses on the public health significance of these findings, especially in light of the SARS-CoV-2 pandemic.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Genome-Wide Association Study
Depression/genetics
Loneliness
Mendelian Randomization Analysis
Prospective Studies
*Depressive Disorder, Major/genetics
RevDate: 2025-09-22
CmpDate: 2024-02-02
Bayesian hierarchical models incorporating study-level covariates for multivariate meta-analysis of diagnostic tests without a gold standard with application to COVID-19.
Statistics in medicine, 42(28):5085-5099.
When evaluating a diagnostic test, it is common that a gold standard may not be available. One example is the diagnosis of SARS-CoV-2 infection using saliva sampling or nasopharyngeal swabs. Without a gold standard, a pragmatic approach is to postulate a "reference standard," defined as positive if either test is positive, or negative if both are negative. However, this pragmatic approach may overestimate sensitivities because subjects infected with SARS-CoV-2 may still have double-negative test results even when both tests exhibit perfect specificity. To address this limitation, we propose a Bayesian hierarchical model for simultaneously estimating sensitivity, specificity, and disease prevalence in the absence of a gold standard. The proposed model allows adjusting for study-level covariates. We evaluate the model performance using an example based on a recently published meta-analysis on the diagnosis of SARS-CoV-2 infection and extensive simulations. Compared with the pragmatic reference standard approach, we demonstrate that the proposed Bayesian method provides a more accurate evaluation of prevalence, specificity, and sensitivity in a meta-analytic framework.
Additional Links: PMID-37724773
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37724773,
year = {2023},
author = {Wang, Z and Murray, TA and Xiao, M and Lin, L and Alemayehu, D and Chu, H},
title = {Bayesian hierarchical models incorporating study-level covariates for multivariate meta-analysis of diagnostic tests without a gold standard with application to COVID-19.},
journal = {Statistics in medicine},
volume = {42},
number = {28},
pages = {5085-5099},
pmid = {37724773},
issn = {1097-0258},
support = {UL1 TR002494/TR/NCATS NIH HHS/United States ; UL1 TR002535/TR/NCATS NIH HHS/United States ; R01 LM012982/LM/NLM NIH HHS/United States ; UL1-TR002535/TR/NCATS NIH HHS/United States ; R03 MH128727/MH/NIMH NIH HHS/United States ; },
mesh = {Humans ; *COVID-19/diagnosis/epidemiology ; SARS-CoV-2 ; Bayes Theorem ; Sensitivity and Specificity ; Diagnostic Tests, Routine/methods ; COVID-19 Testing ; },
abstract = {When evaluating a diagnostic test, it is common that a gold standard may not be available. One example is the diagnosis of SARS-CoV-2 infection using saliva sampling or nasopharyngeal swabs. Without a gold standard, a pragmatic approach is to postulate a "reference standard," defined as positive if either test is positive, or negative if both are negative. However, this pragmatic approach may overestimate sensitivities because subjects infected with SARS-CoV-2 may still have double-negative test results even when both tests exhibit perfect specificity. To address this limitation, we propose a Bayesian hierarchical model for simultaneously estimating sensitivity, specificity, and disease prevalence in the absence of a gold standard. The proposed model allows adjusting for study-level covariates. We evaluate the model performance using an example based on a recently published meta-analysis on the diagnosis of SARS-CoV-2 infection and extensive simulations. Compared with the pragmatic reference standard approach, we demonstrate that the proposed Bayesian method provides a more accurate evaluation of prevalence, specificity, and sensitivity in a meta-analytic framework.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/diagnosis/epidemiology
SARS-CoV-2
Bayes Theorem
Sensitivity and Specificity
Diagnostic Tests, Routine/methods
COVID-19 Testing
RevDate: 2025-05-30
CmpDate: 2023-09-13
Global diversity and antimicrobial resistance of typhoid fever pathogens: Insights from a meta-analysis of 13,000 Salmonella Typhi genomes.
eLife, 12:.
BACKGROUND: The Global Typhoid Genomics Consortium was established to bring together the typhoid research community to aggregate and analyse Salmonella enterica serovar Typhi (Typhi) genomic data to inform public health action. This analysis, which marks 22 years since the publication of the first Typhi genome, represents the largest Typhi genome sequence collection to date (n=13,000).
METHODS: This is a meta-analysis of global genotype and antimicrobial resistance (AMR) determinants extracted from previously sequenced genome data and analysed using consistent methods implemented in open analysis platforms GenoTyphi and Pathogenwatch.
RESULTS: Compared with previous global snapshots, the data highlight that genotype 4.3.1 (H58) has not spread beyond Asia and Eastern/Southern Africa; in other regions, distinct genotypes dominate and have independently evolved AMR. Data gaps remain in many parts of the world, and we show the potential of travel-associated sequences to provide informal 'sentinel' surveillance for such locations. The data indicate that ciprofloxacin non-susceptibility (>1 resistance determinant) is widespread across geographies and genotypes, with high-level ciprofloxacin resistance (≥3 determinants) reaching 20% prevalence in South Asia. Extensively drug-resistant (XDR) typhoid has become dominant in Pakistan (70% in 2020) but has not yet become established elsewhere. Ceftriaxone resistance has emerged in eight non-XDR genotypes, including a ciprofloxacin-resistant lineage (4.3.1.2.1) in India. Azithromycin resistance mutations were detected at low prevalence in South Asia, including in two common ciprofloxacin-resistant genotypes.
CONCLUSIONS: The consortium's aim is to encourage continued data sharing and collaboration to monitor the emergence and global spread of AMR Typhi, and to inform decision-making around the introduction of typhoid conjugate vaccines (TCVs) and other prevention and control strategies.
FUNDING: No specific funding was awarded for this meta-analysis. Coordinators were supported by fellowships from the European Union (ZAD received funding from the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 845681), the Wellcome Trust (SB, Wellcome Trust Senior Fellowship), and the National Health and Medical Research Council (DJI is supported by an NHMRC Investigator Grant [GNT1195210]).
Additional Links: PMID-37697804
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37697804,
year = {2023},
author = {Carey, ME and Dyson, ZA and Ingle, DJ and Amir, A and Aworh, MK and Chattaway, MA and Chew, KL and Crump, JA and Feasey, NA and Howden, BP and Keddy, KH and Maes, M and Parry, CM and Van Puyvelde, S and Webb, HE and Afolayan, AO and Alexander, AP and Anandan, S and Andrews, JR and Ashton, PM and Basnyat, B and Bavdekar, A and Bogoch, II and Clemens, JD and da Silva, KE and De, A and de Ligt, J and Diaz Guevara, PL and Dolecek, C and Dutta, S and Ehlers, MM and Francois Watkins, L and Garrett, DO and Godbole, G and Gordon, MA and Greenhill, AR and Griffin, C and Gupta, M and Hendriksen, RS and Heyderman, RS and Hooda, Y and Hormazabal, JC and Ikhimiukor, OO and Iqbal, J and Jacob, JJ and Jenkins, C and Jinka, DR and John, J and Kang, G and Kanteh, A and Kapil, A and Karkey, A and Kariuki, S and Kingsley, RA and Koshy, RM and Lauer, AC and Levine, MM and Lingegowda, RK and Luby, SP and Mackenzie, GA and Mashe, T and Msefula, C and Mutreja, A and Nagaraj, G and Nagaraj, S and Nair, S and Naseri, TK and Nimarota-Brown, S and Njamkepo, E and Okeke, IN and Perumal, SPB and Pollard, AJ and Pragasam, AK and Qadri, F and Qamar, FN and Rahman, SIA and Rambocus, SD and Rasko, DA and Ray, P and Robins-Browne, R and Rongsen-Chandola, T and Rutanga, JP and Saha, SK and Saha, S and Saigal, K and Sajib, MSI and Seidman, JC and Shakya, J and Shamanna, V and Shastri, J and Shrestha, R and Sia, S and Sikorski, MJ and Singh, A and Smith, AM and Tagg, KA and Tamrakar, D and Tanmoy, AM and Thomas, M and Thomas, MS and Thomsen, R and Thomson, NR and Tupua, S and Vaidya, K and Valcanis, M and Veeraraghavan, B and Weill, FX and Wright, J and Dougan, G and Argimón, S and Keane, JA and Aanensen, DM and Baker, S and Holt, KE and , },
title = {Global diversity and antimicrobial resistance of typhoid fever pathogens: Insights from a meta-analysis of 13,000 Salmonella Typhi genomes.},
journal = {eLife},
volume = {12},
number = {},
pages = {},
pmid = {37697804},
issn = {2050-084X},
support = {R01 TW009237/TW/FIC NIH HHS/United States ; BBS/E/F/00PR10348/BB_/Biotechnology and Biological Sciences Research Council/United Kingdom ; F30 AI156973/AI/NIAID NIH HHS/United States ; BB/R012504/1/BB_/Biotechnology and Biological Sciences Research Council/United Kingdom ; U01 AI062563/AI/NIAID NIH HHS/United States ; MR/TOO5033/1/MRC_/Medical Research Council/United Kingdom ; 206545/7/17/Z/WT_/Wellcome Trust/United Kingdom ; /WT_/Wellcome Trust/United Kingdom ; 106158/7/14/Z/WT_/Wellcome Trust/United Kingdom ; 001/WHO_/World Health Organization/International ; R01 AI121378/AI/NIAID NIH HHS/United States ; U19 AI110820/AI/NIAID NIH HHS/United States ; R01 AI099525/AI/NIAID NIH HHS/United States ; 206194/WT_/Wellcome Trust/United Kingdom ; MR/L00464X//MRC_/Medical Research Council/United Kingdom ; },
mesh = {Humans ; *Salmonella typhi/genetics ; *Typhoid Fever/epidemiology ; Anti-Bacterial Agents/pharmacology ; Travel ; Drug Resistance, Bacterial/genetics ; Ciprofloxacin ; },
abstract = {BACKGROUND: The Global Typhoid Genomics Consortium was established to bring together the typhoid research community to aggregate and analyse Salmonella enterica serovar Typhi (Typhi) genomic data to inform public health action. This analysis, which marks 22 years since the publication of the first Typhi genome, represents the largest Typhi genome sequence collection to date (n=13,000).
METHODS: This is a meta-analysis of global genotype and antimicrobial resistance (AMR) determinants extracted from previously sequenced genome data and analysed using consistent methods implemented in open analysis platforms GenoTyphi and Pathogenwatch.
RESULTS: Compared with previous global snapshots, the data highlight that genotype 4.3.1 (H58) has not spread beyond Asia and Eastern/Southern Africa; in other regions, distinct genotypes dominate and have independently evolved AMR. Data gaps remain in many parts of the world, and we show the potential of travel-associated sequences to provide informal 'sentinel' surveillance for such locations. The data indicate that ciprofloxacin non-susceptibility (>1 resistance determinant) is widespread across geographies and genotypes, with high-level ciprofloxacin resistance (≥3 determinants) reaching 20% prevalence in South Asia. Extensively drug-resistant (XDR) typhoid has become dominant in Pakistan (70% in 2020) but has not yet become established elsewhere. Ceftriaxone resistance has emerged in eight non-XDR genotypes, including a ciprofloxacin-resistant lineage (4.3.1.2.1) in India. Azithromycin resistance mutations were detected at low prevalence in South Asia, including in two common ciprofloxacin-resistant genotypes.
CONCLUSIONS: The consortium's aim is to encourage continued data sharing and collaboration to monitor the emergence and global spread of AMR Typhi, and to inform decision-making around the introduction of typhoid conjugate vaccines (TCVs) and other prevention and control strategies.
FUNDING: No specific funding was awarded for this meta-analysis. Coordinators were supported by fellowships from the European Union (ZAD received funding from the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 845681), the Wellcome Trust (SB, Wellcome Trust Senior Fellowship), and the National Health and Medical Research Council (DJI is supported by an NHMRC Investigator Grant [GNT1195210]).},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Salmonella typhi/genetics
*Typhoid Fever/epidemiology
Anti-Bacterial Agents/pharmacology
Travel
Drug Resistance, Bacterial/genetics
Ciprofloxacin
RevDate: 2023-11-29
CmpDate: 2023-08-28
Use of Nirsevimab for the Prevention of Respiratory Syncytial Virus Disease Among Infants and Young Children: Recommendations of the Advisory Committee on Immunization Practices - United States, 2023.
MMWR. Morbidity and mortality weekly report, 72(34):920-925.
Respiratory syncytial virus (RSV) is the leading cause of hospitalization among U.S. infants. In July 2023, the Food and Drug Administration approved nirsevimab, a long-acting monoclonal antibody, for passive immunization to prevent RSV-associated lower respiratory tract infection among infants and young children. Since October 2021, the Advisory Committee on Immunization Practices (ACIP) Maternal and Pediatric RSV Work Group has reviewed evidence on the safety and efficacy of nirsevimab among infants and young children. On August 3, 2023, ACIP recommended nirsevimab for all infants aged <8 months who are born during or entering their first RSV season and for infants and children aged 8-19 months who are at increased risk for severe RSV disease and are entering their second RSV season. On the basis of pre-COVID-19 pandemic patterns, nirsevimab could be administered in most of the continental United States from October through the end of March. Nirsevimab can prevent severe RSV disease among infants and young children at increased risk for severe RSV disease.
Additional Links: PMID-37616235
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37616235,
year = {2023},
author = {Jones, JM and Fleming-Dutra, KE and Prill, MM and Roper, LE and Brooks, O and Sánchez, PJ and Kotton, CN and Mahon, BE and Meyer, S and Long, SS and McMorrow, ML},
title = {Use of Nirsevimab for the Prevention of Respiratory Syncytial Virus Disease Among Infants and Young Children: Recommendations of the Advisory Committee on Immunization Practices - United States, 2023.},
journal = {MMWR. Morbidity and mortality weekly report},
volume = {72},
number = {34},
pages = {920-925},
pmid = {37616235},
issn = {1545-861X},
mesh = {Humans ; Infant ; Advisory Committees ; *Communicable Diseases ; *COVID-19 ; Immunization ; Pandemics ; *Respiratory Syncytial Virus Infections/epidemiology/prevention & control ; *Respiratory Syncytial Virus, Human ; United States/epidemiology ; },
abstract = {Respiratory syncytial virus (RSV) is the leading cause of hospitalization among U.S. infants. In July 2023, the Food and Drug Administration approved nirsevimab, a long-acting monoclonal antibody, for passive immunization to prevent RSV-associated lower respiratory tract infection among infants and young children. Since October 2021, the Advisory Committee on Immunization Practices (ACIP) Maternal and Pediatric RSV Work Group has reviewed evidence on the safety and efficacy of nirsevimab among infants and young children. On August 3, 2023, ACIP recommended nirsevimab for all infants aged <8 months who are born during or entering their first RSV season and for infants and children aged 8-19 months who are at increased risk for severe RSV disease and are entering their second RSV season. On the basis of pre-COVID-19 pandemic patterns, nirsevimab could be administered in most of the continental United States from October through the end of March. Nirsevimab can prevent severe RSV disease among infants and young children at increased risk for severe RSV disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Infant
Advisory Committees
*Communicable Diseases
*COVID-19
Immunization
Pandemics
*Respiratory Syncytial Virus Infections/epidemiology/prevention & control
*Respiratory Syncytial Virus, Human
United States/epidemiology
RevDate: 2023-11-21
CmpDate: 2023-08-16
Maast: genotyping thousands of microbial strains efficiently.
Genome biology, 24(1):186.
Existing single nucleotide polymorphism (SNP) genotyping algorithms do not scale for species with thousands of sequenced strains, nor do they account for conspecific redundancy. Here we present a bioinformatics tool, Maast, which empowers population genetic meta-analysis of microbes at an unrivaled scale. Maast implements a novel algorithm to heuristically identify a minimal set of diverse conspecific genomes, then constructs a reliable SNP panel for each species, and enables rapid and accurate genotyping using a hybrid of whole-genome alignment and k-mer exact matching. We demonstrate Maast's utility by genotyping thousands of Helicobacter pylori strains and tracking SARS-CoV-2 diversification.
Additional Links: PMID-37563669
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37563669,
year = {2023},
author = {Shi, ZJ and Nayfach, S and Pollard, KS},
title = {Maast: genotyping thousands of microbial strains efficiently.},
journal = {Genome biology},
volume = {24},
number = {1},
pages = {186},
pmid = {37563669},
issn = {1474-760X},
support = {R01 HL160862/HL/NHLBI NIH HHS/United States ; },
mesh = {Humans ; Genotype ; *SARS-CoV-2/genetics ; *COVID-19 ; Genome ; Algorithms ; Polymorphism, Single Nucleotide ; Genotyping Techniques ; },
abstract = {Existing single nucleotide polymorphism (SNP) genotyping algorithms do not scale for species with thousands of sequenced strains, nor do they account for conspecific redundancy. Here we present a bioinformatics tool, Maast, which empowers population genetic meta-analysis of microbes at an unrivaled scale. Maast implements a novel algorithm to heuristically identify a minimal set of diverse conspecific genomes, then constructs a reliable SNP panel for each species, and enables rapid and accurate genotyping using a hybrid of whole-genome alignment and k-mer exact matching. We demonstrate Maast's utility by genotyping thousands of Helicobacter pylori strains and tracking SARS-CoV-2 diversification.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Genotype
*SARS-CoV-2/genetics
*COVID-19
Genome
Algorithms
Polymorphism, Single Nucleotide
Genotyping Techniques
RevDate: 2025-05-30
CmpDate: 2023-12-04
Meta-Analysis of COVID-19 BAL Single-Cell RNA Sequencing Reveals Alveolar Epithelial Transitions and Unique Alveolar Epithelial Cell Fates.
American journal of respiratory cell and molecular biology, 69(6):623-637.
Single-cell RNA sequencing (scRNA-seq) of BAL cells has provided insights into coronavirus disease (COVID-19). However, reports have been limited by small patient cohorts. We performed a meta-analysis of BAL scRNA-seq data from healthy control subjects (n = 13) and patients with COVID-19 (n = 20), sourced from six independent studies (167,280 high-quality cells in total). Consistent with the source reports, increases in infiltrating leukocyte subtypes were noted, several with type I IFN signatures and unique gene expression signatures associated with transcellular chemokine signaling. Noting dramatic reductions of inferred NKX2-1 and NR4A1 activity in alveolar epithelial type II (AT-II) cells, we modeled pseudotemporal AT-II-to-AT-I progression. This revealed changes in inferred AT-II cell metabolic activity, increased transitional cells, and a previously undescribed AT-I state. This cell state was conspicuously marked by the induction of genes of the epidermal differentiation complex, including the cornified envelope protein SPRR3 (small proline-rich protein 3), upregulation of multiple KRT (keratin) genes, inferred mitochondrial dysfunction, and cell death signatures including apoptosis and ferroptosis. Immunohistochemistry of lungs from patients with COVID-19 confirmed upregulation and colocalization of KRT13 and SPRR3 in the distal airspaces. Forced overexpression of SPRR3 in human alveolar epithelial cells ex vivo did not activate caspase-3 or upregulate KRT13, suggesting that SPRR3 marks an AT-I cornification program in COVID-19 but is not sufficient for phenotypic changes.
Additional Links: PMID-37523502
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37523502,
year = {2023},
author = {Karmaus, PWF and Tata, A and Meacham, JM and Day, F and Thrower, D and Tata, PR and Fessler, MB},
title = {Meta-Analysis of COVID-19 BAL Single-Cell RNA Sequencing Reveals Alveolar Epithelial Transitions and Unique Alveolar Epithelial Cell Fates.},
journal = {American journal of respiratory cell and molecular biology},
volume = {69},
number = {6},
pages = {623-637},
pmid = {37523502},
issn = {1535-4989},
support = {Z01 ES102005/ES/NIEHS NIH HHS/United States ; R01 HL153375/HL/NHLBI NIH HHS/United States ; Z01 ES102005/ImNIH/Intramural NIH HHS/United States ; P30 CA014236/CA/NCI NIH HHS/United States ; UM1 CA239755/CA/NCI NIH HHS/United States ; },
mesh = {Humans ; *Alveolar Epithelial Cells ; *COVID-19/genetics/metabolism ; Lung ; Epithelial Cells/metabolism ; Sequence Analysis, RNA ; },
abstract = {Single-cell RNA sequencing (scRNA-seq) of BAL cells has provided insights into coronavirus disease (COVID-19). However, reports have been limited by small patient cohorts. We performed a meta-analysis of BAL scRNA-seq data from healthy control subjects (n = 13) and patients with COVID-19 (n = 20), sourced from six independent studies (167,280 high-quality cells in total). Consistent with the source reports, increases in infiltrating leukocyte subtypes were noted, several with type I IFN signatures and unique gene expression signatures associated with transcellular chemokine signaling. Noting dramatic reductions of inferred NKX2-1 and NR4A1 activity in alveolar epithelial type II (AT-II) cells, we modeled pseudotemporal AT-II-to-AT-I progression. This revealed changes in inferred AT-II cell metabolic activity, increased transitional cells, and a previously undescribed AT-I state. This cell state was conspicuously marked by the induction of genes of the epidermal differentiation complex, including the cornified envelope protein SPRR3 (small proline-rich protein 3), upregulation of multiple KRT (keratin) genes, inferred mitochondrial dysfunction, and cell death signatures including apoptosis and ferroptosis. Immunohistochemistry of lungs from patients with COVID-19 confirmed upregulation and colocalization of KRT13 and SPRR3 in the distal airspaces. Forced overexpression of SPRR3 in human alveolar epithelial cells ex vivo did not activate caspase-3 or upregulate KRT13, suggesting that SPRR3 marks an AT-I cornification program in COVID-19 but is not sufficient for phenotypic changes.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alveolar Epithelial Cells
*COVID-19/genetics/metabolism
Lung
Epithelial Cells/metabolism
Sequence Analysis, RNA
RevDate: 2025-12-18
CmpDate: 2023-07-03
Immunomodulatory Therapy for MIS-C.
Pediatrics, 152(1):.
CONTEXT: Studies comparing initial therapy for multisystem inflammatory syndrome in children (MIS-C) provided conflicting results.
OBJECTIVE: To compare outcomes in MIS-C patients treated with intravenous immunoglobulin (IVIG), glucocorticoids, or the combination thereof.
DATA SOURCES: Medline, Embase, CENTRAL and WOS, from January 2020 to February 2022.
STUDY SELECTION: Randomized or observational comparative studies including MIS-C patients <21 years.
DATA EXTRACTION: Two reviewers independently selected studies and obtained individual participant data. The main outcome was cardiovascular dysfunction (CD), defined as left ventricular ejection fraction < 55% or vasopressor requirement ≥ day 2 of initial therapy, analyzed with a propensity score-matched analysis.
RESULTS: Of 2635 studies identified, 3 nonrandomized cohorts were included. The meta-analysis included 958 children. IVIG plus glucocorticoids group as compared with IVIG alone had improved CD (odds ratio [OR] 0.62 [0.42-0.91]). Glucocorticoids alone group as compared with IVIG alone did not have improved CD (OR 0.57 [0.31-1.05]). Glucocorticoids alone group as compared with IVIG plus glucocorticoids did not have improved CD (OR 0.67 [0.24-1.86]). Secondary analyses found better outcomes associated with IVIG plus glucocorticoids compared with glucocorticoids alone (fever ≥ day 2, need for secondary therapies) and better outcomes associated with glucocorticoids alone compared with IVIG alone (left ventricular ejection fraction < 55% ≥ day 2).
LIMITATIONS: Nonrandomized nature of included studies.
CONCLUSIONS: In a meta-analysis of MIS-C patients, IVIG plus glucocorticoids was associated with improved CD compared with IVIG alone. Glucocorticoids alone was not associated with improved CD compared with IVIG alone or IVIG plus glucocorticoids.
Additional Links: PMID-37376963
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37376963,
year = {2023},
author = {Ouldali, N and Son, MBF and McArdle, AJ and Vito, O and Vaugon, E and Belot, A and Leblanc, C and Murray, NL and Patel, MM and Levin, M and Randolph, AG and Angoulvant, F and , },
title = {Immunomodulatory Therapy for MIS-C.},
journal = {Pediatrics},
volume = {152},
number = {1},
pages = {},
doi = {10.1542/peds.2022-061173},
pmid = {37376963},
issn = {1098-4275},
support = {/CC/CDC HHS/United States ; },
mesh = {Systemic Inflammatory Response Syndrome ; Child ; Immunomodulation ; *Glucocorticoids/therapeutic use ; *Immunoglobulins, Intravenous/therapeutic use ; Ventricular Function, Left ; Humans ; COVID-19/complications ; Stroke Volume ; },
abstract = {CONTEXT: Studies comparing initial therapy for multisystem inflammatory syndrome in children (MIS-C) provided conflicting results.
OBJECTIVE: To compare outcomes in MIS-C patients treated with intravenous immunoglobulin (IVIG), glucocorticoids, or the combination thereof.
DATA SOURCES: Medline, Embase, CENTRAL and WOS, from January 2020 to February 2022.
STUDY SELECTION: Randomized or observational comparative studies including MIS-C patients <21 years.
DATA EXTRACTION: Two reviewers independently selected studies and obtained individual participant data. The main outcome was cardiovascular dysfunction (CD), defined as left ventricular ejection fraction < 55% or vasopressor requirement ≥ day 2 of initial therapy, analyzed with a propensity score-matched analysis.
RESULTS: Of 2635 studies identified, 3 nonrandomized cohorts were included. The meta-analysis included 958 children. IVIG plus glucocorticoids group as compared with IVIG alone had improved CD (odds ratio [OR] 0.62 [0.42-0.91]). Glucocorticoids alone group as compared with IVIG alone did not have improved CD (OR 0.57 [0.31-1.05]). Glucocorticoids alone group as compared with IVIG plus glucocorticoids did not have improved CD (OR 0.67 [0.24-1.86]). Secondary analyses found better outcomes associated with IVIG plus glucocorticoids compared with glucocorticoids alone (fever ≥ day 2, need for secondary therapies) and better outcomes associated with glucocorticoids alone compared with IVIG alone (left ventricular ejection fraction < 55% ≥ day 2).
LIMITATIONS: Nonrandomized nature of included studies.
CONCLUSIONS: In a meta-analysis of MIS-C patients, IVIG plus glucocorticoids was associated with improved CD compared with IVIG alone. Glucocorticoids alone was not associated with improved CD compared with IVIG alone or IVIG plus glucocorticoids.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Systemic Inflammatory Response Syndrome
Child
Immunomodulation
*Glucocorticoids/therapeutic use
*Immunoglobulins, Intravenous/therapeutic use
Ventricular Function, Left
Humans
COVID-19/complications
Stroke Volume
RevDate: 2025-05-03
CmpDate: 2023-06-21
Losartan in hospitalized patients with COVID-19 in North America: An individual participant data meta-analysis.
Medicine, 102(23):e33904.
BACKGROUND: Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers (ARBs) have been hypothesized to benefit patients with COVID-19 via the inhibition of viral entry and other mechanisms. We conducted an individual participant data (IPD) meta-analysis assessing the effect of starting the ARB losartan in recently hospitalized COVID-19 patients.
METHODS: We searched ClinicalTrials.gov in January 2021 for U.S./Canada-based trials where an angiotensin-converting enzyme inhibitors/ARB was a treatment arm, targeted outcomes could be extrapolated, and data sharing was allowed. Our primary outcome was a 7-point COVID-19 ordinal score measured 13 to 16 days post-enrollment. We analyzed data by fitting multilevel Bayesian ordinal regression models and standardizing the resulting predictions.
RESULTS: 325 participants (156 losartan vs 169 control) from 4 studies contributed IPD. Three were randomized trials; one used non-randomized concurrent and historical controls. Baseline covariates were reasonably balanced for the randomized trials. All studies evaluated losartan. We found equivocal evidence of a difference in ordinal scores 13-16 days post-enrollment (model-standardized odds ratio [OR] 1.10, 95% credible interval [CrI] 0.76-1.71; adjusted OR 1.15, 95% CrI 0.15-3.59) and no compelling evidence of treatment effect heterogeneity among prespecified subgroups. Losartan had worse effects for those taking corticosteroids at baseline after adjusting for covariates (ratio of adjusted ORs 0.29, 95% CrI 0.08-0.99). Hypotension serious adverse event rates were numerically higher with losartan.
CONCLUSIONS: In this IPD meta-analysis of hospitalized COVID-19 patients, we found no convincing evidence for the benefit of losartan versus control treatment, but a higher rate of hypotension adverse events with losartan.
Additional Links: PMID-37335665
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37335665,
year = {2023},
author = {Di Stefano, L and Ram, M and Scharfstein, DO and Li, T and Khanal, P and Baksh, SN and McBee, N and Bengtson, CD and Gadomski, A and Geriak, M and Puskarich, MA and Salathe, MA and Schutte, AE and Tignanelli, CJ and Victory, J and Bierer, BE and Hanley, DF and Freilich, DA and , },
title = {Losartan in hospitalized patients with COVID-19 in North America: An individual participant data meta-analysis.},
journal = {Medicine},
volume = {102},
number = {23},
pages = {e33904},
pmid = {37335665},
issn = {1536-5964},
support = {U24 TR001609/TR/NCATS NIH HHS/United States ; },
mesh = {Humans ; Losartan/adverse effects ; Angiotensin Receptor Antagonists/adverse effects ; Angiotensin-Converting Enzyme Inhibitors/adverse effects ; Bayes Theorem ; *COVID-19 ; *Hypotension/chemically induced ; },
abstract = {BACKGROUND: Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers (ARBs) have been hypothesized to benefit patients with COVID-19 via the inhibition of viral entry and other mechanisms. We conducted an individual participant data (IPD) meta-analysis assessing the effect of starting the ARB losartan in recently hospitalized COVID-19 patients.
METHODS: We searched ClinicalTrials.gov in January 2021 for U.S./Canada-based trials where an angiotensin-converting enzyme inhibitors/ARB was a treatment arm, targeted outcomes could be extrapolated, and data sharing was allowed. Our primary outcome was a 7-point COVID-19 ordinal score measured 13 to 16 days post-enrollment. We analyzed data by fitting multilevel Bayesian ordinal regression models and standardizing the resulting predictions.
RESULTS: 325 participants (156 losartan vs 169 control) from 4 studies contributed IPD. Three were randomized trials; one used non-randomized concurrent and historical controls. Baseline covariates were reasonably balanced for the randomized trials. All studies evaluated losartan. We found equivocal evidence of a difference in ordinal scores 13-16 days post-enrollment (model-standardized odds ratio [OR] 1.10, 95% credible interval [CrI] 0.76-1.71; adjusted OR 1.15, 95% CrI 0.15-3.59) and no compelling evidence of treatment effect heterogeneity among prespecified subgroups. Losartan had worse effects for those taking corticosteroids at baseline after adjusting for covariates (ratio of adjusted ORs 0.29, 95% CrI 0.08-0.99). Hypotension serious adverse event rates were numerically higher with losartan.
CONCLUSIONS: In this IPD meta-analysis of hospitalized COVID-19 patients, we found no convincing evidence for the benefit of losartan versus control treatment, but a higher rate of hypotension adverse events with losartan.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Losartan/adverse effects
Angiotensin Receptor Antagonists/adverse effects
Angiotensin-Converting Enzyme Inhibitors/adverse effects
Bayes Theorem
*COVID-19
*Hypotension/chemically induced
RevDate: 2025-09-30
CmpDate: 2023-09-25
A meta-analysis of correction effects in science-relevant misinformation.
Nature human behaviour, 7(9):1514-1525.
Scientifically relevant misinformation, defined as false claims concerning a scientific measurement procedure or scientific evidence, regardless of the author's intent, is illustrated by the fiction that the coronavirus disease 2019 vaccine contained microchips to track citizens. Updating science-relevant misinformation after a correction can be challenging, and little is known about what theoretical factors can influence the correction. Here this meta-analysis examined 205 effect sizes (that is, k, obtained from 74 reports; N = 60,861), which showed that attempts to debunk science-relevant misinformation were, on average, not successful (d = 0.19, P = 0.131, 95% confidence interval -0.06 to 0.43). However, corrections were more successful when the initial science-relevant belief concerned negative topics and domains other than health. Corrections fared better when they were detailed, when recipients were likely familiar with both sides of the issue ahead of the study and when the issue was not politically polarized.
Additional Links: PMID-37322236
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37322236,
year = {2023},
author = {Chan, MS and AlbarracÃn, D},
title = {A meta-analysis of correction effects in science-relevant misinformation.},
journal = {Nature human behaviour},
volume = {7},
number = {9},
pages = {1514-1525},
pmid = {37322236},
issn = {2397-3374},
support = {R01 MH132415/MH/NIMH NIH HHS/United States ; R01 MH094241/MH/NIMH NIH HHS/United States ; P30 AI045008/AI/NIAID NIH HHS/United States ; DP1 DA048570/DA/NIDA NIH HHS/United States ; R01 AI147487/AI/NIAID NIH HHS/United States ; K02 MH075616/MH/NIMH NIH HHS/United States ; R01 MH114847/MH/NIMH NIH HHS/United States ; },
mesh = {Humans ; *COVID-19/prevention & control ; COVID-19 Vaccines ; Intention ; },
abstract = {Scientifically relevant misinformation, defined as false claims concerning a scientific measurement procedure or scientific evidence, regardless of the author's intent, is illustrated by the fiction that the coronavirus disease 2019 vaccine contained microchips to track citizens. Updating science-relevant misinformation after a correction can be challenging, and little is known about what theoretical factors can influence the correction. Here this meta-analysis examined 205 effect sizes (that is, k, obtained from 74 reports; N = 60,861), which showed that attempts to debunk science-relevant misinformation were, on average, not successful (d = 0.19, P = 0.131, 95% confidence interval -0.06 to 0.43). However, corrections were more successful when the initial science-relevant belief concerned negative topics and domains other than health. Corrections fared better when they were detailed, when recipients were likely familiar with both sides of the issue ahead of the study and when the issue was not politically polarized.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/prevention & control
COVID-19 Vaccines
Intention
RevDate: 2024-09-02
CmpDate: 2023-09-04
Genome-Wide Association Study of CKD Progression.
Journal of the American Society of Nephrology : JASN, 34(9):1547-1559.
SIGNIFICANCE STATEMENT: Rapid progression of CKD is associated with poor clinical outcomes. Most previous studies looking for genetic factors associated with low eGFR have used cross-sectional data. The authors conducted a meta-analysis of genome-wide association studies of eGFR decline among 116,870 participants with CKD, focusing on longitudinal data. They identified three loci (two of them novel) associated with longitudinal eGFR decline. In addition to the known UMOD/PDILT locus, variants within BICC1 were associated with significant differences in longitudinal eGFR slope. Variants within HEATR4 also were associated with differences in eGFR decline, but only among Black/African American individuals without diabetes. These findings help characterize molecular mechanisms of eGFR decline in CKD and may inform new therapeutic approaches for progressive kidney disease.
BACKGROUND: Rapid progression of CKD is associated with poor clinical outcomes. Despite extensive study of the genetics of cross-sectional eGFR, only a few loci associated with eGFR decline over time have been identified.
METHODS: We performed a meta-analysis of genome-wide association studies of eGFR decline among 116,870 participants with CKD-defined by two outpatient eGFR measurements of <60 ml/min per 1.73 m 2 , obtained 90-365 days apart-from the Million Veteran Program and Vanderbilt University Medical Center's DNA biobank. The primary outcome was the annualized relative slope in outpatient eGFR. Analyses were stratified by ethnicity and diabetes status and meta-analyzed thereafter.
RESULTS: In cross-ancestry meta-analysis, the strongest association was rs77924615, near UMOD / PDILT ; each copy of the G allele was associated with a 0.30%/yr faster eGFR decline (P = 4.9×10 -27). We also observed an association within BICC1 (rs11592748), where every additional minor allele was associated with a 0.13%/yr slower eGFR decline (P = 5.6×10 -9). Among participants without diabetes, the strongest association was the UMOD/PDILT variant rs36060036, associated with a 0.27%/yr faster eGFR decline per copy of the C allele (P = 1.9×10 -17). Among Black participants, a significantly faster eGFR decline was associated with variant rs16996674 near APOL1 (R 2 =0.29 with the G1 high-risk genotype); among Black participants with diabetes, lead variant rs11624911 near HEATR4 also was associated with a significantly faster eGFR decline. We also nominally replicated loci with known associations with eGFR decline, near PRKAG2, FGF5, and C15ORF54.
CONCLUSIONS: Three loci were significantly associated with longitudinal eGFR change at genome-wide significance. These findings help characterize molecular mechanisms of eGFR decline and may contribute to the development of new therapeutic approaches for progressive CKD.
Additional Links: PMID-37261792
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37261792,
year = {2023},
author = {Robinson-Cohen, C and Triozzi, JL and Rowan, B and He, J and Chen, HC and Zheng, NS and Wei, WQ and Wilson, OD and Hellwege, JN and Tsao, PS and Gaziano, JM and Bick, A and Matheny, ME and Chung, CP and Lipworth, L and Siew, ED and Ikizler, TA and Tao, R and Hung, AM},
title = {Genome-Wide Association Study of CKD Progression.},
journal = {Journal of the American Society of Nephrology : JASN},
volume = {34},
number = {9},
pages = {1547-1559},
pmid = {37261792},
issn = {1533-3450},
support = {U01 HG004798/HG/NHGRI NIH HHS/United States ; UL1 RR024975/RR/NCRR NIH HHS/United States ; U19 HL065962/HL/NHLBI NIH HHS/United States ; I01 CX001897/CX/CSRD VA/United States ; P50 GM115305/GM/NIGMS NIH HHS/United States ; R01 HD074711/HD/NICHD NIH HHS/United States ; R01 NS032830/NS/NINDS NIH HHS/United States ; K12 HD043483/HD/NICHD NIH HHS/United States ; R01 DK122075/DK/NIDDK NIH HHS/United States ; U01 HG006378/HG/NHGRI NIH HHS/United States ; S10 RR025141/RR/NCRR NIH HHS/United States ; UL1 TR002243/TR/NCATS NIH HHS/United States ; UL1 TR000445/TR/NCATS NIH HHS/United States ; RC2 GM092618/GM/NIGMS NIH HHS/United States ; K12 AR084232/AR/NIAMS NIH HHS/United States ; },
mesh = {Humans ; *Genome-Wide Association Study ; *Renal Insufficiency, Chronic/therapy ; Cross-Sectional Studies ; Kidney ; Genotype ; Glomerular Filtration Rate/genetics ; Disease Progression ; Apolipoprotein L1/genetics ; Protein Disulfide-Isomerases/genetics ; },
abstract = {SIGNIFICANCE STATEMENT: Rapid progression of CKD is associated with poor clinical outcomes. Most previous studies looking for genetic factors associated with low eGFR have used cross-sectional data. The authors conducted a meta-analysis of genome-wide association studies of eGFR decline among 116,870 participants with CKD, focusing on longitudinal data. They identified three loci (two of them novel) associated with longitudinal eGFR decline. In addition to the known UMOD/PDILT locus, variants within BICC1 were associated with significant differences in longitudinal eGFR slope. Variants within HEATR4 also were associated with differences in eGFR decline, but only among Black/African American individuals without diabetes. These findings help characterize molecular mechanisms of eGFR decline in CKD and may inform new therapeutic approaches for progressive kidney disease.
BACKGROUND: Rapid progression of CKD is associated with poor clinical outcomes. Despite extensive study of the genetics of cross-sectional eGFR, only a few loci associated with eGFR decline over time have been identified.
METHODS: We performed a meta-analysis of genome-wide association studies of eGFR decline among 116,870 participants with CKD-defined by two outpatient eGFR measurements of <60 ml/min per 1.73 m 2 , obtained 90-365 days apart-from the Million Veteran Program and Vanderbilt University Medical Center's DNA biobank. The primary outcome was the annualized relative slope in outpatient eGFR. Analyses were stratified by ethnicity and diabetes status and meta-analyzed thereafter.
RESULTS: In cross-ancestry meta-analysis, the strongest association was rs77924615, near UMOD / PDILT ; each copy of the G allele was associated with a 0.30%/yr faster eGFR decline (P = 4.9×10 -27). We also observed an association within BICC1 (rs11592748), where every additional minor allele was associated with a 0.13%/yr slower eGFR decline (P = 5.6×10 -9). Among participants without diabetes, the strongest association was the UMOD/PDILT variant rs36060036, associated with a 0.27%/yr faster eGFR decline per copy of the C allele (P = 1.9×10 -17). Among Black participants, a significantly faster eGFR decline was associated with variant rs16996674 near APOL1 (R 2 =0.29 with the G1 high-risk genotype); among Black participants with diabetes, lead variant rs11624911 near HEATR4 also was associated with a significantly faster eGFR decline. We also nominally replicated loci with known associations with eGFR decline, near PRKAG2, FGF5, and C15ORF54.
CONCLUSIONS: Three loci were significantly associated with longitudinal eGFR change at genome-wide significance. These findings help characterize molecular mechanisms of eGFR decline and may contribute to the development of new therapeutic approaches for progressive CKD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Genome-Wide Association Study
*Renal Insufficiency, Chronic/therapy
Cross-Sectional Studies
Kidney
Genotype
Glomerular Filtration Rate/genetics
Disease Progression
Apolipoprotein L1/genetics
Protein Disulfide-Isomerases/genetics
RevDate: 2023-06-17
CmpDate: 2023-06-16
Prognostic performance of blood neurofilament light chain protein in hospitalized COVID-19 patients without major central nervous system manifestations: an individual participant data meta-analysis.
Journal of neurology, 270(7):3315-3328.
BACKGROUND AND AIMS: To investigate the prognostic value of blood neurofilament light chain protein (NfL) levels in the acute phase of coronavirus disease 2019 (COVID-19).
METHODS: We conducted an individual participant data (IPD) meta-analysis after screening on MEDLINE and Scopus to May 23rd 2022. We included studies with hospitalized adult COVID-19 patients without major COVID-19-associated central nervous system (CNS) manifestations and with a measurement of blood NfL in the acute phase as well as data regarding at least one clinical outcome including intensive care unit (ICU) admission, need of mechanical ventilation (MV) and death. We derived the age-adjusted measures NfL Z scores and conducted mixed-effects modelling to test associations between NfL Z scores and other variables, encompassing clinical outcomes. Summary receiver operating characteristic curves (SROCs) were used to calculate the area under the curve (AUC) for blood NfL.
RESULTS: We identified 382 records, of which 7 studies were included with a total of 669 hospitalized COVID-19 cases (mean age 66.2 ± 15.0 years, 68.1% males). Median NfL Z score at admission was elevated compared to the age-corrected reference population (2.37, IQR: 1.13-3.06, referring to 99th percentile in healthy controls). NfL Z scores were significantly associated with disease duration and severity. Higher NfL Z scores were associated with a higher likelihood of ICU admission, need of MV, and death. SROCs revealed AUCs of 0.74, 0.80 and 0.71 for mortality, need of MV and ICU admission, respectively.
CONCLUSIONS: Blood NfL levels were elevated in the acute phase of COVID-19 patients without major CNS manifestations and associated with clinical severity and poor outcome. The marker might ameliorate the performance of prognostic multivariable algorithms in COVID-19.
Additional Links: PMID-37184659
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37184659,
year = {2023},
author = {Abdelhak, A and Barba, L and Romoli, M and Benkert, P and Conversi, F and D'Anna, L and Masvekar, RR and Bielekova, B and Prudencio, M and Petrucelli, L and Meschia, JF and Erben, Y and Furlan, R and De Lorenzo, R and Mandelli, A and Sutter, R and Hert, L and Epple, V and Marastoni, D and Sellner, J and Steinacker, P and Aamodt, AH and Heggelund, L and Dyrhol-Riise, AM and Virhammar, J and Fällmar, D and Rostami, E and Kumlien, E and Blennow, K and Zetterberg, H and Tumani, H and Sacco, S and Green, AJ and Otto, M and Kuhle, J and Ornello, R and Foschi, M and Abu-Rumeileh, S},
title = {Prognostic performance of blood neurofilament light chain protein in hospitalized COVID-19 patients without major central nervous system manifestations: an individual participant data meta-analysis.},
journal = {Journal of neurology},
volume = {270},
number = {7},
pages = {3315-3328},
pmid = {37184659},
issn = {1432-1459},
support = {R35 NS097273/NS/NINDS NIH HHS/United States ; },
mesh = {Adult ; Male ; Humans ; Middle Aged ; Aged ; Aged, 80 and over ; Female ; Prognosis ; *COVID-19 ; Biomarkers ; Intermediate Filaments ; Central Nervous System ; Neurofilament Proteins ; },
abstract = {BACKGROUND AND AIMS: To investigate the prognostic value of blood neurofilament light chain protein (NfL) levels in the acute phase of coronavirus disease 2019 (COVID-19).
METHODS: We conducted an individual participant data (IPD) meta-analysis after screening on MEDLINE and Scopus to May 23rd 2022. We included studies with hospitalized adult COVID-19 patients without major COVID-19-associated central nervous system (CNS) manifestations and with a measurement of blood NfL in the acute phase as well as data regarding at least one clinical outcome including intensive care unit (ICU) admission, need of mechanical ventilation (MV) and death. We derived the age-adjusted measures NfL Z scores and conducted mixed-effects modelling to test associations between NfL Z scores and other variables, encompassing clinical outcomes. Summary receiver operating characteristic curves (SROCs) were used to calculate the area under the curve (AUC) for blood NfL.
RESULTS: We identified 382 records, of which 7 studies were included with a total of 669 hospitalized COVID-19 cases (mean age 66.2 ± 15.0 years, 68.1% males). Median NfL Z score at admission was elevated compared to the age-corrected reference population (2.37, IQR: 1.13-3.06, referring to 99th percentile in healthy controls). NfL Z scores were significantly associated with disease duration and severity. Higher NfL Z scores were associated with a higher likelihood of ICU admission, need of MV, and death. SROCs revealed AUCs of 0.74, 0.80 and 0.71 for mortality, need of MV and ICU admission, respectively.
CONCLUSIONS: Blood NfL levels were elevated in the acute phase of COVID-19 patients without major CNS manifestations and associated with clinical severity and poor outcome. The marker might ameliorate the performance of prognostic multivariable algorithms in COVID-19.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Male
Humans
Middle Aged
Aged
Aged, 80 and over
Female
Prognosis
*COVID-19
Biomarkers
Intermediate Filaments
Central Nervous System
Neurofilament Proteins
RevDate: 2024-11-02
CmpDate: 2023-11-16
Meta-analysis of age-related cognitive decline reveals a novel locus for the attention domain and implicates a COVID-19-related gene for global cognitive function.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 19(11):5010-5022.
INTRODUCTION: Cognitive abilities have substantial heritability throughout life, as shown by twin- and population-based studies. However, there is limited understanding of the genetic factors related to cognitive decline in aging across neurocognitive domains.
METHODS: We conducted a meta-analysis on 3045 individuals aged ≥65, derived from three population-based cohorts, to identify genetic variants associated with the decline of five neurocognitive domains (attention, memory, executive function, language, visuospatial function) and global cognitive decline. We also conducted gene-based and functional bioinformatics analyses.
RESULTS: Apolipoprotein E (APOE)4 was significantly associated with decline of memory (p = 5.58E-09) and global cognitive function (p = 1.84E-08). We identified a novel association with attention decline on chromosome 9, rs6559700 (p = 2.69E-08), near RASEF. Gene-based analysis also identified a novel gene, TMPRSS11D, involved in the activation of SARS-CoV-2, to be associated with the decline in global cognitive function (p = 4.28E-07).
DISCUSSION: Domain-specific genetic studies can aid in the identification of novel genes and pathways associated with decline across neurocognitive domains.
HIGHLIGHTS: rs6559700 was associated with decline of attention. APOE4 was associated with decline of memory and global cognitive decline. TMPRSS11D, a gene involved in the activation of SARS-CoV-2, was implicated in global cognitive decline. Cognitive domain abilities had both unique and shared molecular pathways across the domains.
Additional Links: PMID-37089073
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37089073,
year = {2023},
author = {Acharya, V and Fan, KH and Snitz, BE and Ganguli, M and DeKosky, ST and Lopez, OL and Feingold, E and Kamboh, MI},
title = {Meta-analysis of age-related cognitive decline reveals a novel locus for the attention domain and implicates a COVID-19-related gene for global cognitive function.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {19},
number = {11},
pages = {5010-5022},
pmid = {37089073},
issn = {1552-5279},
support = {R01 AG064877/AG/NIA NIH HHS/United States ; R37 AG023651/AG/NIA NIH HHS/United States ; AG007562/GF/NIH HHS/United States ; AG023651/GF/NIH HHS/United States ; R56 AG064877/AG/NIA NIH HHS/United States ; P30 AG066468/AG/NIA NIH HHS/United States ; R01 AG041718/AG/NIA NIH HHS/United States ; AG041718/GF/NIH HHS/United States ; U01 AT000162/GF/NIH HHS/United States ; U01 AT000162/AT/NCCIH NIH HHS/United States ; U24 AG021886/AG/NIA NIH HHS/United States ; AG030653/GF/NIH HHS/United States ; R01 AG023651/AG/NIA NIH HHS/United States ; R01 AG030653/AG/NIA NIH HHS/United States ; AG064877/GF/NIH HHS/United States ; },
mesh = {Humans ; *COVID-19/genetics ; SARS-CoV-2 ; Cognition/physiology ; *Cognitive Dysfunction/genetics ; Attention ; Apolipoprotein E4/genetics ; Neuropsychological Tests ; },
abstract = {INTRODUCTION: Cognitive abilities have substantial heritability throughout life, as shown by twin- and population-based studies. However, there is limited understanding of the genetic factors related to cognitive decline in aging across neurocognitive domains.
METHODS: We conducted a meta-analysis on 3045 individuals aged ≥65, derived from three population-based cohorts, to identify genetic variants associated with the decline of five neurocognitive domains (attention, memory, executive function, language, visuospatial function) and global cognitive decline. We also conducted gene-based and functional bioinformatics analyses.
RESULTS: Apolipoprotein E (APOE)4 was significantly associated with decline of memory (p = 5.58E-09) and global cognitive function (p = 1.84E-08). We identified a novel association with attention decline on chromosome 9, rs6559700 (p = 2.69E-08), near RASEF. Gene-based analysis also identified a novel gene, TMPRSS11D, involved in the activation of SARS-CoV-2, to be associated with the decline in global cognitive function (p = 4.28E-07).
DISCUSSION: Domain-specific genetic studies can aid in the identification of novel genes and pathways associated with decline across neurocognitive domains.
HIGHLIGHTS: rs6559700 was associated with decline of attention. APOE4 was associated with decline of memory and global cognitive decline. TMPRSS11D, a gene involved in the activation of SARS-CoV-2, was implicated in global cognitive decline. Cognitive domain abilities had both unique and shared molecular pathways across the domains.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/genetics
SARS-CoV-2
Cognition/physiology
*Cognitive Dysfunction/genetics
Attention
Apolipoprotein E4/genetics
Neuropsychological Tests
RevDate: 2023-06-14
CmpDate: 2023-05-02
Home-based exercise improves quality of life in breast and prostate cancer survivors: A meta-analysis.
PloS one, 18(4):e0284427.
BACKGROUND: Breast (BCa) and prostate (PCa) cancer are two of the most common but survivable cancers. One important component of survivorship that is impacted by treatment long term is diminished quality of life (QoL). Supervised exercise improves QoL and subsequent outcomes but is not accessible for all survivors. Additionally, many factors influence QoL including physical activity (PA), cardiorespiratory fitness (CRF), physical function, and fatigue. However, the COVID-19 pandemic has highlighted the need to increase access to exercise beyond supervised exercise facilities. Home-based exercise may provide a feasible alternative for cancer survivors especially for those living in rural communities.
OBJECTIVES: The primary aim is to investigate the effects of home-based exercise training (Pre-training vs. Post-training) on QoL in BCa/PCa. A secondary aim is to investigate PA, CRF, physical function, and fatigue and potential moderators (age, cancer-type, intervention duration and type). Home-based exercise trials (randomized crossover or quasi-experimental design) with adults (aged 18 years and over) breast or prostate cancer survivors (not currently undergoing chemotherapy or radiation treatment) were eligible for inclusion.
DATA SOURCES: Electronic databases were searched (inception-December 2022) for studies which included adult BCa or PCa survivors (not currently on chemotherapy/radiation), at least measured QoL, and undergoing unsupervised, home-based exercise training.
Initially, 819 studies were identified, from which 17 studies (20 effects) involving 692 participants were extracted. Effect sizes were calculated as standardized mean differences (SMD). Data were pooled using a 3-level model with restricted maximum likelihood estimation. Pooled SMD was used to assess the magnitude of effect, where <0.2, 0.2, 0.5, and 0.8 was defined as trivial, small, moderate, and large respectively.
RESULTS: Home-based exercise resulted in small improvements in QoL (SMD = 0.30, 95% CI 0.01, 0.60, p = 0.042), PA (SMD = 0.49, 95% CI 0.26, 0.75, p<0.001) and CRF (SMD = 0.45, 95% CI -0.01, 0.91, p = 0.056). Physical function (SMD = 0.00, 95% CI -0.21, 0.21, p = 1.000) and fatigue (SMD = -0.61, 95%CI -1.53, 0.32, p = 0.198) did not change.
CONCLUSIONS: Home-based exercise results in small improves QoL in BCa/PCa survivors, independent of cancer type, intervention duration and type, or age. Home-based exercise also improves PA and CRF enhancing survivorship. Therefore, home-based exercise is an efficacious alternative option to improve QoL for BCa and PCa survivors especially for those who live in rural communities or lack access to exercise facilities.
Additional Links: PMID-37079635
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid37079635,
year = {2023},
author = {Bates-Fraser, LC and Riley, S and Stopforth, C and Moertl, K and Edgar, K and Stoner, L and Hanson, ED},
title = {Home-based exercise improves quality of life in breast and prostate cancer survivors: A meta-analysis.},
journal = {PloS one},
volume = {18},
number = {4},
pages = {e0284427},
pmid = {37079635},
issn = {1932-6203},
support = {T32 CA116339/CA/NCI NIH HHS/United States ; },
mesh = {Adolescent ; Adult ; Humans ; Male ; *Cancer Survivors ; Exercise/physiology ; *Fatigue/etiology/physiopathology/therapy ; *Prostatic Neoplasms/complications/physiopathology/therapy ; Quality of Life ; *Breast Neoplasms/complications/physiopathology/therapy ; Female ; *Physical Fitness/physiology ; Cardiorespiratory Fitness/physiology ; Functional Status ; *Self Care/methods ; },
abstract = {BACKGROUND: Breast (BCa) and prostate (PCa) cancer are two of the most common but survivable cancers. One important component of survivorship that is impacted by treatment long term is diminished quality of life (QoL). Supervised exercise improves QoL and subsequent outcomes but is not accessible for all survivors. Additionally, many factors influence QoL including physical activity (PA), cardiorespiratory fitness (CRF), physical function, and fatigue. However, the COVID-19 pandemic has highlighted the need to increase access to exercise beyond supervised exercise facilities. Home-based exercise may provide a feasible alternative for cancer survivors especially for those living in rural communities.
OBJECTIVES: The primary aim is to investigate the effects of home-based exercise training (Pre-training vs. Post-training) on QoL in BCa/PCa. A secondary aim is to investigate PA, CRF, physical function, and fatigue and potential moderators (age, cancer-type, intervention duration and type). Home-based exercise trials (randomized crossover or quasi-experimental design) with adults (aged 18 years and over) breast or prostate cancer survivors (not currently undergoing chemotherapy or radiation treatment) were eligible for inclusion.
DATA SOURCES: Electronic databases were searched (inception-December 2022) for studies which included adult BCa or PCa survivors (not currently on chemotherapy/radiation), at least measured QoL, and undergoing unsupervised, home-based exercise training.
Initially, 819 studies were identified, from which 17 studies (20 effects) involving 692 participants were extracted. Effect sizes were calculated as standardized mean differences (SMD). Data were pooled using a 3-level model with restricted maximum likelihood estimation. Pooled SMD was used to assess the magnitude of effect, where <0.2, 0.2, 0.5, and 0.8 was defined as trivial, small, moderate, and large respectively.
RESULTS: Home-based exercise resulted in small improvements in QoL (SMD = 0.30, 95% CI 0.01, 0.60, p = 0.042), PA (SMD = 0.49, 95% CI 0.26, 0.75, p<0.001) and CRF (SMD = 0.45, 95% CI -0.01, 0.91, p = 0.056). Physical function (SMD = 0.00, 95% CI -0.21, 0.21, p = 1.000) and fatigue (SMD = -0.61, 95%CI -1.53, 0.32, p = 0.198) did not change.
CONCLUSIONS: Home-based exercise results in small improves QoL in BCa/PCa survivors, independent of cancer type, intervention duration and type, or age. Home-based exercise also improves PA and CRF enhancing survivorship. Therefore, home-based exercise is an efficacious alternative option to improve QoL for BCa and PCa survivors especially for those who live in rural communities or lack access to exercise facilities.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adolescent
Adult
Humans
Male
*Cancer Survivors
Exercise/physiology
*Fatigue/etiology/physiopathology/therapy
*Prostatic Neoplasms/complications/physiopathology/therapy
Quality of Life
*Breast Neoplasms/complications/physiopathology/therapy
Female
*Physical Fitness/physiology
Cardiorespiratory Fitness/physiology
Functional Status
*Self Care/methods
RevDate: 2024-05-11
CmpDate: 2023-03-31
Burden of Influenza and Respiratory Syncytial Viruses in Suspected COVID-19 Patients: A Cross-Sectional and Meta-Analysis Study.
Viruses, 15(3):.
Non-SARS-CoV-2 respiratory viral infections, such as influenza virus (FluV) and human respiratory syncytial virus (RSV), have contributed considerably to the burden of infectious diseases in the non-COVID-19 era. While the rates of co-infection in SARS-CoV-2-positive group (SCPG) patients have been determined, the burden of other respiratory viruses in the SARS-CoV-2-negative group (SCNG) remains unclear. Here, we conducted a cross-sectional study (São José do Rio Preto county, Brazil), and we collected our data using a meta-analysis to evaluate the pooled prevalence of FluV and RSV among SCNG patients. Out of the 901 patients suspected of COVID-19, our molecular results showed positivity of FluV and RSV in the SCNG was 2% (15/733) and 0.27% (2/733), respectively. Co-infection with SARS-CoV-2 and FluV, or RSV, was identified in 1.7% of the patients (3/168). Following our meta-analysis, 28 studies were selected (n = 114,318 suspected COVID-19 patients), with a pooled prevalence of 4% (95% CI: 3-6) for FluV and 2% (95% CI: 1-3) for RSV among SCNG patients were observed. Interestingly, FluV positivity in the SCNG was four times higher (OR = 4, 95% CI: 3.6-5.4, p < 0.01) than in the SCPG. Similarly, RSV positivity was significantly associated with SCNG patients (OR = 2.9, 95% CI: 2-4, p < 0.01). For subgroup analysis, cold-like symptoms, including fever, cough, sore throat, headache, myalgia, diarrhea, and nausea/vomiting, were positively associated (p < 0.05) with the SCPG. In conclusion, these results show that the pooled prevalence of FluV and RSV were significantly higher in the SCNG than in the SCPG during the early phase of the COVID-19 pandemic.
Additional Links: PMID-36992374
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36992374,
year = {2023},
author = {Costa, VGD and Gomes, AJC and Bittar, C and Geraldini, DB and Previdelli da Conceição, PJ and Cabral, ÁS and Carvalho, T and Biselli, JM and Provazzi, PJS and Campos, GRF and Sanches, PRDS and Costa, PI and Nogueira, ML and Araujo, JP and Spilki, FR and Calmon, MF and Rahal, P},
title = {Burden of Influenza and Respiratory Syncytial Viruses in Suspected COVID-19 Patients: A Cross-Sectional and Meta-Analysis Study.},
journal = {Viruses},
volume = {15},
number = {3},
pages = {},
pmid = {36992374},
issn = {1999-4915},
support = {U01 AI151807/AI/NIAID NIH HHS/United States ; },
mesh = {Humans ; *Coinfection/epidemiology ; *COVID-19/epidemiology ; Cross-Sectional Studies ; *Influenza, Human/epidemiology ; Pandemics ; *Respiratory Syncytial Virus Infections/epidemiology ; Respiratory Syncytial Virus, Human ; SARS-CoV-2 ; },
abstract = {Non-SARS-CoV-2 respiratory viral infections, such as influenza virus (FluV) and human respiratory syncytial virus (RSV), have contributed considerably to the burden of infectious diseases in the non-COVID-19 era. While the rates of co-infection in SARS-CoV-2-positive group (SCPG) patients have been determined, the burden of other respiratory viruses in the SARS-CoV-2-negative group (SCNG) remains unclear. Here, we conducted a cross-sectional study (São José do Rio Preto county, Brazil), and we collected our data using a meta-analysis to evaluate the pooled prevalence of FluV and RSV among SCNG patients. Out of the 901 patients suspected of COVID-19, our molecular results showed positivity of FluV and RSV in the SCNG was 2% (15/733) and 0.27% (2/733), respectively. Co-infection with SARS-CoV-2 and FluV, or RSV, was identified in 1.7% of the patients (3/168). Following our meta-analysis, 28 studies were selected (n = 114,318 suspected COVID-19 patients), with a pooled prevalence of 4% (95% CI: 3-6) for FluV and 2% (95% CI: 1-3) for RSV among SCNG patients were observed. Interestingly, FluV positivity in the SCNG was four times higher (OR = 4, 95% CI: 3.6-5.4, p < 0.01) than in the SCPG. Similarly, RSV positivity was significantly associated with SCNG patients (OR = 2.9, 95% CI: 2-4, p < 0.01). For subgroup analysis, cold-like symptoms, including fever, cough, sore throat, headache, myalgia, diarrhea, and nausea/vomiting, were positively associated (p < 0.05) with the SCPG. In conclusion, these results show that the pooled prevalence of FluV and RSV were significantly higher in the SCNG than in the SCPG during the early phase of the COVID-19 pandemic.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Coinfection/epidemiology
*COVID-19/epidemiology
Cross-Sectional Studies
*Influenza, Human/epidemiology
Pandemics
*Respiratory Syncytial Virus Infections/epidemiology
Respiratory Syncytial Virus, Human
SARS-CoV-2
RevDate: 2025-10-08
CmpDate: 2023-06-19
Coronavirus Disease 2019 Convalescent Plasma Outpatient Therapy to Prevent Outpatient Hospitalization: A Meta-Analysis of Individual Participant Data From 5 Randomized Trials.
Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, 76(12):2077-2086.
BACKGROUND: Outpatient monoclonal antibodies are no longer effective and antiviral treatments for coronavirus disease 2019 (COVID-19) disease remain largely unavailable in many countries worldwide. Although treatment with COVID-19 convalescent plasma (CCP) is promising, clinical trials among outpatients have shown mixed results.
METHODS: We conducted an individual participant data meta-analysis from outpatient trials to assess the overall risk reduction for all-cause hospitalizations by day 28 in transfused participants. Relevant trials were identified by searching Medline, Embase, medRxiv, World Health Organization COVID-19 Research Database, Cochrane Library, and Web of Science from January 2020 to September 2022.
RESULTS: Five included studies from 4 countries enrolled and transfused 2620 adult patients. Comorbidities were present in 1795 (69%). The virus neutralizing antibody dilutional titer levels ranged from 8 to 14 580 in diverse assays. One hundred sixty of 1315 (12.2%) control patients were hospitalized, versus 111 of 1305 (8.5%) CCP-treated patients, yielding a 3.7% (95% confidence interval [CI], 1.3%-6.0%; P = .001) absolute risk reduction and 30.1% relative risk reduction for all-cause hospitalization. The hospitalization reduction was greatest in those with both early transfusion and high titer with a 7.6% absolute risk reduction (95% CI, 4.0%-11.1%; P = .0001) accompanied by at 51.4% relative risk reduction. No significant reduction in hospitalization was seen with treatment >5 days after symptom onset or in those receiving CCP with antibody titers below the median titer.
CONCLUSIONS: Among outpatients with COVID-19, treatment with CCP reduced the rate of all-cause hospitalization and may be most effective when given within 5 days of symptom onset and when antibody titer is higher.
Additional Links: PMID-36809473
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36809473,
year = {2023},
author = {Levine, AC and Fukuta, Y and Huaman, MA and Ou, J and Meisenberg, BR and Patel, B and Paxton, JH and Hanley, DF and Rijnders, BJA and Gharbharan, A and Rokx, C and Zwaginga, JJ and Alemany, A and Mitjà , O and Ouchi, D and Millat-Martinez, P and Durkalski-Mauldin, V and Korley, FK and Dumont, LJ and Callaway, CW and Libster, R and Marc, GP and Wappner, D and Esteban, I and Polack, F and Sullivan, DJ},
title = {Coronavirus Disease 2019 Convalescent Plasma Outpatient Therapy to Prevent Outpatient Hospitalization: A Meta-Analysis of Individual Participant Data From 5 Randomized Trials.},
journal = {Clinical infectious diseases : an official publication of the Infectious Diseases Society of America},
volume = {76},
number = {12},
pages = {2077-2086},
pmid = {36809473},
issn = {1537-6591},
support = {R01 AI152078/AI/NIAID NIH HHS/United States ; U24 NS100659/NS/NINDS NIH HHS/United States ; 75A50120C00094/NS/NINDS NIH HHS/United States ; U24 NS100655/NS/NINDS NIH HHS/United States ; U24 TR001609/TR/NCATS NIH HHS/United States ; 1OT2HL156812-01/GATES/Gates Foundation/United States ; OT2 HL156812/HL/NHLBI NIH HHS/United States ; },
mesh = {Adult ; Humans ; *COVID-19/therapy ; Outpatients ; SARS-CoV-2 ; COVID-19 Serotherapy ; Randomized Controlled Trials as Topic ; Hospitalization ; },
abstract = {BACKGROUND: Outpatient monoclonal antibodies are no longer effective and antiviral treatments for coronavirus disease 2019 (COVID-19) disease remain largely unavailable in many countries worldwide. Although treatment with COVID-19 convalescent plasma (CCP) is promising, clinical trials among outpatients have shown mixed results.
METHODS: We conducted an individual participant data meta-analysis from outpatient trials to assess the overall risk reduction for all-cause hospitalizations by day 28 in transfused participants. Relevant trials were identified by searching Medline, Embase, medRxiv, World Health Organization COVID-19 Research Database, Cochrane Library, and Web of Science from January 2020 to September 2022.
RESULTS: Five included studies from 4 countries enrolled and transfused 2620 adult patients. Comorbidities were present in 1795 (69%). The virus neutralizing antibody dilutional titer levels ranged from 8 to 14 580 in diverse assays. One hundred sixty of 1315 (12.2%) control patients were hospitalized, versus 111 of 1305 (8.5%) CCP-treated patients, yielding a 3.7% (95% confidence interval [CI], 1.3%-6.0%; P = .001) absolute risk reduction and 30.1% relative risk reduction for all-cause hospitalization. The hospitalization reduction was greatest in those with both early transfusion and high titer with a 7.6% absolute risk reduction (95% CI, 4.0%-11.1%; P = .0001) accompanied by at 51.4% relative risk reduction. No significant reduction in hospitalization was seen with treatment >5 days after symptom onset or in those receiving CCP with antibody titers below the median titer.
CONCLUSIONS: Among outpatients with COVID-19, treatment with CCP reduced the rate of all-cause hospitalization and may be most effective when given within 5 days of symptom onset and when antibody titer is higher.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Humans
*COVID-19/therapy
Outpatients
SARS-CoV-2
COVID-19 Serotherapy
Randomized Controlled Trials as Topic
Hospitalization
RevDate: 2026-01-15
CmpDate: 2023-04-28
American Academy of Nursing consensus recommendations to advance system level change for nurse well-being.
Nursing outlook, 71(2):101917.
The COVID-19 pandemic has required close examination of workforce-related stressors that over decades have contributed to widespread burnout, negative health outcomes, including mental health outcomes, and the loss of the well-educated professionals who are the future of the nursing profession. In the United States and globally, evidence points to factors known to diminish well-being, including inequities, issues of minority status, persistent discrimination, and demanding work environments. The American Academy of Nursing (AAN), dedicated to organizational excellence, nursing leadership and evidence-based policy, develops statements reflecting its mission and those of its nursing affiliates and corporate member, The American Nurses Association. Within nursing, despite the efforts of its members toward advancement, professional fulfillment is often constrained by the systems in which nurses practice and workplace factors over which they have little control. Action by key organizations to initiate changes at systems levels in workplace safety, to increase professional mobility, and propel policies that increase access to health care resources could improve nurse well-being. This paper proposes recommendations from the AAN Expert Panels on Building Health Care System Excellence, Psychiatric Mental Health and Substance Use, and Global Health Expert Panels for the American Academy of Nursing to leverage related policy in the arenas of government and professional/healthcare organizations. Transforming health care work environments and advancing nurse well-being and equity can be accomplished through key, innovative policy changes. These will be achieved through collaboration among associations, organizations, nonprofit groups, and with the public and the media.
Additional Links: PMID-36736029
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36736029,
year = {2023},
author = {Naegle, MA and Kelly, LA and Embree, JL and Valentine, N and Sharp, D and Grinspun, D and Hines-Martin, VP and Crawford, CL and Rosa, WE},
title = {American Academy of Nursing consensus recommendations to advance system level change for nurse well-being.},
journal = {Nursing outlook},
volume = {71},
number = {2},
pages = {101917},
pmid = {36736029},
issn = {1528-3968},
support = {P30 CA008748/CA/NCI NIH HHS/United States ; P30 ES030283/ES/NIEHS NIH HHS/United States ; },
mesh = {Humans ; United States ; *Pandemics ; *COVID-19/epidemiology ; Delivery of Health Care ; Workplace ; Leadership ; },
abstract = {The COVID-19 pandemic has required close examination of workforce-related stressors that over decades have contributed to widespread burnout, negative health outcomes, including mental health outcomes, and the loss of the well-educated professionals who are the future of the nursing profession. In the United States and globally, evidence points to factors known to diminish well-being, including inequities, issues of minority status, persistent discrimination, and demanding work environments. The American Academy of Nursing (AAN), dedicated to organizational excellence, nursing leadership and evidence-based policy, develops statements reflecting its mission and those of its nursing affiliates and corporate member, The American Nurses Association. Within nursing, despite the efforts of its members toward advancement, professional fulfillment is often constrained by the systems in which nurses practice and workplace factors over which they have little control. Action by key organizations to initiate changes at systems levels in workplace safety, to increase professional mobility, and propel policies that increase access to health care resources could improve nurse well-being. This paper proposes recommendations from the AAN Expert Panels on Building Health Care System Excellence, Psychiatric Mental Health and Substance Use, and Global Health Expert Panels for the American Academy of Nursing to leverage related policy in the arenas of government and professional/healthcare organizations. Transforming health care work environments and advancing nurse well-being and equity can be accomplished through key, innovative policy changes. These will be achieved through collaboration among associations, organizations, nonprofit groups, and with the public and the media.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
United States
*Pandemics
*COVID-19/epidemiology
Delivery of Health Care
Workplace
Leadership
RevDate: 2025-05-30
CmpDate: 2023-01-18
Adverse maternal, fetal, and newborn outcomes among pregnant women with SARS-CoV-2 infection: an individual participant data meta-analysis.
BMJ global health, 8(1):.
INTRODUCTION: Despite a growing body of research on the risks of SARS-CoV-2 infection during pregnancy, there is continued controversy given heterogeneity in the quality and design of published studies.
METHODS: We screened ongoing studies in our sequential, prospective meta-analysis. We pooled individual participant data to estimate the absolute and relative risk (RR) of adverse outcomes among pregnant women with SARS-CoV-2 infection, compared with confirmed negative pregnancies. We evaluated the risk of bias using a modified Newcastle-Ottawa Scale.
RESULTS: We screened 137 studies and included 12 studies in 12 countries involving 13 136 pregnant women.Pregnant women with SARS-CoV-2 infection-as compared with uninfected pregnant women-were at significantly increased risk of maternal mortality (10 studies; n=1490; RR 7.68, 95% CI 1.70 to 34.61); admission to intensive care unit (8 studies; n=6660; RR 3.81, 95% CI 2.03 to 7.17); receiving mechanical ventilation (7 studies; n=4887; RR 15.23, 95% CI 4.32 to 53.71); receiving any critical care (7 studies; n=4735; RR 5.48, 95% CI 2.57 to 11.72); and being diagnosed with pneumonia (6 studies; n=4573; RR 23.46, 95% CI 3.03 to 181.39) and thromboembolic disease (8 studies; n=5146; RR 5.50, 95% CI 1.12 to 27.12).Neonates born to women with SARS-CoV-2 infection were more likely to be admitted to a neonatal care unit after birth (7 studies; n=7637; RR 1.86, 95% CI 1.12 to 3.08); be born preterm (7 studies; n=6233; RR 1.71, 95% CI 1.28 to 2.29) or moderately preterm (7 studies; n=6071; RR 2.92, 95% CI 1.88 to 4.54); and to be born low birth weight (12 studies; n=11 930; RR 1.19, 95% CI 1.02 to 1.40). Infection was not linked to stillbirth. Studies were generally at low or moderate risk of bias.
CONCLUSIONS: This analysis indicates that SARS-CoV-2 infection at any time during pregnancy increases the risk of maternal death, severe maternal morbidities and neonatal morbidity, but not stillbirth or intrauterine growth restriction. As more data become available, we will update these findings per the published protocol.
Additional Links: PMID-36646475
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36646475,
year = {2023},
author = {Smith, ER and Oakley, E and Grandner, GW and Ferguson, K and Farooq, F and Afshar, Y and Ahlberg, M and Ahmadzia, H and Akelo, V and Aldrovandi, G and Tippett Barr, BA and Bevilacqua, E and Brandt, JS and Broutet, N and Fernández Buhigas, I and Carrillo, J and Clifton, R and Conry, J and Cosmi, E and Crispi, F and Crovetto, F and Delgado-López, C and Divakar, H and Driscoll, AJ and Favre, G and Flaherman, VJ and Gale, C and Gil, MM and Gottlieb, SL and Gratacós, E and Hernandez, O and Jones, S and Kalafat, E and Khagayi, S and Knight, M and Kotloff, K and Lanzone, A and Le Doare, K and Lees, C and Litman, E and Lokken, EM and Laurita Longo, V and Madhi, SA and Magee, LA and Martinez-Portilla, RJ and McClure, EM and Metz, TD and Miller, ES and Money, D and Moungmaithong, S and Mullins, E and Nachega, JB and Nunes, MC and Onyango, D and Panchaud, A and Poon, LC and Raiten, D and Regan, L and Rukundo, G and Sahota, D and Sakowicz, A and Sanin-Blair, J and Söderling, J and Stephansson, O and Temmerman, M and Thorson, A and Tolosa, JE and Townson, J and Valencia-Prado, M and Visentin, S and von Dadelszen, P and Adams Waldorf, K and Whitehead, C and Yassa, M and Tielsch, JM and , and , },
title = {Adverse maternal, fetal, and newborn outcomes among pregnant women with SARS-CoV-2 infection: an individual participant data meta-analysis.},
journal = {BMJ global health},
volume = {8},
number = {1},
pages = {},
pmid = {36646475},
issn = {2059-7908},
support = {T32 AI007524/AI/NIAID NIH HHS/United States ; 001/WHO_/World Health Organization/International ; U01 HD036801/HD/NICHD NIH HHS/United States ; U10 HD036801/HD/NICHD NIH HHS/United States ; U24 HD036801/HD/NICHD NIH HHS/United States ; },
mesh = {Infant, Newborn ; Pregnancy ; Female ; Humans ; *Pregnant People ; Prospective Studies ; *COVID-19 ; SARS-CoV-2 ; },
abstract = {INTRODUCTION: Despite a growing body of research on the risks of SARS-CoV-2 infection during pregnancy, there is continued controversy given heterogeneity in the quality and design of published studies.
METHODS: We screened ongoing studies in our sequential, prospective meta-analysis. We pooled individual participant data to estimate the absolute and relative risk (RR) of adverse outcomes among pregnant women with SARS-CoV-2 infection, compared with confirmed negative pregnancies. We evaluated the risk of bias using a modified Newcastle-Ottawa Scale.
RESULTS: We screened 137 studies and included 12 studies in 12 countries involving 13 136 pregnant women.Pregnant women with SARS-CoV-2 infection-as compared with uninfected pregnant women-were at significantly increased risk of maternal mortality (10 studies; n=1490; RR 7.68, 95% CI 1.70 to 34.61); admission to intensive care unit (8 studies; n=6660; RR 3.81, 95% CI 2.03 to 7.17); receiving mechanical ventilation (7 studies; n=4887; RR 15.23, 95% CI 4.32 to 53.71); receiving any critical care (7 studies; n=4735; RR 5.48, 95% CI 2.57 to 11.72); and being diagnosed with pneumonia (6 studies; n=4573; RR 23.46, 95% CI 3.03 to 181.39) and thromboembolic disease (8 studies; n=5146; RR 5.50, 95% CI 1.12 to 27.12).Neonates born to women with SARS-CoV-2 infection were more likely to be admitted to a neonatal care unit after birth (7 studies; n=7637; RR 1.86, 95% CI 1.12 to 3.08); be born preterm (7 studies; n=6233; RR 1.71, 95% CI 1.28 to 2.29) or moderately preterm (7 studies; n=6071; RR 2.92, 95% CI 1.88 to 4.54); and to be born low birth weight (12 studies; n=11 930; RR 1.19, 95% CI 1.02 to 1.40). Infection was not linked to stillbirth. Studies were generally at low or moderate risk of bias.
CONCLUSIONS: This analysis indicates that SARS-CoV-2 infection at any time during pregnancy increases the risk of maternal death, severe maternal morbidities and neonatal morbidity, but not stillbirth or intrauterine growth restriction. As more data become available, we will update these findings per the published protocol.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Infant, Newborn
Pregnancy
Female
Humans
*Pregnant People
Prospective Studies
*COVID-19
SARS-CoV-2
RevDate: 2023-09-14
CmpDate: 2023-03-27
Hydroxychloroquine for treatment of non-hospitalized adults with COVID-19: A meta-analysis of individual participant data of randomized trials.
Clinical and translational science, 16(3):524-535.
Hydroxychloroquine (HCQ) was initially promoted as an oral therapy for early treatment of coronavirus disease 2019 (COVID-19). Conventional meta-analyses cannot fully address the heterogeneity of different designs and outcomes of randomized controlled trials (RCTs) assessing the efficacy of HCQ in outpatients with mild COVID-19. We conducted a pooled analysis of individual participant data from RCTs that evaluated the effect of HCQ on hospitalization and viral load reduction in outpatients with confirmed COVID-19. We evaluated the overall treatment group effect by log-likelihood ratio test (-2LL) from a generalized linear mixed model to accommodate correlated longitudinal binary data. The analysis included data from 11 RCTs. The outcome of virological effect, assessed in 1560 participants (N = 795 HCQ, N = 765 control), did not differ significantly between the two treatment groups (-2LL = 7.66; p = 0.18) when adjusting for cohort, duration of symptoms, and comorbidities. The decline in polymerase chain reaction positive tests from day 1 to 7 was 42.0 and 41.6 percentage points in the HCQ and control groups, respectively. Among the 2037 participants evaluable for hospitalization (N = 1058 HCQ, N = 979 control), we found no significant differences in hospitalization rate between participants receiving HCQ and controls (odds ratio 0.995; 95% confidence interval 0.614-1.610; -2LL = 0.0; p = 0.98) when adjusting for cohort, duration of symptoms, and comorbidities. This individual participant data meta-analysis of 11 HCQ trials that evaluated severe acute respiratory syndrome-coronavirus 2 viral clearance and COVID-19 hospitalization did not show a clinical benefit of HCQ. Our meta-analysis provides evidence to support the interruption in the use of HCQ in mild COVID-19 outpatients to reduce progression to severe disease.
Additional Links: PMID-36601684
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36601684,
year = {2023},
author = {Mitjà , O and Reis, G and Boulware, DR and Spivak, AM and Sarwar, A and Johnston, C and Webb, B and Hill, MD and Smith, D and Kremsner, P and Curran, M and Carter, D and Alexander, J and Corbacho, M and Lee, TC and Hullsiek, KH and McDonald, EG and Hess, R and Hughes, M and Baeten, JM and Schwartz, I and Metz, L and Richer, L and Chew, KW and Daar, E and Wohl, D and Dunne, M},
title = {Hydroxychloroquine for treatment of non-hospitalized adults with COVID-19: A meta-analysis of individual participant data of randomized trials.},
journal = {Clinical and translational science},
volume = {16},
number = {3},
pages = {524-535},
pmid = {36601684},
issn = {1752-8062},
support = {UM1 AI069423/AI/NIAID NIH HHS/United States ; UM1 AI069424/AI/NIAID NIH HHS/United States ; UL1 TR002538/TR/NCATS NIH HHS/United States ; UM1 AI068634/AI/NIAID NIH HHS/United States ; U01 AI069424/AI/NIAID NIH HHS/United States ; UM1 AI068636/AI/NIAID NIH HHS/United States ; },
mesh = {Adult ; Humans ; *COVID-19 ; COVID-19 Drug Treatment ; Hydroxychloroquine ; Randomized Controlled Trials as Topic ; Treatment Outcome ; },
abstract = {Hydroxychloroquine (HCQ) was initially promoted as an oral therapy for early treatment of coronavirus disease 2019 (COVID-19). Conventional meta-analyses cannot fully address the heterogeneity of different designs and outcomes of randomized controlled trials (RCTs) assessing the efficacy of HCQ in outpatients with mild COVID-19. We conducted a pooled analysis of individual participant data from RCTs that evaluated the effect of HCQ on hospitalization and viral load reduction in outpatients with confirmed COVID-19. We evaluated the overall treatment group effect by log-likelihood ratio test (-2LL) from a generalized linear mixed model to accommodate correlated longitudinal binary data. The analysis included data from 11 RCTs. The outcome of virological effect, assessed in 1560 participants (N = 795 HCQ, N = 765 control), did not differ significantly between the two treatment groups (-2LL = 7.66; p = 0.18) when adjusting for cohort, duration of symptoms, and comorbidities. The decline in polymerase chain reaction positive tests from day 1 to 7 was 42.0 and 41.6 percentage points in the HCQ and control groups, respectively. Among the 2037 participants evaluable for hospitalization (N = 1058 HCQ, N = 979 control), we found no significant differences in hospitalization rate between participants receiving HCQ and controls (odds ratio 0.995; 95% confidence interval 0.614-1.610; -2LL = 0.0; p = 0.98) when adjusting for cohort, duration of symptoms, and comorbidities. This individual participant data meta-analysis of 11 HCQ trials that evaluated severe acute respiratory syndrome-coronavirus 2 viral clearance and COVID-19 hospitalization did not show a clinical benefit of HCQ. Our meta-analysis provides evidence to support the interruption in the use of HCQ in mild COVID-19 outpatients to reduce progression to severe disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Humans
*COVID-19
COVID-19 Drug Treatment
Hydroxychloroquine
Randomized Controlled Trials as Topic
Treatment Outcome
RevDate: 2024-06-04
CmpDate: 2022-11-22
Tissue factor links inflammation, thrombosis, and senescence in COVID-19.
Scientific reports, 12(1):19842.
COVID-19 is a highly contagious respiratory infection caused by the SARS-CoV-2 virus. The infected lung epithelial cells secrete a group of chemokines and cytokines, which triggers harmful cytokine storms and hyper-thrombotic responses. Recent studies have proposed that viral-induced senescence is responsible for cytokine release and inflammation in COVID-19 patients. However, it is unknown whether cellular senescence is commonly triggered after viral infection and how inflammation and thrombosis, hyper-activated in these patients, are functionally connected. To address these questions, we conducted a bioinformatics-based meta-analysis using single-cell and bulk RNA sequencing datasets obtained from human patient studies, animal models, and cell lines infected with SARS-CoV-2 and other respiratory viruses. We found that the senescence phenotype is robustly upregulated in most SARS-CoV-2-infected patients, especially in the infected lung epithelial cells. Notably, the upregulation of Tissue factor (F3), a key initiator of the extrinsic blood coagulation pathway, occurs concurrently with the upregulation of the senescence-associated secretory phenotype (SASP) factors. Furthermore, F3 levels are positively correlated with the senescence and hyper-coagulation gene signatures in COVID-19 patients. Together, these data demonstrate the prevalence of senescence in respiratory viral infection and suggest F3 as a critical link between inflammation, thrombosis, and senescence in these disease states.
Additional Links: PMID-36400883
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36400883,
year = {2022},
author = {Nguyen, D and Jeon, HM and Lee, J},
title = {Tissue factor links inflammation, thrombosis, and senescence in COVID-19.},
journal = {Scientific reports},
volume = {12},
number = {1},
pages = {19842},
pmid = {36400883},
issn = {2045-2322},
support = {R01 CA223370/CA/NCI NIH HHS/United States ; },
mesh = {Humans ; Animals ; Thromboplastin/genetics ; *COVID-19 ; SARS-CoV-2 ; Inflammation ; *Thrombosis ; Cytokines/metabolism ; },
abstract = {COVID-19 is a highly contagious respiratory infection caused by the SARS-CoV-2 virus. The infected lung epithelial cells secrete a group of chemokines and cytokines, which triggers harmful cytokine storms and hyper-thrombotic responses. Recent studies have proposed that viral-induced senescence is responsible for cytokine release and inflammation in COVID-19 patients. However, it is unknown whether cellular senescence is commonly triggered after viral infection and how inflammation and thrombosis, hyper-activated in these patients, are functionally connected. To address these questions, we conducted a bioinformatics-based meta-analysis using single-cell and bulk RNA sequencing datasets obtained from human patient studies, animal models, and cell lines infected with SARS-CoV-2 and other respiratory viruses. We found that the senescence phenotype is robustly upregulated in most SARS-CoV-2-infected patients, especially in the infected lung epithelial cells. Notably, the upregulation of Tissue factor (F3), a key initiator of the extrinsic blood coagulation pathway, occurs concurrently with the upregulation of the senescence-associated secretory phenotype (SASP) factors. Furthermore, F3 levels are positively correlated with the senescence and hyper-coagulation gene signatures in COVID-19 patients. Together, these data demonstrate the prevalence of senescence in respiratory viral infection and suggest F3 as a critical link between inflammation, thrombosis, and senescence in these disease states.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Animals
Thromboplastin/genetics
*COVID-19
SARS-CoV-2
Inflammation
*Thrombosis
Cytokines/metabolism
RevDate: 2022-12-17
CmpDate: 2022-11-16
Interim Recommendations from the Advisory Committee on Immunization Practices for the Use of Bivalent Booster Doses of COVID-19 Vaccines - United States, October 2022.
MMWR. Morbidity and mortality weekly report, 71(45):1436-1441.
Four COVID-19 vaccines are currently approved for primary series vaccination in the United States under a Biologics License Application or authorized under an emergency use authorization (EUA) by the Food and Drug Administration (FDA), and recommended for primary series vaccination by the Advisory Committee on Immunization Practices (ACIP): 1) the 2- or 3-dose monovalent mRNA BNT162b2 (Pfizer-BioNTech, Comirnaty) COVID-19 vaccine; 2) the 2- or 3-dose monovalent mRNA mRNA-1273 (Moderna, Spikevax) COVID-19 vaccine; 3) the single-dose adenovirus vector-based Ad26.COV.S (Janssen [Johnson & Johnson]) COVID-19 vaccine; and 4) the 2-dose adjuvanted, protein subunit-based NVX-CoV2373 (Novavax) COVID-19 vaccine. The number of doses recommended is based on recipient age and immunocompromise status (1). For additional protection, FDA has amended EUAs to allow for COVID-19 booster doses in eligible persons (1). Because COVID-19 vaccines have demonstrated decreased effectiveness during the period when the Omicron variant (B.1.1.529) of SARS-CoV-2 predominated, bivalent booster doses (i.e., vaccine with equal components from the ancestral and Omicron strains) were considered for the express purpose of improving protection conferred by COVID-19 vaccine booster doses (2). During September-October 2022, FDA authorized bivalent mRNA vaccines for use as a booster dose in persons aged ≥5 years who completed any FDA-approved or FDA-authorized primary series and removed EUAs for monovalent COVID-19 booster doses (1). Pfizer-BioNTech and Moderna bivalent booster vaccines each contain equal amounts of spike mRNA from the ancestral and Omicron BA.4/BA.5 strains. After the EUA amendments, ACIP and CDC recommended that all persons aged ≥5 years receive 1 bivalent mRNA booster dose ≥2 months after completion of any FDA-approved or FDA-authorized monovalent primary series or monovalent booster doses.
Additional Links: PMID-36355612
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36355612,
year = {2022},
author = {Rosenblum, HG and Wallace, M and Godfrey, M and Roper, LE and Hall, E and Fleming-Dutra, KE and Link-Gelles, R and Pilishvili, T and Williams, J and Moulia, DL and Brooks, O and Talbot, HK and Lee, GM and Bell, BP and Daley, MF and Meyer, S and Oliver, SE and Twentyman, E},
title = {Interim Recommendations from the Advisory Committee on Immunization Practices for the Use of Bivalent Booster Doses of COVID-19 Vaccines - United States, October 2022.},
journal = {MMWR. Morbidity and mortality weekly report},
volume = {71},
number = {45},
pages = {1436-1441},
pmid = {36355612},
issn = {1545-861X},
mesh = {Vaccination ; *COVID-19/prevention & control ; *COVID-19 Vaccines ; United States/epidemiology ; Immunization ; BNT162 Vaccine ; SARS-CoV-2 ; Immunization, Secondary ; RNA, Messenger ; Humans ; Advisory Committees ; },
abstract = {Four COVID-19 vaccines are currently approved for primary series vaccination in the United States under a Biologics License Application or authorized under an emergency use authorization (EUA) by the Food and Drug Administration (FDA), and recommended for primary series vaccination by the Advisory Committee on Immunization Practices (ACIP): 1) the 2- or 3-dose monovalent mRNA BNT162b2 (Pfizer-BioNTech, Comirnaty) COVID-19 vaccine; 2) the 2- or 3-dose monovalent mRNA mRNA-1273 (Moderna, Spikevax) COVID-19 vaccine; 3) the single-dose adenovirus vector-based Ad26.COV.S (Janssen [Johnson & Johnson]) COVID-19 vaccine; and 4) the 2-dose adjuvanted, protein subunit-based NVX-CoV2373 (Novavax) COVID-19 vaccine. The number of doses recommended is based on recipient age and immunocompromise status (1). For additional protection, FDA has amended EUAs to allow for COVID-19 booster doses in eligible persons (1). Because COVID-19 vaccines have demonstrated decreased effectiveness during the period when the Omicron variant (B.1.1.529) of SARS-CoV-2 predominated, bivalent booster doses (i.e., vaccine with equal components from the ancestral and Omicron strains) were considered for the express purpose of improving protection conferred by COVID-19 vaccine booster doses (2). During September-October 2022, FDA authorized bivalent mRNA vaccines for use as a booster dose in persons aged ≥5 years who completed any FDA-approved or FDA-authorized primary series and removed EUAs for monovalent COVID-19 booster doses (1). Pfizer-BioNTech and Moderna bivalent booster vaccines each contain equal amounts of spike mRNA from the ancestral and Omicron BA.4/BA.5 strains. After the EUA amendments, ACIP and CDC recommended that all persons aged ≥5 years receive 1 bivalent mRNA booster dose ≥2 months after completion of any FDA-approved or FDA-authorized monovalent primary series or monovalent booster doses.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Vaccination
*COVID-19/prevention & control
*COVID-19 Vaccines
United States/epidemiology
Immunization
BNT162 Vaccine
SARS-CoV-2
Immunization, Secondary
RNA, Messenger
Humans
Advisory Committees
RevDate: 2024-03-25
CmpDate: 2022-11-14
An Experimental Framework for Developing Point-of-Need Biosensors: Connecting Bio-Layer Interferometry and Electrochemical Impedance Spectroscopy.
Biosensors, 12(11):.
Biolayer interferometry (BLI) is a well-established laboratory technique for studying biomolecular interactions important for applications such as drug development. Currently, there are interesting opportunities for expanding the use of BLI in other fields, including the development of rapid diagnostic tools. To date, there are no detailed frameworks for implementing BLI in target-recognition studies that are pivotal for developing point-of-need biosensors. Here, we attempt to bridge these domains by providing a framework that connects output(s) of molecular interaction studies with key performance indicators used in the development of point-of-need biosensors. First, we briefly review the governing theory for protein-ligand interactions, and we then summarize the approach for real-time kinetic quantification using various techniques. The 2020 PRISMA guideline was used for all governing theory reviews and meta-analyses. Using the information from the meta-analysis, we introduce an experimental framework for connecting outcomes from BLI experiments (KD, kon, koff) with electrochemical (capacitive) biosensor design. As a first step in the development of a larger framework, we specifically focus on mapping BLI outcomes to five biosensor key performance indicators (sensitivity, selectivity, response time, hysteresis, operating range). The applicability of our framework was demonstrated in a study of case based on published literature related to SARS-CoV-2 spike protein to show the development of a capacitive biosensor based on truncated angiotensin-converting enzyme 2 (ACE2) as the receptor. The case study focuses on non-specific binding and selectivity as research goals. The proposed framework proved to be an important first step toward modeling/simulation efforts that map molecular interactions to sensor design.
Additional Links: PMID-36354449
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36354449,
year = {2022},
author = {Ullah, SF and Moreira, G and Datta, SPA and McLamore, E and Vanegas, D},
title = {An Experimental Framework for Developing Point-of-Need Biosensors: Connecting Bio-Layer Interferometry and Electrochemical Impedance Spectroscopy.},
journal = {Biosensors},
volume = {12},
number = {11},
pages = {},
pmid = {36354449},
issn = {2079-6374},
support = {U01 AA029328/AA/NIAAA NIH HHS/United States ; U01AA029328/NH/NIH HHS/United States ; },
mesh = {Humans ; Dielectric Spectroscopy ; SARS-CoV-2 ; *COVID-19/diagnosis ; Interferometry/methods ; *Biosensing Techniques/methods ; },
abstract = {Biolayer interferometry (BLI) is a well-established laboratory technique for studying biomolecular interactions important for applications such as drug development. Currently, there are interesting opportunities for expanding the use of BLI in other fields, including the development of rapid diagnostic tools. To date, there are no detailed frameworks for implementing BLI in target-recognition studies that are pivotal for developing point-of-need biosensors. Here, we attempt to bridge these domains by providing a framework that connects output(s) of molecular interaction studies with key performance indicators used in the development of point-of-need biosensors. First, we briefly review the governing theory for protein-ligand interactions, and we then summarize the approach for real-time kinetic quantification using various techniques. The 2020 PRISMA guideline was used for all governing theory reviews and meta-analyses. Using the information from the meta-analysis, we introduce an experimental framework for connecting outcomes from BLI experiments (KD, kon, koff) with electrochemical (capacitive) biosensor design. As a first step in the development of a larger framework, we specifically focus on mapping BLI outcomes to five biosensor key performance indicators (sensitivity, selectivity, response time, hysteresis, operating range). The applicability of our framework was demonstrated in a study of case based on published literature related to SARS-CoV-2 spike protein to show the development of a capacitive biosensor based on truncated angiotensin-converting enzyme 2 (ACE2) as the receptor. The case study focuses on non-specific binding and selectivity as research goals. The proposed framework proved to be an important first step toward modeling/simulation efforts that map molecular interactions to sensor design.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Dielectric Spectroscopy
SARS-CoV-2
*COVID-19/diagnosis
Interferometry/methods
*Biosensing Techniques/methods
RevDate: 2023-11-29
CmpDate: 2023-01-10
Frequently Asked Questions on Coronavirus Disease 2019 Vaccination for Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T-Cell Recipients From the American Society for Transplantation and Cellular Therapy and the American Society of Hematology.
Transplantation and cellular therapy, 29(1):10-18.
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), disproportionately affects immunocompromised and elderly patients. Not only are hematopoietic cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cell recipients at greater risk for severe COVID-19 and COVID-19-related complications, but they also may experience suboptimal immune responses to currently available COVID-19 vaccines. Optimizing the use, timing, and number of doses of the COVID-19 vaccines in these patients may provide better protection against SARS-CoV-2 infection and better outcomes after infection. To this end, current guidelines for COVID-19 vaccination in HCT and CAR T-cell recipients from the American Society of Transplantation and Cellular Therapy Transplant Infectious Disease Special Interest Group and the American Society of Hematology are provided in a frequently asked questions format.
Additional Links: PMID-36273782
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36273782,
year = {2023},
author = {Khawaja, F and Papanicolaou, G and Dadwal, S and Pergam, SA and Wingard, JR and Boghdadly, ZE and Abidi, MZ and Waghmare, A and Shahid, Z and Michaels, L and Hill, JA and Kamboj, M and Boeckh, M and Auletta, JJ and Chemaly, RF},
title = {Frequently Asked Questions on Coronavirus Disease 2019 Vaccination for Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T-Cell Recipients From the American Society for Transplantation and Cellular Therapy and the American Society of Hematology.},
journal = {Transplantation and cellular therapy},
volume = {29},
number = {1},
pages = {10-18},
pmid = {36273782},
issn = {2666-6367},
support = {K23 AG073534/AG/NIA NIH HHS/United States ; },
mesh = {Aged ; Humans ; COVID-19 Vaccines ; *Receptors, Chimeric Antigen/therapeutic use ; *COVID-19/prevention & control ; SARS-CoV-2 ; T-Lymphocytes ; *Hematopoietic Stem Cell Transplantation ; Vaccination ; *Hematology ; },
abstract = {Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), disproportionately affects immunocompromised and elderly patients. Not only are hematopoietic cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cell recipients at greater risk for severe COVID-19 and COVID-19-related complications, but they also may experience suboptimal immune responses to currently available COVID-19 vaccines. Optimizing the use, timing, and number of doses of the COVID-19 vaccines in these patients may provide better protection against SARS-CoV-2 infection and better outcomes after infection. To this end, current guidelines for COVID-19 vaccination in HCT and CAR T-cell recipients from the American Society of Transplantation and Cellular Therapy Transplant Infectious Disease Special Interest Group and the American Society of Hematology are provided in a frequently asked questions format.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Aged
Humans
COVID-19 Vaccines
*Receptors, Chimeric Antigen/therapeutic use
*COVID-19/prevention & control
SARS-CoV-2
T-Lymphocytes
*Hematopoietic Stem Cell Transplantation
Vaccination
*Hematology
RevDate: 2023-11-28
CmpDate: 2022-10-28
Estimated Global Proportions of Individuals With Persistent Fatigue, Cognitive, and Respiratory Symptom Clusters Following Symptomatic COVID-19 in 2020 and 2021.
JAMA, 328(16):1604-1615.
IMPORTANCE: Some individuals experience persistent symptoms after initial symptomatic SARS-CoV-2 infection (often referred to as Long COVID).
OBJECTIVE: To estimate the proportion of males and females with COVID-19, younger or older than 20 years of age, who had Long COVID symptoms in 2020 and 2021 and their Long COVID symptom duration.
Bayesian meta-regression and pooling of 54 studies and 2 medical record databases with data for 1.2 million individuals (from 22 countries) who had symptomatic SARS-CoV-2 infection. Of the 54 studies, 44 were published and 10 were collaborating cohorts (conducted in Austria, the Faroe Islands, Germany, Iran, Italy, the Netherlands, Russia, Sweden, Switzerland, and the US). The participant data were derived from the 44 published studies (10 501 hospitalized individuals and 42 891 nonhospitalized individuals), the 10 collaborating cohort studies (10 526 and 1906), and the 2 US electronic medical record databases (250 928 and 846 046). Data collection spanned March 2020 to January 2022.
EXPOSURES: Symptomatic SARS-CoV-2 infection.
MAIN OUTCOMES AND MEASURES: Proportion of individuals with at least 1 of the 3 self-reported Long COVID symptom clusters (persistent fatigue with bodily pain or mood swings; cognitive problems; or ongoing respiratory problems) 3 months after SARS-CoV-2 infection in 2020 and 2021, estimated separately for hospitalized and nonhospitalized individuals aged 20 years or older by sex and for both sexes of nonhospitalized individuals younger than 20 years of age.
RESULTS: A total of 1.2 million individuals who had symptomatic SARS-CoV-2 infection were included (mean age, 4-66 years; males, 26%-88%). In the modeled estimates, 6.2% (95% uncertainty interval [UI], 2.4%-13.3%) of individuals who had symptomatic SARS-CoV-2 infection experienced at least 1 of the 3 Long COVID symptom clusters in 2020 and 2021, including 3.2% (95% UI, 0.6%-10.0%) for persistent fatigue with bodily pain or mood swings, 3.7% (95% UI, 0.9%-9.6%) for ongoing respiratory problems, and 2.2% (95% UI, 0.3%-7.6%) for cognitive problems after adjusting for health status before COVID-19, comprising an estimated 51.0% (95% UI, 16.9%-92.4%), 60.4% (95% UI, 18.9%-89.1%), and 35.4% (95% UI, 9.4%-75.1%), respectively, of Long COVID cases. The Long COVID symptom clusters were more common in women aged 20 years or older (10.6% [95% UI, 4.3%-22.2%]) 3 months after symptomatic SARS-CoV-2 infection than in men aged 20 years or older (5.4% [95% UI, 2.2%-11.7%]). Both sexes younger than 20 years of age were estimated to be affected in 2.8% (95% UI, 0.9%-7.0%) of symptomatic SARS-CoV-2 infections. The estimated mean Long COVID symptom cluster duration was 9.0 months (95% UI, 7.0-12.0 months) among hospitalized individuals and 4.0 months (95% UI, 3.6-4.6 months) among nonhospitalized individuals. Among individuals with Long COVID symptoms 3 months after symptomatic SARS-CoV-2 infection, an estimated 15.1% (95% UI, 10.3%-21.1%) continued to experience symptoms at 12 months.
CONCLUSIONS AND RELEVANCE: This study presents modeled estimates of the proportion of individuals with at least 1 of 3 self-reported Long COVID symptom clusters (persistent fatigue with bodily pain or mood swings; cognitive problems; or ongoing respiratory problems) 3 months after symptomatic SARS-CoV-2 infection.
Additional Links: PMID-36215063
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36215063,
year = {2022},
author = {, and Wulf Hanson, S and Abbafati, C and Aerts, JG and Al-Aly, Z and Ashbaugh, C and Ballouz, T and Blyuss, O and Bobkova, P and Bonsel, G and Borzakova, S and Buonsenso, D and Butnaru, D and Carter, A and Chu, H and De Rose, C and Diab, MM and Ekbom, E and El Tantawi, M and Fomin, V and Frithiof, R and Gamirova, A and Glybochko, PV and Haagsma, JA and Haghjooy Javanmard, S and Hamilton, EB and Harris, G and Heijenbrok-Kal, MH and Helbok, R and Hellemons, ME and Hillus, D and Huijts, SM and Hultström, M and Jassat, W and Kurth, F and Larsson, IM and Lipcsey, M and Liu, C and Loflin, CD and Malinovschi, A and Mao, W and Mazankova, L and McCulloch, D and Menges, D and Mohammadifard, N and Munblit, D and Nekliudov, NA and Ogbuoji, O and Osmanov, IM and Peñalvo, JL and Petersen, MS and Puhan, MA and Rahman, M and Rass, V and Reinig, N and Ribbers, GM and Ricchiuto, A and Rubertsson, S and Samitova, E and Sarrafzadegan, N and Shikhaleva, A and Simpson, KE and Sinatti, D and Soriano, JB and Spiridonova, E and Steinbeis, F and Svistunov, AA and Valentini, P and van de Water, BJ and van den Berg-Emons, R and Wallin, E and Witzenrath, M and Wu, Y and Xu, H and Zoller, T and Adolph, C and Albright, J and Amlag, JO and Aravkin, AY and Bang-Jensen, BL and Bisignano, C and Castellano, R and Castro, E and Chakrabarti, S and Collins, JK and Dai, X and Daoud, F and Dapper, C and Deen, A and Duncan, BB and Erickson, M and Ewald, SB and Ferrari, AJ and Flaxman, AD and Fullman, N and Gamkrelidze, A and Giles, JR and Guo, G and Hay, SI and He, J and Helak, M and Hulland, EN and Kereselidze, M and Krohn, KJ and Lazzar-Atwood, A and Lindstrom, A and Lozano, R and Malta, DC and Månsson, J and Mantilla Herrera, AM and Mokdad, AH and Monasta, L and Nomura, S and Pasovic, M and Pigott, DM and Reiner, RC and Reinke, G and Ribeiro, ALP and Santomauro, DF and Sholokhov, A and Spurlock, EE and Walcott, R and Walker, A and Wiysonge, CS and Zheng, P and Bettger, JP and Murray, CJL and Vos, T},
title = {Estimated Global Proportions of Individuals With Persistent Fatigue, Cognitive, and Respiratory Symptom Clusters Following Symptomatic COVID-19 in 2020 and 2021.},
journal = {JAMA},
volume = {328},
number = {16},
pages = {1604-1615},
pmid = {36215063},
issn = {1538-3598},
support = {K23 NR019019/NR/NINR NIH HHS/United States ; T32 AI007044/AI/NIAID NIH HHS/United States ; },
mesh = {Adolescent ; Adult ; Aged ; Child ; Child, Preschool ; Female ; Humans ; Male ; Middle Aged ; Young Adult ; Bayes Theorem ; *COVID-19/complications/epidemiology ; *Fatigue/epidemiology/etiology ; Pain/epidemiology/etiology ; SARS-CoV-2 ; Syndrome ; *Cognition Disorders/epidemiology/etiology ; *Respiratory Insufficiency/epidemiology/etiology ; Internationality ; Global Health/statistics & numerical data ; Mood Disorders/epidemiology/etiology ; Post-Acute COVID-19 Syndrome ; },
abstract = {IMPORTANCE: Some individuals experience persistent symptoms after initial symptomatic SARS-CoV-2 infection (often referred to as Long COVID).
OBJECTIVE: To estimate the proportion of males and females with COVID-19, younger or older than 20 years of age, who had Long COVID symptoms in 2020 and 2021 and their Long COVID symptom duration.
Bayesian meta-regression and pooling of 54 studies and 2 medical record databases with data for 1.2 million individuals (from 22 countries) who had symptomatic SARS-CoV-2 infection. Of the 54 studies, 44 were published and 10 were collaborating cohorts (conducted in Austria, the Faroe Islands, Germany, Iran, Italy, the Netherlands, Russia, Sweden, Switzerland, and the US). The participant data were derived from the 44 published studies (10 501 hospitalized individuals and 42 891 nonhospitalized individuals), the 10 collaborating cohort studies (10 526 and 1906), and the 2 US electronic medical record databases (250 928 and 846 046). Data collection spanned March 2020 to January 2022.
EXPOSURES: Symptomatic SARS-CoV-2 infection.
MAIN OUTCOMES AND MEASURES: Proportion of individuals with at least 1 of the 3 self-reported Long COVID symptom clusters (persistent fatigue with bodily pain or mood swings; cognitive problems; or ongoing respiratory problems) 3 months after SARS-CoV-2 infection in 2020 and 2021, estimated separately for hospitalized and nonhospitalized individuals aged 20 years or older by sex and for both sexes of nonhospitalized individuals younger than 20 years of age.
RESULTS: A total of 1.2 million individuals who had symptomatic SARS-CoV-2 infection were included (mean age, 4-66 years; males, 26%-88%). In the modeled estimates, 6.2% (95% uncertainty interval [UI], 2.4%-13.3%) of individuals who had symptomatic SARS-CoV-2 infection experienced at least 1 of the 3 Long COVID symptom clusters in 2020 and 2021, including 3.2% (95% UI, 0.6%-10.0%) for persistent fatigue with bodily pain or mood swings, 3.7% (95% UI, 0.9%-9.6%) for ongoing respiratory problems, and 2.2% (95% UI, 0.3%-7.6%) for cognitive problems after adjusting for health status before COVID-19, comprising an estimated 51.0% (95% UI, 16.9%-92.4%), 60.4% (95% UI, 18.9%-89.1%), and 35.4% (95% UI, 9.4%-75.1%), respectively, of Long COVID cases. The Long COVID symptom clusters were more common in women aged 20 years or older (10.6% [95% UI, 4.3%-22.2%]) 3 months after symptomatic SARS-CoV-2 infection than in men aged 20 years or older (5.4% [95% UI, 2.2%-11.7%]). Both sexes younger than 20 years of age were estimated to be affected in 2.8% (95% UI, 0.9%-7.0%) of symptomatic SARS-CoV-2 infections. The estimated mean Long COVID symptom cluster duration was 9.0 months (95% UI, 7.0-12.0 months) among hospitalized individuals and 4.0 months (95% UI, 3.6-4.6 months) among nonhospitalized individuals. Among individuals with Long COVID symptoms 3 months after symptomatic SARS-CoV-2 infection, an estimated 15.1% (95% UI, 10.3%-21.1%) continued to experience symptoms at 12 months.
CONCLUSIONS AND RELEVANCE: This study presents modeled estimates of the proportion of individuals with at least 1 of 3 self-reported Long COVID symptom clusters (persistent fatigue with bodily pain or mood swings; cognitive problems; or ongoing respiratory problems) 3 months after symptomatic SARS-CoV-2 infection.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adolescent
Adult
Aged
Child
Child, Preschool
Female
Humans
Male
Middle Aged
Young Adult
Bayes Theorem
*COVID-19/complications/epidemiology
*Fatigue/epidemiology/etiology
Pain/epidemiology/etiology
SARS-CoV-2
Syndrome
*Cognition Disorders/epidemiology/etiology
*Respiratory Insufficiency/epidemiology/etiology
Internationality
Global Health/statistics & numerical data
Mood Disorders/epidemiology/etiology
Post-Acute COVID-19 Syndrome
RevDate: 2024-12-31
CmpDate: 2022-10-18
Baseline host determinants of robust human HIV-1 vaccine-induced immune responses: A meta-analysis of 26 vaccine regimens.
EBioMedicine, 84:104271.
BACKGROUND: The identification of baseline host determinants that associate with robust HIV-1 vaccine-induced immune responses could aid HIV-1 vaccine development. We aimed to assess both the collective and relative performance of baseline characteristics in classifying individual participants in nine different Phase 1-2 HIV-1 vaccine clinical trials (26 vaccine regimens, conducted in Africa and in the Americas) as High HIV-1 vaccine responders.
METHODS: This was a meta-analysis of individual participant data, with studies chosen based on participant-level (vs. study-level summary) data availability within the HIV-1 Vaccine Trials Network. We assessed the performance of 25 baseline characteristics (demographics, safety haematological measurements, vital signs, assay background measurements) and estimated the relative importance of each characteristic in classifying 831 participants as High (defined as within the top 25th percentile among positive responders or above the assay upper limit of quantification) versus Non-High responders. Immune response outcomes included HIV-1-specific serum IgG binding antibodies and Env-specific CD4+ T-cell responses assessed two weeks post-last dose, all measured at central HVTN laboratories. Three variable importance approaches based on SuperLearner ensemble machine learning were considered.
FINDINGS: Overall, 30.1%, 50.5%, 36.2%, and 13.9% of participants were categorized as High responders for gp120 IgG, gp140 IgG, gp41 IgG, and Env-specific CD4+ T-cell vaccine-induced responses, respectively. When including all baseline characteristics, moderate performance was achieved for the classification of High responder status for the binding antibody responses, with cross-validated areas under the ROC curve (CV-AUC) of 0.72 (95% CI: 0.68, 0.76) for gp120 IgG, 0.73 (0.69, 0.76) for gp140 IgG, and 0.67 (95% CI: 0.63, 0.72) for gp41 IgG. In contrast, the collection of all baseline characteristics yielded little improvement over chance for predicting High Env-specific CD4+ T-cell responses [CV-AUC: 0.53 (0.48, 0.58)]. While estimated variable importance patterns differed across the three approaches, female sex assigned at birth, lower height, and higher total white blood cell count emerged as significant predictors of High responder status across multiple immune response outcomes using Approach 1. Of these three baseline variables, total white blood cell count ranked highly across all three approaches for predicting vaccine-induced gp41 and gp140 High responder status.
INTERPRETATION: The identified features should be studied further in pursuit of intervention strategies to improve vaccine responses and may be adjusted for in analyses of immune response data to enhance statistical power.
FUNDING: National Institute of Allergy and Infectious Diseases (UM1AI068635 to YH, UM1AI068614 to GDT, UM1AI068618 to MJM, and UM1 AI069511 to MCK), the Duke CFAR P30 AI064518 to GDT, and National Institute of Dental and Craniofacial Research (R01DE027245 to JJK). This work was also supported by the Bill and Melinda Gates Foundation. The content is solely the responsibility of the authors and does not necessarily represent the official views of any of the funding sources.
Additional Links: PMID-36179551
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36179551,
year = {2022},
author = {Huang, Y and Zhang, Y and Seaton, KE and De Rosa, S and Heptinstall, J and Carpp, LN and Randhawa, AK and McKinnon, LR and McLaren, P and Viegas, E and Gray, GE and Churchyard, G and Buchbinder, SP and Edupuganti, S and Bekker, LG and Keefer, MC and Hosseinipour, MC and Goepfert, PA and Cohen, KW and Williamson, BD and McElrath, MJ and Tomaras, GD and Thakar, J and Kobie, JJ and , },
title = {Baseline host determinants of robust human HIV-1 vaccine-induced immune responses: A meta-analysis of 26 vaccine regimens.},
journal = {EBioMedicine},
volume = {84},
number = {},
pages = {104271},
pmid = {36179551},
issn = {2352-3964},
support = {UM1 AI069423/AI/NIAID NIH HHS/United States ; R01 DE027245/DE/NIDCR NIH HHS/United States ; UM1 AI069481/AI/NIAID NIH HHS/United States ; UM1 AI068614/AI/NIAID NIH HHS/United States ; UM1 AI068618/AI/NIAID NIH HHS/United States ; UM1 AI069496/AI/NIAID NIH HHS/United States ; UM1 AI068635/AI/NIAID NIH HHS/United States ; P30 AI064518/AI/NIAID NIH HHS/United States ; UM1 AI069511/AI/NIAID NIH HHS/United States ; R01 AI117787/AI/NIAID NIH HHS/United States ; UM1 AI154463/AI/NIAID NIH HHS/United States ; },
mesh = {Female ; Humans ; *AIDS Vaccines ; Antibody Formation ; HIV Antibodies ; *HIV Infections/prevention & control ; *HIV Seropositivity ; *HIV-1 ; Immunoglobulin G ; Male ; Adult ; },
abstract = {BACKGROUND: The identification of baseline host determinants that associate with robust HIV-1 vaccine-induced immune responses could aid HIV-1 vaccine development. We aimed to assess both the collective and relative performance of baseline characteristics in classifying individual participants in nine different Phase 1-2 HIV-1 vaccine clinical trials (26 vaccine regimens, conducted in Africa and in the Americas) as High HIV-1 vaccine responders.
METHODS: This was a meta-analysis of individual participant data, with studies chosen based on participant-level (vs. study-level summary) data availability within the HIV-1 Vaccine Trials Network. We assessed the performance of 25 baseline characteristics (demographics, safety haematological measurements, vital signs, assay background measurements) and estimated the relative importance of each characteristic in classifying 831 participants as High (defined as within the top 25th percentile among positive responders or above the assay upper limit of quantification) versus Non-High responders. Immune response outcomes included HIV-1-specific serum IgG binding antibodies and Env-specific CD4+ T-cell responses assessed two weeks post-last dose, all measured at central HVTN laboratories. Three variable importance approaches based on SuperLearner ensemble machine learning were considered.
FINDINGS: Overall, 30.1%, 50.5%, 36.2%, and 13.9% of participants were categorized as High responders for gp120 IgG, gp140 IgG, gp41 IgG, and Env-specific CD4+ T-cell vaccine-induced responses, respectively. When including all baseline characteristics, moderate performance was achieved for the classification of High responder status for the binding antibody responses, with cross-validated areas under the ROC curve (CV-AUC) of 0.72 (95% CI: 0.68, 0.76) for gp120 IgG, 0.73 (0.69, 0.76) for gp140 IgG, and 0.67 (95% CI: 0.63, 0.72) for gp41 IgG. In contrast, the collection of all baseline characteristics yielded little improvement over chance for predicting High Env-specific CD4+ T-cell responses [CV-AUC: 0.53 (0.48, 0.58)]. While estimated variable importance patterns differed across the three approaches, female sex assigned at birth, lower height, and higher total white blood cell count emerged as significant predictors of High responder status across multiple immune response outcomes using Approach 1. Of these three baseline variables, total white blood cell count ranked highly across all three approaches for predicting vaccine-induced gp41 and gp140 High responder status.
INTERPRETATION: The identified features should be studied further in pursuit of intervention strategies to improve vaccine responses and may be adjusted for in analyses of immune response data to enhance statistical power.
FUNDING: National Institute of Allergy and Infectious Diseases (UM1AI068635 to YH, UM1AI068614 to GDT, UM1AI068618 to MJM, and UM1 AI069511 to MCK), the Duke CFAR P30 AI064518 to GDT, and National Institute of Dental and Craniofacial Research (R01DE027245 to JJK). This work was also supported by the Bill and Melinda Gates Foundation. The content is solely the responsibility of the authors and does not necessarily represent the official views of any of the funding sources.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Female
Humans
*AIDS Vaccines
Antibody Formation
HIV Antibodies
*HIV Infections/prevention & control
*HIV Seropositivity
*HIV-1
Immunoglobulin G
Male
Adult
RevDate: 2025-08-28
CmpDate: 2022-10-03
Hydroxychloroquine/chloroquine for the treatment of hospitalized patients with COVID-19: An individual participant data meta-analysis.
PloS one, 17(9):e0273526.
BACKGROUND: Results from observational studies and randomized clinical trials (RCTs) have led to the consensus that hydroxychloroquine (HCQ) and chloroquine (CQ) are not effective for COVID-19 prevention or treatment. Pooling individual participant data, including unanalyzed data from trials terminated early, enables more detailed investigation of the efficacy and safety of HCQ/CQ among subgroups of hospitalized patients.
METHODS: We searched ClinicalTrials.gov in May and June 2020 for US-based RCTs evaluating HCQ/CQ in hospitalized COVID-19 patients in which the outcomes defined in this study were recorded or could be extrapolated. The primary outcome was a 7-point ordinal scale measured between day 28 and 35 post enrollment; comparisons used proportional odds ratios. Harmonized de-identified data were collected via a common template spreadsheet sent to each principal investigator. The data were analyzed by fitting a prespecified Bayesian ordinal regression model and standardizing the resulting predictions.
RESULTS: Eight of 19 trials met eligibility criteria and agreed to participate. Patient-level data were available from 770 participants (412 HCQ/CQ vs 358 control). Baseline characteristics were similar between groups. We did not find evidence of a difference in COVID-19 ordinal scores between days 28 and 35 post-enrollment in the pooled patient population (odds ratio, 0.97; 95% credible interval, 0.76-1.24; higher favors HCQ/CQ), and found no convincing evidence of meaningful treatment effect heterogeneity among prespecified subgroups. Adverse event and serious adverse event rates were numerically higher with HCQ/CQ vs control (0.39 vs 0.29 and 0.13 vs 0.09 per patient, respectively).
CONCLUSIONS: The findings of this individual participant data meta-analysis reinforce those of individual RCTs that HCQ/CQ is not efficacious for treatment of COVID-19 in hospitalized patients.
Additional Links: PMID-36173983
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36173983,
year = {2022},
author = {Di Stefano, L and Ogburn, EL and Ram, M and Scharfstein, DO and Li, T and Khanal, P and Baksh, SN and McBee, N and Gruber, J and Gildea, MR and Clark, MR and Goldenberg, NA and Bennani, Y and Brown, SM and Buckel, WR and Clement, ME and Mulligan, MJ and O'Halloran, JA and Rauseo, AM and Self, WH and Semler, MW and Seto, T and Stout, JE and Ulrich, RJ and Victory, J and Bierer, BE and Hanley, DF and Freilich, D and , },
title = {Hydroxychloroquine/chloroquine for the treatment of hospitalized patients with COVID-19: An individual participant data meta-analysis.},
journal = {PloS one},
volume = {17},
number = {9},
pages = {e0273526},
pmid = {36173983},
issn = {1932-6203},
support = {U01 HL123018/HL/NHLBI NIH HHS/United States ; U01 HL123031/HL/NHLBI NIH HHS/United States ; U01 HL123020/HL/NHLBI NIH HHS/United States ; U01 HL122989/HL/NHLBI NIH HHS/United States ; U01 HL123004/HL/NHLBI NIH HHS/United States ; U24 TR001609/TR/NCATS NIH HHS/United States ; U01 HL123008/HL/NHLBI NIH HHS/United States ; U01 HL123022/HL/NHLBI NIH HHS/United States ; U01 HL123023/HL/NHLBI NIH HHS/United States ; U01 HL123027/HL/NHLBI NIH HHS/United States ; U01 HL122998/HL/NHLBI NIH HHS/United States ; U01 HL123009/HL/NHLBI NIH HHS/United States ; UL1 TR002541/TR/NCATS NIH HHS/United States ; U01 HL123033/HL/NHLBI NIH HHS/United States ; U01 HL123010/HL/NHLBI NIH HHS/United States ; UL1 TR002345/TR/NCATS NIH HHS/United States ; },
mesh = {Chloroquine/adverse effects ; Data Analysis ; Humans ; *Hydroxychloroquine/adverse effects ; *COVID-19 Drug Treatment ; },
abstract = {BACKGROUND: Results from observational studies and randomized clinical trials (RCTs) have led to the consensus that hydroxychloroquine (HCQ) and chloroquine (CQ) are not effective for COVID-19 prevention or treatment. Pooling individual participant data, including unanalyzed data from trials terminated early, enables more detailed investigation of the efficacy and safety of HCQ/CQ among subgroups of hospitalized patients.
METHODS: We searched ClinicalTrials.gov in May and June 2020 for US-based RCTs evaluating HCQ/CQ in hospitalized COVID-19 patients in which the outcomes defined in this study were recorded or could be extrapolated. The primary outcome was a 7-point ordinal scale measured between day 28 and 35 post enrollment; comparisons used proportional odds ratios. Harmonized de-identified data were collected via a common template spreadsheet sent to each principal investigator. The data were analyzed by fitting a prespecified Bayesian ordinal regression model and standardizing the resulting predictions.
RESULTS: Eight of 19 trials met eligibility criteria and agreed to participate. Patient-level data were available from 770 participants (412 HCQ/CQ vs 358 control). Baseline characteristics were similar between groups. We did not find evidence of a difference in COVID-19 ordinal scores between days 28 and 35 post-enrollment in the pooled patient population (odds ratio, 0.97; 95% credible interval, 0.76-1.24; higher favors HCQ/CQ), and found no convincing evidence of meaningful treatment effect heterogeneity among prespecified subgroups. Adverse event and serious adverse event rates were numerically higher with HCQ/CQ vs control (0.39 vs 0.29 and 0.13 vs 0.09 per patient, respectively).
CONCLUSIONS: The findings of this individual participant data meta-analysis reinforce those of individual RCTs that HCQ/CQ is not efficacious for treatment of COVID-19 in hospitalized patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Chloroquine/adverse effects
Data Analysis
Humans
*Hydroxychloroquine/adverse effects
*COVID-19 Drug Treatment
RevDate: 2024-09-18
CmpDate: 2022-12-07
Revised Guidelines for Coronavirus Disease 19 Management in Hematopoietic Cell Transplantation and Cellular Therapy Recipients (August 2022).
Transplantation and cellular therapy, 28(12):810-821.
This document is intended as a guide for diagnosis and management of Coronavirus Disease 2019 (COVID-19), caused by the virus SARS-CoV-2, in adult and pediatric HCT and cellular therapy patients. This document was prepared using available data and with expert opinion provided by members of the (ASTCT) Infectious Diseases Special Interest Group (ID-SIG) and is an update of pervious publication. Since our original publication in 2020, the NIH and IDSA have published extensive guidelines for management of COVID-19 which are readily accessible (NIH Guidelines , IDSA Guidelines). This update focuses primarily on issues pertaining specifically to HCT/cellular therapy recipients. Information provided in this manuscript may change as new information becomes available.
Additional Links: PMID-36103987
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36103987,
year = {2022},
author = {Dioverti, V and Boghdadly, ZE and Shahid, Z and Waghmare, A and Abidi, MZ and Pergam, S and Boeckh, M and Dadwal, S and Kamboj, M and Seo, S and Chemaly, RF and Papanicolaou, GA},
title = {Revised Guidelines for Coronavirus Disease 19 Management in Hematopoietic Cell Transplantation and Cellular Therapy Recipients (August 2022).},
journal = {Transplantation and cellular therapy},
volume = {28},
number = {12},
pages = {810-821},
pmid = {36103987},
issn = {2666-6367},
support = {K23 AG073534/AG/NIA NIH HHS/United States ; },
mesh = {Adult ; Humans ; Child ; *COVID-19/therapy ; SARS-CoV-2 ; *Hematopoietic Stem Cell Transplantation ; Cell- and Tissue-Based Therapy ; },
abstract = {This document is intended as a guide for diagnosis and management of Coronavirus Disease 2019 (COVID-19), caused by the virus SARS-CoV-2, in adult and pediatric HCT and cellular therapy patients. This document was prepared using available data and with expert opinion provided by members of the (ASTCT) Infectious Diseases Special Interest Group (ID-SIG) and is an update of pervious publication. Since our original publication in 2020, the NIH and IDSA have published extensive guidelines for management of COVID-19 which are readily accessible (NIH Guidelines , IDSA Guidelines). This update focuses primarily on issues pertaining specifically to HCT/cellular therapy recipients. Information provided in this manuscript may change as new information becomes available.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Humans
Child
*COVID-19/therapy
SARS-CoV-2
*Hematopoietic Stem Cell Transplantation
Cell- and Tissue-Based Therapy
RevDate: 2025-02-25
CmpDate: 2024-06-27
Infectious Diseases Society of America Guidelines on the Treatment and Management of Patients With COVID-19 (September 2022).
Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, 78(7):e250-e349.
There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19), with rapidly changing efficacy and safety evidence from trials. The objective was to develop evidence-based, rapid, living guidelines intended to support patients, clinicians, and other healthcare professionals in their decisions about treatment and management of patients with COVID-19. In March 2020, the Infectious Diseases Society of America (IDSA) formed a multidisciplinary guideline panel of infectious disease clinicians, pharmacists, and methodologists with varied areas of expertise to regularly review the evidence and make recommendations about the treatment and management of persons with COVID-19. The process used a living guideline approach and followed a rapid recommendation development checklist. The panel prioritized questions and outcomes. A systematic review of the peer-reviewed and grey literature was conducted at regular intervals. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to assess the certainty of evidence and make recommendations. Based on the most recent search conducted on 31 May 2022, the IDSA guideline panel has made 32 recommendations for the treatment and management of the following groups/populations: pre- and postexposure prophylaxis, ambulatory with mild-to-moderate disease, and hospitalized with mild-to-moderate, severe but not critical, and critical disease. As these are living guidelines, the most recent recommendations can be found online at: https://idsociety.org/COVID19guidelines. At the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials. Since then, many trials were conducted that provided much-needed evidence for COVID-19 therapies. There still remain many unanswered questions as the pandemic evolved, which we hope future trials can answer.
Additional Links: PMID-36063397
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid36063397,
year = {2024},
author = {Bhimraj, A and Morgan, RL and Shumaker, AH and Baden, LR and Cheng, VC and Edwards, KM and Gallagher, JC and Gandhi, RT and Muller, WJ and Nakamura, MM and O'Horo, JC and Shafer, RW and Shoham, S and Murad, MH and Mustafa, RA and Sultan, S and Falck-Ytter, Y},
title = {Infectious Diseases Society of America Guidelines on the Treatment and Management of Patients With COVID-19 (September 2022).},
journal = {Clinical infectious diseases : an official publication of the Infectious Diseases Society of America},
volume = {78},
number = {7},
pages = {e250-e349},
doi = {10.1093/cid/ciac724},
pmid = {36063397},
issn = {1537-6591},
support = {R24 AI136618/AI/NIAID NIH HHS/United States ; /CC/CDC HHS/United States ; /HH/HHS/United States ; },
mesh = {Humans ; *COVID-19/therapy ; *SARS-CoV-2 ; United States ; Antiviral Agents/therapeutic use ; COVID-19 Drug Treatment ; },
abstract = {There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19), with rapidly changing efficacy and safety evidence from trials. The objective was to develop evidence-based, rapid, living guidelines intended to support patients, clinicians, and other healthcare professionals in their decisions about treatment and management of patients with COVID-19. In March 2020, the Infectious Diseases Society of America (IDSA) formed a multidisciplinary guideline panel of infectious disease clinicians, pharmacists, and methodologists with varied areas of expertise to regularly review the evidence and make recommendations about the treatment and management of persons with COVID-19. The process used a living guideline approach and followed a rapid recommendation development checklist. The panel prioritized questions and outcomes. A systematic review of the peer-reviewed and grey literature was conducted at regular intervals. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to assess the certainty of evidence and make recommendations. Based on the most recent search conducted on 31 May 2022, the IDSA guideline panel has made 32 recommendations for the treatment and management of the following groups/populations: pre- and postexposure prophylaxis, ambulatory with mild-to-moderate disease, and hospitalized with mild-to-moderate, severe but not critical, and critical disease. As these are living guidelines, the most recent recommendations can be found online at: https://idsociety.org/COVID19guidelines. At the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials. Since then, many trials were conducted that provided much-needed evidence for COVID-19 therapies. There still remain many unanswered questions as the pandemic evolved, which we hope future trials can answer.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/therapy
*SARS-CoV-2
United States
Antiviral Agents/therapeutic use
COVID-19 Drug Treatment
RevDate: 2025-12-03
CmpDate: 2022-12-27
Longitudinal lung function and gas transfer in individuals with idiopathic pulmonary fibrosis: a genome-wide association study.
The Lancet. Respiratory medicine, 11(1):65-73.
BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is an incurable lung disease characterised by progressive scarring leading to alveolar stiffness, reduced lung capacity, and impeded gas transfer. We aimed to identify genetic variants associated with declining lung capacity or declining gas transfer after diagnosis of IPF.
METHODS: We did a genome-wide meta-analysis of longitudinal measures of forced vital capacity (FVC) and diffusing capacity of the lung for carbon monoxide (DLCO) in individuals diagnosed with IPF. Individuals were recruited to three studies between June, 1996, and August, 2017, from across centres in the US, UK, and Spain. Suggestively significant variants were investigated further in an additional independent study (CleanUP-IPF). All four studies diagnosed cases following American Thoracic Society/European Respiratory Society guidelines. Variants were defined as significantly associated if they had a meta-analysis p<5 × 10[-8] when meta-analysing across all discovery and follow-up studies, had consistent direction of effects across all four studies, and were nominally significant (p<0·05) in each study.
FINDINGS: 1329 individuals with a total of 5216 measures were included in the FVC analysis. 975 individuals with a total of 3361 measures were included in the DLCO analysis. For the discovery genome-wide analyses, 7 611 174 genetic variants were included in the FVC analysis and 7 536 843 in the DLCO analysis. One variant (rs115982800) located in an antisense RNA gene for protein kinase N2 (PKN2) showed a genome-wide significant association with FVC decline (-140 mL/year per risk allele [95% CI -180 to -100]; p=9·14 × 10[-12]).
INTERPRETATION: Our analysis identifies a genetic variant associated with disease progression, which might highlight a new biological mechanism for IPF. We found that PKN2, a Rho and Rac effector protein, is the most likely gene of interest from this analysis. PKN2 inhibitors are currently in development and signify a potential novel therapeutic approach for IPF.
FUNDING: Action for Pulmonary Fibrosis, Medical Research Council, Wellcome Trust, and National Institutes of Health National Heart, Lung, and Blood Institute.
Additional Links: PMID-35985358
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35985358,
year = {2023},
author = {Allen, RJ and Oldham, JM and Jenkins, DA and Leavy, OC and Guillen-Guio, B and Melbourne, CA and Ma, SF and Jou, J and Kim, JS and , and Fahy, WA and Oballa, E and Hubbard, RB and Navaratnam, V and Braybrooke, R and Saini, G and Roach, KM and Tobin, MD and Hirani, N and Whyte, MKB and Kaminski, N and Zhang, Y and Martinez, FJ and Linderholm, AL and Adegunsoye, A and Strek, ME and Maher, TM and Molyneaux, PL and Flores, C and Noth, I and Gisli Jenkins, R and Wain, LV},
title = {Longitudinal lung function and gas transfer in individuals with idiopathic pulmonary fibrosis: a genome-wide association study.},
journal = {The Lancet. Respiratory medicine},
volume = {11},
number = {1},
pages = {65-73},
pmid = {35985358},
issn = {2213-2619},
support = {R01 HL141852/HL/NHLBI NIH HHS/United States ; UH3 HL145266/HL/NHLBI NIH HHS/United States ; 225221/WT_/Wellcome Trust/United Kingdom ; R01 HL127349/HL/NHLBI NIH HHS/United States ; R01 HL130796/HL/NHLBI NIH HHS/United States ; MC_PC_19004/MRC_/Medical Research Council/United Kingdom ; MC_PC_17228/MRC_/Medical Research Council/United Kingdom ; K23 HL138190/HL/NHLBI NIH HHS/United States ; U01 HL145567/HL/NHLBI NIH HHS/United States ; MR/V00235X/1/MRC_/Medical Research Council/United Kingdom ; R21 HL079394/HL/NHLBI NIH HHS/United States ; K23 HL146942/HL/NHLBI NIH HHS/United States ; MC_QA137853/MRC_/Medical Research Council/United Kingdom ; 221680/Z/20/Z/WT_/Wellcome Trust/United Kingdom ; R56 HL158935/HL/NHLBI NIH HHS/United States ; UH2 HL123886/HL/NHLBI NIH HHS/United States ; UG3 HL145266/HL/NHLBI NIH HHS/United States ; R21 HL161723/HL/NHLBI NIH HHS/United States ; },
mesh = {Humans ; *Genome-Wide Association Study ; *Idiopathic Pulmonary Fibrosis/diagnosis ; Lung ; Vital Capacity ; Lung Volume Measurements ; },
abstract = {BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is an incurable lung disease characterised by progressive scarring leading to alveolar stiffness, reduced lung capacity, and impeded gas transfer. We aimed to identify genetic variants associated with declining lung capacity or declining gas transfer after diagnosis of IPF.
METHODS: We did a genome-wide meta-analysis of longitudinal measures of forced vital capacity (FVC) and diffusing capacity of the lung for carbon monoxide (DLCO) in individuals diagnosed with IPF. Individuals were recruited to three studies between June, 1996, and August, 2017, from across centres in the US, UK, and Spain. Suggestively significant variants were investigated further in an additional independent study (CleanUP-IPF). All four studies diagnosed cases following American Thoracic Society/European Respiratory Society guidelines. Variants were defined as significantly associated if they had a meta-analysis p<5 × 10[-8] when meta-analysing across all discovery and follow-up studies, had consistent direction of effects across all four studies, and were nominally significant (p<0·05) in each study.
FINDINGS: 1329 individuals with a total of 5216 measures were included in the FVC analysis. 975 individuals with a total of 3361 measures were included in the DLCO analysis. For the discovery genome-wide analyses, 7 611 174 genetic variants were included in the FVC analysis and 7 536 843 in the DLCO analysis. One variant (rs115982800) located in an antisense RNA gene for protein kinase N2 (PKN2) showed a genome-wide significant association with FVC decline (-140 mL/year per risk allele [95% CI -180 to -100]; p=9·14 × 10[-12]).
INTERPRETATION: Our analysis identifies a genetic variant associated with disease progression, which might highlight a new biological mechanism for IPF. We found that PKN2, a Rho and Rac effector protein, is the most likely gene of interest from this analysis. PKN2 inhibitors are currently in development and signify a potential novel therapeutic approach for IPF.
FUNDING: Action for Pulmonary Fibrosis, Medical Research Council, Wellcome Trust, and National Institutes of Health National Heart, Lung, and Blood Institute.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Genome-Wide Association Study
*Idiopathic Pulmonary Fibrosis/diagnosis
Lung
Vital Capacity
Lung Volume Measurements
RevDate: 2024-09-03
CmpDate: 2022-09-22
Clinical Practice Guidelines From the Association for the Advancement of Blood and Biotherapies (AABB): COVID-19 Convalescent Plasma.
Annals of internal medicine, 175(9):1310-1321.
DESCRIPTION: Coronavirus disease 2019 convalescent plasma (CCP) has emerged as a potential treatment of COVID-19. However, meta-analysis data and recommendations are limited. The Association for the Advancement of Blood and Biotherapies (AABB) developed clinical practice guidelines for the appropriate use of CCP.
METHODS: These guidelines are based on 2 living systematic reviews of randomized controlled trials (RCTs) evaluating CCP from 1 January 2019 to 26 January 2022. There were 33 RCTs assessing 21 916 participants. The results were summarized using the GRADE (Grading of Recommendations Assessment, Development and Evaluation) method. An expert panel reviewed the data using the GRADE framework to formulate recommendations.
RECOMMENDATION 1 (OUTPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for outpatients with COVID-19 who are at high risk for disease progression (weak recommendation, moderate-certainty evidence).
RECOMMENDATION 2 (INPATIENT): The AABB recommends against CCP transfusion for unselected hospitalized persons with moderate or severe disease (strong recommendation, high-certainty evidence). This recommendation does not apply to immunosuppressed patients or those who lack antibodies against SARS-CoV-2.
RECOMMENDATION 3 (INPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for hospitalized patients with COVID-19 who do not have SARS-CoV-2 antibodies detected at admission (weak recommendation, low-certainty evidence).
RECOMMENDATION 4 (INPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for hospitalized patients with COVID-19 and preexisting immunosuppression (weak recommendation, low-certainty evidence).
The AABB suggests against prophylactic CCP transfusion for uninfected persons with close contact exposure to a person with COVID-19 (weak recommendation, low-certainty evidence).
CCP is most effective when transfused with high neutralizing titers to infected patients early after symptom onset.
Additional Links: PMID-35969859
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35969859,
year = {2022},
author = {Estcourt, LJ and Cohn, CS and Pagano, MB and Iannizzi, C and Kreuzberger, N and Skoetz, N and Allen, ES and Bloch, EM and Beaudoin, G and Casadevall, A and Devine, DV and Foroutan, F and Gniadek, TJ and Goel, R and Gorlin, J and Grossman, BJ and Joyner, MJ and Metcalf, RA and Raval, JS and Rice, TW and Shaz, BH and Vassallo, RR and Winters, JL and Tobian, AAR},
title = {Clinical Practice Guidelines From the Association for the Advancement of Blood and Biotherapies (AABB): COVID-19 Convalescent Plasma.},
journal = {Annals of internal medicine},
volume = {175},
number = {9},
pages = {1310-1321},
pmid = {35969859},
issn = {1539-3704},
support = {K23 HL151826/HL/NHLBI NIH HHS/United States ; },
mesh = {*COVID-19/therapy ; Hospitalization ; Humans ; Immunization, Passive/methods ; *SARS-CoV-2 ; COVID-19 Serotherapy ; },
abstract = {DESCRIPTION: Coronavirus disease 2019 convalescent plasma (CCP) has emerged as a potential treatment of COVID-19. However, meta-analysis data and recommendations are limited. The Association for the Advancement of Blood and Biotherapies (AABB) developed clinical practice guidelines for the appropriate use of CCP.
METHODS: These guidelines are based on 2 living systematic reviews of randomized controlled trials (RCTs) evaluating CCP from 1 January 2019 to 26 January 2022. There were 33 RCTs assessing 21 916 participants. The results were summarized using the GRADE (Grading of Recommendations Assessment, Development and Evaluation) method. An expert panel reviewed the data using the GRADE framework to formulate recommendations.
RECOMMENDATION 1 (OUTPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for outpatients with COVID-19 who are at high risk for disease progression (weak recommendation, moderate-certainty evidence).
RECOMMENDATION 2 (INPATIENT): The AABB recommends against CCP transfusion for unselected hospitalized persons with moderate or severe disease (strong recommendation, high-certainty evidence). This recommendation does not apply to immunosuppressed patients or those who lack antibodies against SARS-CoV-2.
RECOMMENDATION 3 (INPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for hospitalized patients with COVID-19 who do not have SARS-CoV-2 antibodies detected at admission (weak recommendation, low-certainty evidence).
RECOMMENDATION 4 (INPATIENT): The AABB suggests CCP transfusion in addition to the usual standard of care for hospitalized patients with COVID-19 and preexisting immunosuppression (weak recommendation, low-certainty evidence).
The AABB suggests against prophylactic CCP transfusion for uninfected persons with close contact exposure to a person with COVID-19 (weak recommendation, low-certainty evidence).
CCP is most effective when transfused with high neutralizing titers to infected patients early after symptom onset.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/therapy
Hospitalization
Humans
Immunization, Passive/methods
*SARS-CoV-2
COVID-19 Serotherapy
RevDate: 2025-05-30
CmpDate: 2022-10-17
Using time-weighted average change from baseline of SARS-CoV-2 viral load to assess impact of hydroxychloroquine as postexposure prophylaxis and early treatment for COVID-19.
Journal of medical virology, 94(12):6091-6096.
Two randomized controlled trials demonstrated no clinical benefit of hydroxychloroquine (HCQ) for either postexposure prophylaxis or early treatment of SARS-CoV-2 infection. Using data from these studies, we calculated the time-weighted average change from baseline SARS-CoV-2 viral load and demonstrated that HCQ did not affect viral clearance.
Additional Links: PMID-35940869
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35940869,
year = {2022},
author = {Kumbhakar, R and Neradilek, M and Barnabas, RV and Stewart, J and Stankiewicz Karita, HC and Landovitz, RJ and Kissinger, PJ and Jerome, KR and Paasche-Orlow, MK and Bershteyn, A and Chu, HY and Neuzil, KM and Greninger, AL and Luk, A and Wald, A and Brown, ER and Johnston, C},
title = {Using time-weighted average change from baseline of SARS-CoV-2 viral load to assess impact of hydroxychloroquine as postexposure prophylaxis and early treatment for COVID-19.},
journal = {Journal of medical virology},
volume = {94},
number = {12},
pages = {6091-6096},
pmid = {35940869},
issn = {1096-9071},
support = {R01 CA213130/CA/NCI NIH HHS/United States ; T32 AI007140/AI/NIAID NIH HHS/United States ; /NH/NIH HHS/United States ; },
mesh = {*COVID-19/prevention & control ; Humans ; Hydroxychloroquine/therapeutic use ; *SARS-CoV-2 ; Viral Load ; *COVID-19 Drug Treatment ; },
abstract = {Two randomized controlled trials demonstrated no clinical benefit of hydroxychloroquine (HCQ) for either postexposure prophylaxis or early treatment of SARS-CoV-2 infection. Using data from these studies, we calculated the time-weighted average change from baseline SARS-CoV-2 viral load and demonstrated that HCQ did not affect viral clearance.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/prevention & control
Humans
Hydroxychloroquine/therapeutic use
*SARS-CoV-2
Viral Load
*COVID-19 Drug Treatment
RevDate: 2025-07-28
CmpDate: 2022-08-17
Mental and social wellbeing and the UK coronavirus job retention scheme: Evidence from nine longitudinal studies.
Social science & medicine (1982), 308:115226.
BACKGROUND: The COVID-19 pandemic has led to major economic disruptions. In March 2020, the UK implemented the Coronavirus Job Retention Scheme - known as furlough - to minimize the impact of job losses. We investigate associations between change in employment status and mental and social wellbeing during the early stages of the pandemic.
METHODS: Data were from 25,670 respondents, aged 17-66, across nine UK longitudinal studies. Furlough and other employment changes were defined using employment status pre-pandemic and during the first lockdown (April-June 2020). Mental and social wellbeing outcomes included psychological distress, life satisfaction, self-rated health, social contact, and loneliness. Study-specific modified Poisson regression estimates, adjusting for socio-demographic characteristics and pre-pandemic mental and social wellbeing, were pooled using meta-analysis. Associations were also stratified by sex, age, education, and household composition.
RESULTS: Compared to those who remained working, furloughed workers were at greater risk of psychological distress (adjusted risk ratio, ARR = 1.12; 95%CI: 0.97, 1.29), low life satisfaction (ARR = 1.14; 95%CI: 1.07, 1.22), loneliness (ARR = 1.12; 95%CI: 1.01, 1.23), and poor self-rated health (ARR = 1.26; 95%CI: 1.05, 1.50). Nevertheless, compared to furloughed workers, those who became unemployed had greater risk of psychological distress (ARR = 1.30; 95%CI: 1.12, 1.52), low life satisfaction (ARR = 1.16; 95%CI: 0.98, 1.38), and loneliness (ARR = 1.67; 95%CI: 1.08, 2.59). Effects were not uniform across all sub-groups.
CONCLUSIONS: During the early stages of the pandemic, those furloughed had increased risk of poor mental and social wellbeing, but furloughed workers fared better than those who became unemployed, suggesting that furlough may have partly mitigated poorer outcomes.
Additional Links: PMID-35932537
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35932537,
year = {2022},
author = {Jacques Wels, and Booth, C and Wielgoszewska, B and Green, MJ and Di Gessa, G and Huggins, CF and Griffith, GJ and Kwong, ASF and Bowyer, RCE and Maddock, J and Patalay, P and Silverwood, RJ and Fitzsimons, E and Shaw, R and Thompson, EJ and Steptoe, A and Hughes, A and Chaturvedi, N and Steves, CJ and Katikireddi, SV and Ploubidis, GB},
title = {Mental and social wellbeing and the UK coronavirus job retention scheme: Evidence from nine longitudinal studies.},
journal = {Social science & medicine (1982)},
volume = {308},
number = {},
pages = {115226},
pmid = {35932537},
issn = {1873-5347},
support = {CZD/16/6/CSO_/Chief Scientist Office/United Kingdom ; /DH_/Department of Health/United Kingdom ; MC_UU_00019/1/MRC_/Medical Research Council/United Kingdom ; MC_PC_15018/MRC_/Medical Research Council/United Kingdom ; SCAF/15/02/CSO_/Chief Scientist Office/United Kingdom ; R01 AG017644/AG/NIA NIH HHS/United States ; 216767/Z/19/Z/WT_/Wellcome Trust/United Kingdom ; MC_PC_20059/MRC_/Medical Research Council/United Kingdom ; SPHSU17/CSO_/Chief Scientist Office/United Kingdom ; MC_UU_00019/3/MRC_/Medical Research Council/United Kingdom ; 217065/Z/19/Z/WT_/Wellcome Trust/United Kingdom ; MC_PC_19009/MRC_/Medical Research Council/United Kingdom ; MC_UU_00022/2/MRC_/Medical Research Council/United Kingdom ; 104036/Z/14/Z/WT_/Wellcome Trust/United Kingdom ; G9815508/MRC_/Medical Research Council/United Kingdom ; MC_PC_20030/MRC_/Medical Research Council/United Kingdom ; },
mesh = {*COVID-19/epidemiology ; Communicable Disease Control ; Humans ; Longitudinal Studies ; Mental Health ; *Pandemics ; United Kingdom/epidemiology ; },
abstract = {BACKGROUND: The COVID-19 pandemic has led to major economic disruptions. In March 2020, the UK implemented the Coronavirus Job Retention Scheme - known as furlough - to minimize the impact of job losses. We investigate associations between change in employment status and mental and social wellbeing during the early stages of the pandemic.
METHODS: Data were from 25,670 respondents, aged 17-66, across nine UK longitudinal studies. Furlough and other employment changes were defined using employment status pre-pandemic and during the first lockdown (April-June 2020). Mental and social wellbeing outcomes included psychological distress, life satisfaction, self-rated health, social contact, and loneliness. Study-specific modified Poisson regression estimates, adjusting for socio-demographic characteristics and pre-pandemic mental and social wellbeing, were pooled using meta-analysis. Associations were also stratified by sex, age, education, and household composition.
RESULTS: Compared to those who remained working, furloughed workers were at greater risk of psychological distress (adjusted risk ratio, ARR = 1.12; 95%CI: 0.97, 1.29), low life satisfaction (ARR = 1.14; 95%CI: 1.07, 1.22), loneliness (ARR = 1.12; 95%CI: 1.01, 1.23), and poor self-rated health (ARR = 1.26; 95%CI: 1.05, 1.50). Nevertheless, compared to furloughed workers, those who became unemployed had greater risk of psychological distress (ARR = 1.30; 95%CI: 1.12, 1.52), low life satisfaction (ARR = 1.16; 95%CI: 0.98, 1.38), and loneliness (ARR = 1.67; 95%CI: 1.08, 2.59). Effects were not uniform across all sub-groups.
CONCLUSIONS: During the early stages of the pandemic, those furloughed had increased risk of poor mental and social wellbeing, but furloughed workers fared better than those who became unemployed, suggesting that furlough may have partly mitigated poorer outcomes.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/epidemiology
Communicable Disease Control
Humans
Longitudinal Studies
Mental Health
*Pandemics
United Kingdom/epidemiology
RevDate: 2025-08-26
CmpDate: 2022-08-09
Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal HCoVs.
Nature genetics, 54(8):1090-1102.
CRISPR knockout (KO) screens have identified host factors regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication. Here, we conducted a meta-analysis of these screens, which showed a high level of cell-type specificity of the identified hits, highlighting the necessity of additional models to uncover the full landscape of host factors. Thus, we performed genome-wide KO and activation screens in Calu-3 lung cells and KO screens in Caco-2 colorectal cells, followed by secondary screens in four human cell lines. This revealed host-dependency factors, including AP1G1 adaptin and ATP8B1 flippase, as well as inhibitors, including mucins. Interestingly, some of the identified genes also modulate Middle East respiratory syndrome coronavirus (MERS-CoV) and seasonal human coronavirus (HCoV) (HCoV-NL63 and HCoV-229E) replication. Moreover, most genes had an impact on viral entry, with AP1G1 likely regulating TMPRSS2 activity at the plasma membrane. These results demonstrate the value of multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential targets for therapeutic interventions.
Additional Links: PMID-35879413
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35879413,
year = {2022},
author = {Rebendenne, A and Roy, P and Bonaventure, B and Chaves Valadão, AL and Desmarets, L and Arnaud-Arnould, M and Rouillé, Y and Tauziet, M and Giovannini, D and Touhami, J and Lee, Y and DeWeirdt, P and Hegde, M and Urbach, S and Koulali, KE and de Gracia, FG and McKellar, J and Dubuisson, J and Wencker, M and Belouzard, S and Moncorgé, O and Doench, JG and Goujon, C},
title = {Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal HCoVs.},
journal = {Nature genetics},
volume = {54},
number = {8},
pages = {1090-1102},
pmid = {35879413},
issn = {1546-1718},
support = {759226/ERC_/European Research Council/International ; R21 AI157835/AI/NIAID NIH HHS/United States ; },
mesh = {*COVID-19/genetics ; Caco-2 Cells ; Clustered Regularly Interspaced Short Palindromic Repeats/genetics ; Humans ; *Middle East Respiratory Syndrome Coronavirus/genetics ; SARS-CoV-2/genetics ; Seasons ; },
abstract = {CRISPR knockout (KO) screens have identified host factors regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication. Here, we conducted a meta-analysis of these screens, which showed a high level of cell-type specificity of the identified hits, highlighting the necessity of additional models to uncover the full landscape of host factors. Thus, we performed genome-wide KO and activation screens in Calu-3 lung cells and KO screens in Caco-2 colorectal cells, followed by secondary screens in four human cell lines. This revealed host-dependency factors, including AP1G1 adaptin and ATP8B1 flippase, as well as inhibitors, including mucins. Interestingly, some of the identified genes also modulate Middle East respiratory syndrome coronavirus (MERS-CoV) and seasonal human coronavirus (HCoV) (HCoV-NL63 and HCoV-229E) replication. Moreover, most genes had an impact on viral entry, with AP1G1 likely regulating TMPRSS2 activity at the plasma membrane. These results demonstrate the value of multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential targets for therapeutic interventions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/genetics
Caco-2 Cells
Clustered Regularly Interspaced Short Palindromic Repeats/genetics
Humans
*Middle East Respiratory Syndrome Coronavirus/genetics
SARS-CoV-2/genetics
Seasons
RevDate: 2023-02-15
CmpDate: 2022-11-30
Detailed stratified GWAS analysis for severe COVID-19 in four European populations.
Human molecular genetics, 31(23):3945-3966.
Given the highly variable clinical phenotype of Coronavirus disease 2019 (COVID-19), a deeper analysis of the host genetic contribution to severe COVID-19 is important to improve our understanding of underlying disease mechanisms. Here, we describe an extended genome-wide association meta-analysis of a well-characterized cohort of 3255 COVID-19 patients with respiratory failure and 12 488 population controls from Italy, Spain, Norway and Germany/Austria, including stratified analyses based on age, sex and disease severity, as well as targeted analyses of chromosome Y haplotypes, the human leukocyte antigen region and the SARS-CoV-2 peptidome. By inversion imputation, we traced a reported association at 17q21.31 to a ~0.9-Mb inversion polymorphism that creates two highly differentiated haplotypes and characterized the potential effects of the inversion in detail. Our data, together with the 5th release of summary statistics from the COVID-19 Host Genetics Initiative including non-Caucasian individuals, also identified a new locus at 19q13.33, including NAPSA, a gene which is expressed primarily in alveolar cells responsible for gas exchange in the lung.
Additional Links: PMID-35848942
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35848942,
year = {2022},
author = {Degenhardt, F and Ellinghaus, D and Juzenas, S and Lerga-Jaso, J and Wendorff, M and Maya-Miles, D and Uellendahl-Werth, F and ElAbd, H and Rühlemann, MC and Arora, J and Özer, O and Lenning, OB and Myhre, R and Vadla, MS and Wacker, EM and Wienbrandt, L and Blandino Ortiz, A and de Salazar, A and Garrido Chercoles, A and Palom, A and Ruiz, A and Garcia-Fernandez, AE and Blanco-Grau, A and Mantovani, A and Zanella, A and Holten, AR and Mayer, A and Bandera, A and Cherubini, A and Protti, A and Aghemo, A and Gerussi, A and Ramirez, A and Braun, A and Nebel, A and Barreira, A and Lleo, A and Teles, A and Kildal, AB and Biondi, A and Caballero-Garralda, A and Ganna, A and Gori, A and Glück, A and Lind, A and Tanck, A and Hinney, A and Carreras Nolla, A and Fracanzani, AL and Peschuck, A and Cavallero, A and Dyrhol-Riise, AM and Ruello, A and Julià , A and Muscatello, A and Pesenti, A and Voza, A and Rando-Segura, A and Solier, A and Schmidt, A and Cortes, B and Mateos, B and Nafria-Jimenez, B and Schaefer, B and Jensen, B and Bellinghausen, C and Maj, C and Ferrando, C and de la Horra, C and Quereda, C and Skurk, C and Thibeault, C and Scollo, C and Herr, C and Spinner, CD and Gassner, C and Lange, C and Hu, C and Paccapelo, C and Lehmann, C and Angelini, C and Cappadona, C and Azuure, C and , and Bianco, C and Cea, C and Sancho, C and Hoff, DAL and Galimberti, D and Prati, D and Haschka, D and Jiménez, D and Pestaña, D and Toapanta, D and Muñiz-Diaz, E and Azzolini, E and Sandoval, E and Binatti, E and Scarpini, E and Helbig, ET and Casalone, E and Urrechaga, E and Paraboschi, EM and Pontali, E and Reverter, E and Calderón, EJ and Navas, E and SolligÃ¥rd, E and Contro, E and Arana-Arri, E and Aziz, F and Garcia, F and GarcÃa Sánchez, F and Ceriotti, F and Martinelli-Boneschi, F and Peyvandi, F and Kurth, F and Blasi, F and Malvestiti, F and Medrano, FJ and Mesonero, F and Rodriguez-Frias, F and Hanses, F and Müller, F and Hemmrich-Stanisak, G and Bellani, G and Grasselli, G and Pezzoli, G and Costantino, G and Albano, G and Cardamone, G and Bellelli, G and Citerio, G and Foti, G and Lamorte, G and Matullo, G and Baselli, G and Kurihara, H and Neb, H and My, I and Kurth, I and Hernández, I and Pink, I and de Rojas, I and Galván-Femenia, I and Holter, JC and Afset, JE and Heyckendorf, J and Kässens, J and DamÃ¥s, JK and Rybniker, J and Altmüller, J and Ampuero, J and MartÃn, J and Erdmann, J and Banales, JM and Badia, JR and Dopazo, J and Schneider, J and Bergan, J and Barretina, J and Walter, J and Hernández Quero, J and Goikoetxea, J and Delgado, J and Guerrero, JM and Fazaal, J and Kraft, J and Schröder, J and Risnes, K and Banasik, K and Müller, KE and Gaede, KI and Garcia-Etxebarria, K and Tonby, K and Heggelund, L and Izquierdo-Sanchez, L and Bettini, LR and Sumoy, L and Sander, LE and Lippert, LJ and Terranova, L and Nkambule, L and Knopp, L and Gustad, LT and Garbarino, L and Santoro, L and Téllez, L and Roade, L and Ostadreza, M and Intxausti, M and Kogevinas, M and Riveiro-Barciela, M and Berger, MM and Schaefer, M and Niemi, MEK and Gutiérrez-Stampa, MA and Carrabba, M and Figuera Basso, ME and Valsecchi, MG and Hernandez-Tejero, M and Vehreschild, MJGT and Manunta, M and Acosta-Herrera, M and D'Angiò, M and Baldini, M and Cazzaniga, M and Grimsrud, MM and Cornberg, M and Nöthen, MM and Marquié, M and Castoldi, M and Cordioli, M and Cecconi, M and D'Amato, M and Augustin, M and Tomasi, M and Boada, M and Dreher, M and Seilmaier, MJ and Joannidis, M and Wittig, M and Mazzocco, M and Ciccarelli, M and RodrÃguez-GandÃa, M and Bocciolone, M and Miozzo, M and Imaz Ayo, N and Blay, N and Chueca, N and Montano, N and Braun, N and Ludwig, N and Marx, N and MartÃnez, N and , and Cornely, OA and Witzke, O and Palmieri, O and , and Faverio, P and Preatoni, P and Bonfanti, P and Omodei, P and Tentorio, P and Castro, P and Rodrigues, PM and España, PP and Hoffmann, P and Rosenstiel, P and Schommers, P and Suwalski, P and de Pablo, R and Ferrer, R and Bals, R and Gualtierotti, R and Gallego-Durán, R and Nieto, R and Carpani, R and Morilla, R and Badalamenti, S and Haider, S and Ciesek, S and May, S and Bombace, S and Marsal, S and Pigazzini, S and Klein, S and Pelusi, S and Wilfling, S and Bosari, S and Volland, S and Brunak, S and Raychaudhuri, S and Schreiber, S and Heilmann-Heimbach, S and Aliberti, S and Ripke, S and Dudman, S and Wesse, T and Zheng, T and , and Bahmer, T and Eggermann, T and Illig, T and Brenner, T and Pumarola, T and Feldt, T and Folseraas, T and Gonzalez Cejudo, T and Landmesser, U and Protzer, U and Hehr, U and Rimoldi, V and Monzani, V and Skogen, V and Keitel, V and Kopfnagel, V and Friaza, V and Andrade, V and Moreno, V and Albrecht, W and Peter, W and Poller, W and Farre, X and Yi, X and Wang, X and Khodamoradi, Y and Karadeniz, Z and Latiano, A and Goerg, S and Bacher, P and Koehler, P and Tran, F and Zoller, H and Schulte, EC and Heidecker, B and Ludwig, KU and Fernández, J and Romero-Gómez, M and Albillos, A and Invernizzi, P and Buti, M and Duga, S and Bujanda, L and Hov, JR and Lenz, TL and Asselta, R and de Cid, R and Valenti, L and Karlsen, TH and Cáceres, M and Franke, A},
title = {Detailed stratified GWAS analysis for severe COVID-19 in four European populations.},
journal = {Human molecular genetics},
volume = {31},
number = {23},
pages = {3945-3966},
pmid = {35848942},
issn = {1460-2083},
support = {R01 AR063759/AR/NIAMS NIH HHS/United States ; },
mesh = {Humans ; *COVID-19/genetics ; SARS-CoV-2/genetics ; Genome-Wide Association Study ; Haplotypes ; Polymorphism, Genetic ; },
abstract = {Given the highly variable clinical phenotype of Coronavirus disease 2019 (COVID-19), a deeper analysis of the host genetic contribution to severe COVID-19 is important to improve our understanding of underlying disease mechanisms. Here, we describe an extended genome-wide association meta-analysis of a well-characterized cohort of 3255 COVID-19 patients with respiratory failure and 12 488 population controls from Italy, Spain, Norway and Germany/Austria, including stratified analyses based on age, sex and disease severity, as well as targeted analyses of chromosome Y haplotypes, the human leukocyte antigen region and the SARS-CoV-2 peptidome. By inversion imputation, we traced a reported association at 17q21.31 to a ~0.9-Mb inversion polymorphism that creates two highly differentiated haplotypes and characterized the potential effects of the inversion in detail. Our data, together with the 5th release of summary statistics from the COVID-19 Host Genetics Initiative including non-Caucasian individuals, also identified a new locus at 19q13.33, including NAPSA, a gene which is expressed primarily in alveolar cells responsible for gas exchange in the lung.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/genetics
SARS-CoV-2/genetics
Genome-Wide Association Study
Haplotypes
Polymorphism, Genetic
RevDate: 2025-05-30
CmpDate: 2022-11-18
A MUC5B Gene Polymorphism, rs35705950-T, Confers Protective Effects Against COVID-19 Hospitalization but Not Severe Disease or Mortality.
American journal of respiratory and critical care medicine, 206(10):1220-1229.
Rationale: A common MUC5B gene polymorphism, rs35705950-T, is associated with idiopathic pulmonary fibrosis (IPF), but its role in severe acute respiratory syndrome coronavirus 2 infection and disease severity is unclear. Objectives: To assess whether rs35705950-T confers differential risk for clinical outcomes associated with coronavirus disease (COVID-19) infection among participants in the Million Veteran Program (MVP). Methods: The MUC5B rs35705950-T allele was directly genotyped among MVP participants; clinical events and comorbidities were extracted from the electronic health records. Associations between the incidence or severity of COVID-19 and rs35705950-T were analyzed within each ancestry group in the MVP followed by transancestry meta-analysis. Replication and joint meta-analysis were conducted using summary statistics from the COVID-19 Host Genetics Initiative (HGI). Sensitivity analyses with adjustment for additional covariates (body mass index, Charlson comorbidity index, smoking, asbestosis, rheumatoid arthritis with interstitial lung disease, and IPF) and associations with post-COVID-19 pneumonia were performed in MVP subjects. Measurements and Main Results: The rs35705950-T allele was associated with fewer COVID-19 hospitalizations in transancestry meta-analyses within the MVP (Ncases = 4,325; Ncontrols = 507,640; OR = 0.89 [0.82-0.97]; P = 6.86 × 10[-3]) and joint meta-analyses with the HGI (Ncases = 13,320; Ncontrols = 1,508,841; OR, 0.90 [0.86-0.95]; P = 8.99 × 10[-5]). The rs35705950-T allele was not associated with reduced COVID-19 positivity in transancestry meta-analysis within the MVP (Ncases = 19,168/Ncontrols = 492,854; OR, 0.98 [0.95-1.01]; P = 0.06) but was nominally significant (P < 0.05) in the joint meta-analysis with the HGI (Ncases = 44,820; Ncontrols = 1,775,827; OR, 0.97 [0.95-1.00]; P = 0.03). Associations were not observed with severe outcomes or mortality. Among individuals of European ancestry in the MVP, rs35705950-T was associated with fewer post-COVID-19 pneumonia events (OR, 0.82 [0.72-0.93]; P = 0.001). Conclusions: The MUC5B variant rs35705950-T may confer protection in COVID-19 hospitalizations.
Additional Links: PMID-35771531
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35771531,
year = {2022},
author = {Verma, A and Minnier, J and Wan, ES and Huffman, JE and Gao, L and Joseph, J and Ho, YL and Wu, WC and Cho, K and Gorman, BR and Rajeevan, N and Pyarajan, S and Garcon, H and Meigs, JB and Sun, YV and Reaven, PD and McGeary, JE and Suzuki, A and Gelernter, J and Lynch, JA and Petersen, JM and Zekavat, SM and Natarajan, P and Dalal, S and Jhala, DN and Arjomandi, M and Gatsby, E and Lynch, KE and Bonomo, RA and Freiberg, M and Pathak, GA and Zhou, JJ and Donskey, CJ and Madduri, RK and Wells, QS and Huang, RDL and Polimanti, R and Chang, KM and Liao, KP and Tsao, PS and Wilson, PWF and Hung, AM and O'Donnell, CJ and Gaziano, JM and Hauger, RL and Iyengar, SK and Luoh, SW and , },
title = {A MUC5B Gene Polymorphism, rs35705950-T, Confers Protective Effects Against COVID-19 Hospitalization but Not Severe Disease or Mortality.},
journal = {American journal of respiratory and critical care medicine},
volume = {206},
number = {10},
pages = {1220-1229},
pmid = {35771531},
issn = {1535-4970},
support = {R01HL142711/HL/NHLBI NIH HHS/United States ; I01 CX001727/CX/CSRD VA/United States ; R01 HG006139/HG/NHGRI NIH HHS/United States ; R01 HL127564/HL/NHLBI NIH HHS/United States ; I01 CX001897/CX/CSRD VA/United States ; R01 HL142711/HL/NHLBI NIH HHS/United States ; R01HL127564/HL/NHLBI NIH HHS/United States ; R01 GM138597/GM/NIGMS NIH HHS/United States ; I01 BX004821/BX/BLRD VA/United States ; I01 BX004188/BX/BLRD VA/United States ; },
mesh = {Humans ; *COVID-19/epidemiology/genetics ; Mucin-5B/genetics ; Polymorphism, Genetic ; *Idiopathic Pulmonary Fibrosis/genetics ; Genotype ; Hospitalization ; Genetic Predisposition to Disease/genetics ; },
abstract = {Rationale: A common MUC5B gene polymorphism, rs35705950-T, is associated with idiopathic pulmonary fibrosis (IPF), but its role in severe acute respiratory syndrome coronavirus 2 infection and disease severity is unclear. Objectives: To assess whether rs35705950-T confers differential risk for clinical outcomes associated with coronavirus disease (COVID-19) infection among participants in the Million Veteran Program (MVP). Methods: The MUC5B rs35705950-T allele was directly genotyped among MVP participants; clinical events and comorbidities were extracted from the electronic health records. Associations between the incidence or severity of COVID-19 and rs35705950-T were analyzed within each ancestry group in the MVP followed by transancestry meta-analysis. Replication and joint meta-analysis were conducted using summary statistics from the COVID-19 Host Genetics Initiative (HGI). Sensitivity analyses with adjustment for additional covariates (body mass index, Charlson comorbidity index, smoking, asbestosis, rheumatoid arthritis with interstitial lung disease, and IPF) and associations with post-COVID-19 pneumonia were performed in MVP subjects. Measurements and Main Results: The rs35705950-T allele was associated with fewer COVID-19 hospitalizations in transancestry meta-analyses within the MVP (Ncases = 4,325; Ncontrols = 507,640; OR = 0.89 [0.82-0.97]; P = 6.86 × 10[-3]) and joint meta-analyses with the HGI (Ncases = 13,320; Ncontrols = 1,508,841; OR, 0.90 [0.86-0.95]; P = 8.99 × 10[-5]). The rs35705950-T allele was not associated with reduced COVID-19 positivity in transancestry meta-analysis within the MVP (Ncases = 19,168/Ncontrols = 492,854; OR, 0.98 [0.95-1.01]; P = 0.06) but was nominally significant (P < 0.05) in the joint meta-analysis with the HGI (Ncases = 44,820; Ncontrols = 1,775,827; OR, 0.97 [0.95-1.00]; P = 0.03). Associations were not observed with severe outcomes or mortality. Among individuals of European ancestry in the MVP, rs35705950-T was associated with fewer post-COVID-19 pneumonia events (OR, 0.82 [0.72-0.93]; P = 0.001). Conclusions: The MUC5B variant rs35705950-T may confer protection in COVID-19 hospitalizations.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/epidemiology/genetics
Mucin-5B/genetics
Polymorphism, Genetic
*Idiopathic Pulmonary Fibrosis/genetics
Genotype
Hospitalization
Genetic Predisposition to Disease/genetics
RevDate: 2025-07-28
CmpDate: 2022-07-25
2022 AHA/ACC Key Data Elements and Definitions for Cardiovascular and Noncardiovascular Complications of COVID-19: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Data Standards.
Journal of the American College of Cardiology, 80(4):388-465.
Additional Links: PMID-35753858
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35753858,
year = {2022},
author = {, and Bozkurt, B and Das, SR and Addison, D and Gupta, A and Jneid, H and Khan, SS and Koromia, GA and Kulkarni, PA and LaPoint, K and Lewis, EF and Michos, ED and Peterson, PN and Turagam, MK and Wang, TY and Yancy, CW},
title = {2022 AHA/ACC Key Data Elements and Definitions for Cardiovascular and Noncardiovascular Complications of COVID-19: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Data Standards.},
journal = {Journal of the American College of Cardiology},
volume = {80},
number = {4},
pages = {388-465},
pmid = {35753858},
issn = {1558-3597},
support = {K23 HL155890/HL/NHLBI NIH HHS/United States ; L30 HL149063/HL/NHLBI NIH HHS/United States ; },
mesh = {Advisory Committees ; American Heart Association ; *COVID-19/complications ; *Cardiology ; Heart ; Humans ; United States/epidemiology ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Advisory Committees
American Heart Association
*COVID-19/complications
*Cardiology
Heart
Humans
United States/epidemiology
RevDate: 2025-05-30
CmpDate: 2022-06-06
Prognostic indicators and outcomes of hospitalised COVID-19 patients with neurological disease: An individual patient data meta-analysis.
PloS one, 17(6):e0263595.
BACKGROUND: Neurological COVID-19 disease has been reported widely, but published studies often lack information on neurological outcomes and prognostic risk factors. We aimed to describe the spectrum of neurological disease in hospitalised COVID-19 patients; characterise clinical outcomes; and investigate factors associated with a poor outcome.
METHODS: We conducted an individual patient data (IPD) meta-analysis of hospitalised patients with neurological COVID-19 disease, using standard case definitions. We invited authors of studies from the first pandemic wave, plus clinicians in the Global COVID-Neuro Network with unpublished data, to contribute. We analysed features associated with poor outcome (moderate to severe disability or death, 3 to 6 on the modified Rankin Scale) using multivariable models.
RESULTS: We included 83 studies (31 unpublished) providing IPD for 1979 patients with COVID-19 and acute new-onset neurological disease. Encephalopathy (978 [49%] patients) and cerebrovascular events (506 [26%]) were the most common diagnoses. Respiratory and systemic symptoms preceded neurological features in 93% of patients; one third developed neurological disease after hospital admission. A poor outcome was more common in patients with cerebrovascular events (76% [95% CI 67-82]), than encephalopathy (54% [42-65]). Intensive care use was high (38% [35-41]) overall, and also greater in the cerebrovascular patients. In the cerebrovascular, but not encephalopathic patients, risk factors for poor outcome included breathlessness on admission and elevated D-dimer. Overall, 30-day mortality was 30% [27-32]. The hazard of death was comparatively lower for patients in the WHO European region.
INTERPRETATION: Neurological COVID-19 disease poses a considerable burden in terms of disease outcomes and use of hospital resources from prolonged intensive care and inpatient admission; preliminary data suggest these may differ according to WHO regions and country income levels. The different risk factors for encephalopathy and stroke suggest different disease mechanisms which may be amenable to intervention, especially in those who develop neurological symptoms after hospital admission.
Additional Links: PMID-35653330
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35653330,
year = {2022},
author = {Singh, B and Lant, S and Cividini, S and Cattrall, JWS and Goodwin, LC and Benjamin, L and Michael, BD and Khawaja, A and Matos, AMB and Alkeridy, W and Pilotto, A and Lahiri, D and Rawlinson, R and Mhlanga, S and Lopez, EC and Sargent, BF and Somasundaran, A and Tamborska, A and Webb, G and Younas, K and Al Sami, Y and Babu, H and Banks, T and Cavallieri, F and Cohen, M and Davies, E and Dhar, S and Fajardo Modol, A and Farooq, H and Harte, J and Hey, S and Joseph, A and Karthikappallil, D and Kassahun, D and Lipunga, G and Mason, R and Minton, T and Mond, G and Poxon, J and Rabas, S and Soothill, G and Zedde, M and Yenkoyan, K and Brew, B and Contini, E and Cysique, L and Zhang, X and Maggi, P and van Pesch, V and Lechien, J and Saussez, S and Heyse, A and Brito Ferreira, ML and Soares, CN and Elicer, I and EugenÃn-von Bernhardi, L and Ñancupil Reyes, W and Yin, R and Azab, MA and Abd-Allah, F and Elkady, A and Escalard, S and Corvol, JC and Delorme, C and Tattevin, P and Bigaut, K and Lorenz, N and Hornuss, D and Hosp, J and Rieg, S and Wagner, D and Knier, B and Lingor, P and Winkler, AS and Sharifi-Razavi, A and Moein, ST and SeyedAlinaghi, S and JamaliMoghadamSiahkali, S and Morassi, M and Padovani, A and Giunta, M and Libri, I and Beretta, S and Ravaglia, S and Foschi, M and Calabresi, P and Primiano, G and Servidei, S and Biagio Mercuri, N and Liguori, C and Pierantozzi, M and Sarmati, L and Boso, F and Garazzino, S and Mariotto, S and Patrick, KN and Costache, O and Pincherle, A and Klok, FA and Meza, R and Cabreira, V and Valdoleiros, SR and Oliveira, V and Kaimovsky, I and Guekht, A and Koh, J and Fernández DÃaz, E and Barrios-López, JM and Guijarro-Castro, C and Beltrán-Corbellini, Á and MartÃnez-Poles, J and Diezma-MartÃn, AM and Morales-Casado, MI and GarcÃa GarcÃa, S and Breville, G and Coen, M and Uginet, M and Bernard-Valnet, R and Du Pasquier, R and Kaya, Y and Abdelnour, LH and Rice, C and Morrison, H and Defres, S and Huda, S and Enright, N and Hassell, J and D'Anna, L and Benger, M and Sztriha, L and Raith, E and Chinthapalli, K and Nortley, R and Paterson, R and Chandratheva, A and Werring, DJ and Dervisevic, S and Harkness, K and Pinto, A and Jillella, D and Beach, S and Gunasekaran, K and Rocha Ferreira Da Silva, I and Nalleballe, K and Santoro, J and Scullen, T and Kahn, L and Kim, CY and Thakur, KT and Jain, R and Umapathi, T and Nicholson, TR and Sejvar, JJ and Hodel, EM and , and Tudur Smith, C and Solomon, T},
title = {Prognostic indicators and outcomes of hospitalised COVID-19 patients with neurological disease: An individual patient data meta-analysis.},
journal = {PloS one},
volume = {17},
number = {6},
pages = {e0263595},
pmid = {35653330},
issn = {1932-6203},
support = {MR/V033441/1/MRC_/Medical Research Council/United Kingdom ; 17/63/110/DH_/Department of Health/United Kingdom ; NIHR200907/DH_/Department of Health/United Kingdom ; },
mesh = {*COVID-19/complications/therapy ; Hospitalization ; Humans ; Prognosis ; Risk Factors ; *Stroke ; },
abstract = {BACKGROUND: Neurological COVID-19 disease has been reported widely, but published studies often lack information on neurological outcomes and prognostic risk factors. We aimed to describe the spectrum of neurological disease in hospitalised COVID-19 patients; characterise clinical outcomes; and investigate factors associated with a poor outcome.
METHODS: We conducted an individual patient data (IPD) meta-analysis of hospitalised patients with neurological COVID-19 disease, using standard case definitions. We invited authors of studies from the first pandemic wave, plus clinicians in the Global COVID-Neuro Network with unpublished data, to contribute. We analysed features associated with poor outcome (moderate to severe disability or death, 3 to 6 on the modified Rankin Scale) using multivariable models.
RESULTS: We included 83 studies (31 unpublished) providing IPD for 1979 patients with COVID-19 and acute new-onset neurological disease. Encephalopathy (978 [49%] patients) and cerebrovascular events (506 [26%]) were the most common diagnoses. Respiratory and systemic symptoms preceded neurological features in 93% of patients; one third developed neurological disease after hospital admission. A poor outcome was more common in patients with cerebrovascular events (76% [95% CI 67-82]), than encephalopathy (54% [42-65]). Intensive care use was high (38% [35-41]) overall, and also greater in the cerebrovascular patients. In the cerebrovascular, but not encephalopathic patients, risk factors for poor outcome included breathlessness on admission and elevated D-dimer. Overall, 30-day mortality was 30% [27-32]. The hazard of death was comparatively lower for patients in the WHO European region.
INTERPRETATION: Neurological COVID-19 disease poses a considerable burden in terms of disease outcomes and use of hospital resources from prolonged intensive care and inpatient admission; preliminary data suggest these may differ according to WHO regions and country income levels. The different risk factors for encephalopathy and stroke suggest different disease mechanisms which may be amenable to intervention, especially in those who develop neurological symptoms after hospital admission.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/complications/therapy
Hospitalization
Humans
Prognosis
Risk Factors
*Stroke
RevDate: 2025-07-28
CmpDate: 2022-06-28
Distributed Quasi-Poisson regression algorithm for modeling multi-site count outcomes in distributed data networks.
Journal of biomedical informatics, 131:104097.
BACKGROUND: Observational studies incorporating real-world data from multiple institutions facilitate study of rare outcomes or exposures and improve generalizability of results. Due to privacy concerns surrounding patient-level data sharing across institutions, methods for performing regression analyses distributively are desirable. Meta-analysis of institution-specific estimates is commonly used, but has been shown to produce biased estimates in certain settings. While distributed regression methods are increasingly available, methods for analyzing count outcomes are currently limited. Count data in practice are commonly subject to overdispersion, exhibiting greater variability than expected under a given statistical model.
OBJECTIVE: We propose a novel computational method, a one-shot distributed algorithm for quasi-Poisson regression (ODAP), to distributively model count outcomes while accounting for overdispersion.
METHODS: ODAP incorporates a surrogate likelihood approach to perform distributed quasi-Poisson regression without requiring patient-level data sharing, only requiring sharing of aggregate data from each participating institution. ODAP requires at most three rounds of non-iterative communication among institutions to generate coefficient estimates and corresponding standard errors. In simulations, we evaluate ODAP under several data scenarios possible in multi-site analyses, comparing ODAP and meta-analysis estimates in terms of error relative to pooled regression estimates, considered the gold standard. In a proof-of-concept real-world data analysis, we similarly compare ODAP and meta-analysis in terms of relative error to pooled estimatation using data from the OneFlorida Clinical Research Consortium, modeling length of stay in COVID-19 patients as a function of various patient characteristics. In a second proof-of-concept analysis, using the same outcome and covariates, we incorporate data from the UnitedHealth Group Clinical Discovery Database together with the OneFlorida data in a distributed analysis to compare estimates produced by ODAP and meta-analysis.
RESULTS: In simulations, ODAP exhibited negligible error relative to pooled regression estimates across all settings explored. Meta-analysis estimates, while largely unbiased, were increasingly variable as heterogeneity in the outcome increased across institutions. When baseline expected count was 0.2, relative error for meta-analysis was above 5% in 25% of iterations (250/1000), while the largest relative error for ODAP in any iteration was 3.59%. In our proof-of-concept analysis using only OneFlorida data, ODAP estimates were closer to pooled regression estimates than those produced by meta-analysis for all 15 covariates. In our distributed analysis incorporating data from both OneFlorida and the UnitedHealth Group Clinical Discovery Database, ODAP and meta-analysis estimates were largely similar, while some differences in estimates (as large as 13.8%) could be indicative of bias in meta-analytic estimates.
CONCLUSIONS: ODAP performs privacy-preserving, communication-efficient distributed quasi-Poisson regression to analyze count outcomes using data stored within multiple institutions. Our method produces estimates nearly matching pooled regression estimates and sometimes more accurate than meta-analysis estimates, most notably in settings with relatively low counts and high outcome heterogeneity across institutions.
Additional Links: PMID-35643272
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35643272,
year = {2022},
author = {Edmondson, MJ and Luo, C and Nazmul Islam, M and Sheils, NE and Buresh, J and Chen, Z and Bian, J and Chen, Y},
title = {Distributed Quasi-Poisson regression algorithm for modeling multi-site count outcomes in distributed data networks.},
journal = {Journal of biomedical informatics},
volume = {131},
number = {},
pages = {104097},
pmid = {35643272},
issn = {1532-0480},
support = {R56 AG069880/AG/NIA NIH HHS/United States ; U18 DP006512/DP/NCCDPHP CDC HHS/United States ; R01 LM013519/LM/NLM NIH HHS/United States ; R56 AG074604/AG/NIA NIH HHS/United States ; R01 AG073435/AG/NIA NIH HHS/United States ; R01 AG076234/AG/NIA NIH HHS/United States ; R01 CA246418/CA/NCI NIH HHS/United States ; },
mesh = {Algorithms ; *COVID-19/epidemiology ; Humans ; Likelihood Functions ; Models, Statistical ; Regression Analysis ; },
abstract = {BACKGROUND: Observational studies incorporating real-world data from multiple institutions facilitate study of rare outcomes or exposures and improve generalizability of results. Due to privacy concerns surrounding patient-level data sharing across institutions, methods for performing regression analyses distributively are desirable. Meta-analysis of institution-specific estimates is commonly used, but has been shown to produce biased estimates in certain settings. While distributed regression methods are increasingly available, methods for analyzing count outcomes are currently limited. Count data in practice are commonly subject to overdispersion, exhibiting greater variability than expected under a given statistical model.
OBJECTIVE: We propose a novel computational method, a one-shot distributed algorithm for quasi-Poisson regression (ODAP), to distributively model count outcomes while accounting for overdispersion.
METHODS: ODAP incorporates a surrogate likelihood approach to perform distributed quasi-Poisson regression without requiring patient-level data sharing, only requiring sharing of aggregate data from each participating institution. ODAP requires at most three rounds of non-iterative communication among institutions to generate coefficient estimates and corresponding standard errors. In simulations, we evaluate ODAP under several data scenarios possible in multi-site analyses, comparing ODAP and meta-analysis estimates in terms of error relative to pooled regression estimates, considered the gold standard. In a proof-of-concept real-world data analysis, we similarly compare ODAP and meta-analysis in terms of relative error to pooled estimatation using data from the OneFlorida Clinical Research Consortium, modeling length of stay in COVID-19 patients as a function of various patient characteristics. In a second proof-of-concept analysis, using the same outcome and covariates, we incorporate data from the UnitedHealth Group Clinical Discovery Database together with the OneFlorida data in a distributed analysis to compare estimates produced by ODAP and meta-analysis.
RESULTS: In simulations, ODAP exhibited negligible error relative to pooled regression estimates across all settings explored. Meta-analysis estimates, while largely unbiased, were increasingly variable as heterogeneity in the outcome increased across institutions. When baseline expected count was 0.2, relative error for meta-analysis was above 5% in 25% of iterations (250/1000), while the largest relative error for ODAP in any iteration was 3.59%. In our proof-of-concept analysis using only OneFlorida data, ODAP estimates were closer to pooled regression estimates than those produced by meta-analysis for all 15 covariates. In our distributed analysis incorporating data from both OneFlorida and the UnitedHealth Group Clinical Discovery Database, ODAP and meta-analysis estimates were largely similar, while some differences in estimates (as large as 13.8%) could be indicative of bias in meta-analytic estimates.
CONCLUSIONS: ODAP performs privacy-preserving, communication-efficient distributed quasi-Poisson regression to analyze count outcomes using data stored within multiple institutions. Our method produces estimates nearly matching pooled regression estimates and sometimes more accurate than meta-analysis estimates, most notably in settings with relatively low counts and high outcome heterogeneity across institutions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Algorithms
*COVID-19/epidemiology
Humans
Likelihood Functions
Models, Statistical
Regression Analysis
RevDate: 2022-11-03
CmpDate: 2022-04-25
Meta-analysis of the robustness of COVID-19 diagnostic kit performance during the early pandemic.
BMJ open, 12(4):e053912.
BACKGROUND: Accurate detection of SARS-CoV-2 is necessary to mitigate the COVID-19 pandemic. However, the test reagents and assay platforms are varied and may not be sufficiently robust to diagnose COVID-19.
METHODS: We reviewed 85 studies (21 530 patients), published from five regions of the world, to highlight issues involved in the diagnosis of COVID-19 in the early phase of the pandemic. All relevant articles, published up to 31 May 2020, in PubMed, BioRiXv, MedRiXv and Google Scholar, were included. We evaluated the qualitative (9749 patients) and quantitative (10 355 patients) performance of RT-PCR and serologic diagnostic tests for real-world samples, and assessed the concordance (5538 patients) between test performance in meta-analyses. Synthesis of results was done using random effects modelling and bias was evaluated according to QUADAS-2 guidelines.
RESULTS: The RT-PCR tests exhibited heterogeneity in the primers and reagents used. Of 1957 positive RT-PCR COVID-19 participants, 1585 had positive serum antibody (IgM±IgG) tests (sensitivity 0.81, 95% CI 0.66 to 0.90). While 3509 of 3581 participants RT-PCR negative for COVID-19 were found negative by serology testing (specificity 0.98, 95% CI 0.94 to 0.99). The chemiluminescent immunoassay exhibited the highest sensitivity, followed by ELISA and lateral flow immunoassays. Serology tests had higher sensitivity and specificity for laboratory approval than for real-world reporting data.
DISCUSSION: The robustness of the assays/platforms is influenced by variability in sampling and reagents. Serological testing complements and may minimise false negative RT-PCR results. Lack of standardised assay protocols in the early phase of pandemic might have contributed to the spread of COVID-19.
Additional Links: PMID-35450897
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35450897,
year = {2022},
author = {Shanmugam, C and Behring, M and Luthra, V and Leal, SM and Varambally, S and Netto, GJ and Manne, U},
title = {Meta-analysis of the robustness of COVID-19 diagnostic kit performance during the early pandemic.},
journal = {BMJ open},
volume = {12},
number = {4},
pages = {e053912},
pmid = {35450897},
issn = {2044-6055},
support = {U54 CA118948/CA/NCI NIH HHS/United States ; R25 CA047888/CA/NCI NIH HHS/United States ; },
mesh = {Antibodies, Viral ; *COVID-19/diagnosis/epidemiology ; COVID-19 Testing ; Humans ; Pandemics ; SARS-CoV-2 ; Sensitivity and Specificity ; },
abstract = {BACKGROUND: Accurate detection of SARS-CoV-2 is necessary to mitigate the COVID-19 pandemic. However, the test reagents and assay platforms are varied and may not be sufficiently robust to diagnose COVID-19.
METHODS: We reviewed 85 studies (21 530 patients), published from five regions of the world, to highlight issues involved in the diagnosis of COVID-19 in the early phase of the pandemic. All relevant articles, published up to 31 May 2020, in PubMed, BioRiXv, MedRiXv and Google Scholar, were included. We evaluated the qualitative (9749 patients) and quantitative (10 355 patients) performance of RT-PCR and serologic diagnostic tests for real-world samples, and assessed the concordance (5538 patients) between test performance in meta-analyses. Synthesis of results was done using random effects modelling and bias was evaluated according to QUADAS-2 guidelines.
RESULTS: The RT-PCR tests exhibited heterogeneity in the primers and reagents used. Of 1957 positive RT-PCR COVID-19 participants, 1585 had positive serum antibody (IgM±IgG) tests (sensitivity 0.81, 95% CI 0.66 to 0.90). While 3509 of 3581 participants RT-PCR negative for COVID-19 were found negative by serology testing (specificity 0.98, 95% CI 0.94 to 0.99). The chemiluminescent immunoassay exhibited the highest sensitivity, followed by ELISA and lateral flow immunoassays. Serology tests had higher sensitivity and specificity for laboratory approval than for real-world reporting data.
DISCUSSION: The robustness of the assays/platforms is influenced by variability in sampling and reagents. Serological testing complements and may minimise false negative RT-PCR results. Lack of standardised assay protocols in the early phase of pandemic might have contributed to the spread of COVID-19.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Antibodies, Viral
*COVID-19/diagnosis/epidemiology
COVID-19 Testing
Humans
Pandemics
SARS-CoV-2
Sensitivity and Specificity
RevDate: 2025-05-30
CmpDate: 2022-05-04
Genetic Landscape of the ACE2 Coronavirus Receptor.
Circulation, 145(18):1398-1411.
BACKGROUND: SARS-CoV-2, the causal agent of COVID-19, enters human cells using the ACE2 (angiotensin-converting enzyme 2) protein as a receptor. ACE2 is thus key to the infection and treatment of the coronavirus. ACE2 is highly expressed in the heart and respiratory and gastrointestinal tracts, playing important regulatory roles in the cardiovascular and other biological systems. However, the genetic basis of the ACE2 protein levels is not well understood.
METHODS: We have conducted the largest genome-wide association meta-analysis of plasma ACE2 levels in >28 000 individuals of the SCALLOP Consortium (Systematic and Combined Analysis of Olink Proteins). We summarize the cross-sectional epidemiological correlates of circulating ACE2. Using the summary statistics-based high-definition likelihood method, we estimate relevant genetic correlations with cardiometabolic phenotypes, COVID-19, and other human complex traits and diseases. We perform causal inference of soluble ACE2 on vascular disease outcomes and COVID-19 severity using mendelian randomization. We also perform in silico functional analysis by integrating with other types of omics data.
RESULTS: We identified 10 loci, including 8 novel, capturing 30% of the heritability of the protein. We detected that plasma ACE2 was genetically correlated with vascular diseases, severe COVID-19, and a wide range of human complex diseases and medications. An X-chromosome cis-protein quantitative trait loci-based mendelian randomization analysis suggested a causal effect of elevated ACE2 levels on COVID-19 severity (odds ratio, 1.63 [95% CI, 1.10-2.42]; P=0.01), hospitalization (odds ratio, 1.52 [95% CI, 1.05-2.21]; P=0.03), and infection (odds ratio, 1.60 [95% CI, 1.08-2.37]; P=0.02). Tissue- and cell type-specific transcriptomic and epigenomic analysis revealed that the ACE2 regulatory variants were enriched for DNA methylation sites in blood immune cells.
CONCLUSIONS: Human plasma ACE2 shares a genetic basis with cardiovascular disease, COVID-19, and other related diseases. The genetic architecture of the ACE2 protein is mapped, providing a useful resource for further biological and clinical studies on this coronavirus receptor.
Additional Links: PMID-35387486
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35387486,
year = {2022},
author = {Yang, Z and Macdonald-Dunlop, E and Chen, J and Zhai, R and Li, T and Richmond, A and Klarić, L and Pirastu, N and Ning, Z and Zheng, C and Wang, Y and Huang, T and He, Y and Guo, H and Ying, K and Gustafsson, S and Prins, B and Ramisch, A and Dermitzakis, ET and Png, G and Eriksson, N and Haessler, J and Hu, X and Zanetti, D and Boutin, T and Hwang, SJ and Wheeler, E and Pietzner, M and Raffield, LM and Kalnapenkis, A and Peters, JE and Viñuela, A and Gilly, A and Elmståhl, S and Dedoussis, G and Petrie, JR and Polašek, O and Folkersen, L and Chen, Y and Yao, C and Võsa, U and Pairo-Castineira, E and Clohisey, S and Bretherick, AD and Rawlik, K and , and , and Esko, T and Enroth, S and Johansson, Šand Gyllensten, U and Langenberg, C and Levy, D and Hayward, C and Assimes, TL and Kooperberg, C and Manichaikul, AW and Siegbahn, A and Wallentin, L and Lind, L and Zeggini, E and Schwenk, JM and Butterworth, AS and Michaëlsson, K and Pawitan, Y and Joshi, PK and Baillie, JK and Mälarstig, A and Reiner, AP and Wilson, JF and Shen, X},
title = {Genetic Landscape of the ACE2 Coronavirus Receptor.},
journal = {Circulation},
volume = {145},
number = {18},
pages = {1398-1411},
pmid = {35387486},
issn = {1524-4539},
support = {MC_UU_00007/10/MRC_/Medical Research Council/United Kingdom ; RG/18/13/33946/BHF_/British Heart Foundation/United Kingdom ; RG/13/13/30194/BHF_/British Heart Foundation/United Kingdom ; MR/R026408/1/MRC_/Medical Research Council/United Kingdom ; MC_UU_12015/1/MRC_/Medical Research Council/United Kingdom ; R01 HL146500/HL/NHLBI NIH HHS/United States ; CH/12/2/29428/BHF_/British Heart Foundation/United Kingdom ; MC_UU_00006/1/MRC_/Medical Research Council/United Kingdom ; 204979/Z/16/Z/WT_/Wellcome Trust/United Kingdom ; R01 DK114183/DK/NIDDK NIH HHS/United States ; R01 HL132947/HL/NHLBI NIH HHS/United States ; },
mesh = {*Angiotensin-Converting Enzyme 2/genetics ; *COVID-19/genetics ; Cross-Sectional Studies ; Genome-Wide Association Study ; Humans ; Receptors, Coronavirus ; SARS-CoV-2 ; },
abstract = {BACKGROUND: SARS-CoV-2, the causal agent of COVID-19, enters human cells using the ACE2 (angiotensin-converting enzyme 2) protein as a receptor. ACE2 is thus key to the infection and treatment of the coronavirus. ACE2 is highly expressed in the heart and respiratory and gastrointestinal tracts, playing important regulatory roles in the cardiovascular and other biological systems. However, the genetic basis of the ACE2 protein levels is not well understood.
METHODS: We have conducted the largest genome-wide association meta-analysis of plasma ACE2 levels in >28 000 individuals of the SCALLOP Consortium (Systematic and Combined Analysis of Olink Proteins). We summarize the cross-sectional epidemiological correlates of circulating ACE2. Using the summary statistics-based high-definition likelihood method, we estimate relevant genetic correlations with cardiometabolic phenotypes, COVID-19, and other human complex traits and diseases. We perform causal inference of soluble ACE2 on vascular disease outcomes and COVID-19 severity using mendelian randomization. We also perform in silico functional analysis by integrating with other types of omics data.
RESULTS: We identified 10 loci, including 8 novel, capturing 30% of the heritability of the protein. We detected that plasma ACE2 was genetically correlated with vascular diseases, severe COVID-19, and a wide range of human complex diseases and medications. An X-chromosome cis-protein quantitative trait loci-based mendelian randomization analysis suggested a causal effect of elevated ACE2 levels on COVID-19 severity (odds ratio, 1.63 [95% CI, 1.10-2.42]; P=0.01), hospitalization (odds ratio, 1.52 [95% CI, 1.05-2.21]; P=0.03), and infection (odds ratio, 1.60 [95% CI, 1.08-2.37]; P=0.02). Tissue- and cell type-specific transcriptomic and epigenomic analysis revealed that the ACE2 regulatory variants were enriched for DNA methylation sites in blood immune cells.
CONCLUSIONS: Human plasma ACE2 shares a genetic basis with cardiovascular disease, COVID-19, and other related diseases. The genetic architecture of the ACE2 protein is mapped, providing a useful resource for further biological and clinical studies on this coronavirus receptor.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Angiotensin-Converting Enzyme 2/genetics
*COVID-19/genetics
Cross-Sectional Studies
Genome-Wide Association Study
Humans
Receptors, Coronavirus
SARS-CoV-2
RevDate: 2023-04-12
CmpDate: 2022-05-25
Protective trend of anti-androgen therapy during the COVID-19 pandemic: A meta-analysis.
The Journal of infection, 84(6):834-872.
Additional Links: PMID-35341831
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35341831,
year = {2022},
author = {Meng, J and Ge, Q and Li, J and Lu, X and Chen, Y and Wang, H and Zhang, M and Du, J and Zhang, L and Hao, Z and Liang, C},
title = {Protective trend of anti-androgen therapy during the COVID-19 pandemic: A meta-analysis.},
journal = {The Journal of infection},
volume = {84},
number = {6},
pages = {834-872},
pmid = {35341831},
issn = {1532-2742},
mesh = {*COVID-19 ; Humans ; Pandemics ; SARS-CoV-2 ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19
Humans
Pandemics
SARS-CoV-2
RevDate: 2022-02-19
CmpDate: 2022-02-16
Meta-Analysis of APP Expression Modulated by SARS-CoV-2 Infection via the ACE2 Receptor.
International journal of molecular sciences, 23(3):.
Alzheimer's disease (AD) is characterized by the deposition of amyloid-beta (Aβ) plaques from improper amyloid-beta precursor protein (APP) cleavage. Following studies of inflammation caused by coronavirus-2019 (COVID-19) infection, this study investigated the impact of COVID-19 on APP expression. A meta-analysis was conducted utilizing QIAGEN Ingenuity Pathway Analysis (IPA) to examine the link between severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) and the modulation of APP expression upon virus binding the Angiotensin-converting enzyme-2 (ACE2) receptor. A Core Analysis was run on the infection by severe acute respiratory syndrome (SARS) coronavirus node, which included molecules affected by SARS-CoV-2, revealing its upstream regulators. Intermediary molecules were found between the upstream regulators and ACE2 and between ACE2 and APP. Activation of the upstream regulators downregulated the expression of ACE2 with a Z-score of -1.719 (p-value = 0.086) and upregulated APP with a Z-score of 1.898 (p-value = 0.058), showing a less than 10% chance of the results occurring by chance and pointing to an inverse relationship between ACE2 and APP expression. The neuroinflammation signaling pathway was the fifth top canonical pathway involved in APP upregulation. The study results suggest that ACE2 could be downregulated by SARS-CoV-2, resulting in APP upregulation, and potentially exacerbating the onset and progression of AD.
Additional Links: PMID-35163117
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35163117,
year = {2022},
author = {Caradonna, A and Patel, T and Toleska, M and Alabed, S and Chang, SL},
title = {Meta-Analysis of APP Expression Modulated by SARS-CoV-2 Infection via the ACE2 Receptor.},
journal = {International journal of molecular sciences},
volume = {23},
number = {3},
pages = {},
pmid = {35163117},
issn = {1422-0067},
support = {DA046258/NH/NIH HHS/United States ; },
mesh = {Amyloid beta-Protein Precursor/*genetics/metabolism ; Angiotensin-Converting Enzyme 2/genetics/*metabolism ; COVID-19/*genetics/metabolism/pathology ; Gene Expression Regulation ; Humans ; Plaque, Amyloid/genetics/metabolism/pathology ; SARS-CoV-2/metabolism/pathogenicity/*physiology ; Signal Transduction/genetics ; },
abstract = {Alzheimer's disease (AD) is characterized by the deposition of amyloid-beta (Aβ) plaques from improper amyloid-beta precursor protein (APP) cleavage. Following studies of inflammation caused by coronavirus-2019 (COVID-19) infection, this study investigated the impact of COVID-19 on APP expression. A meta-analysis was conducted utilizing QIAGEN Ingenuity Pathway Analysis (IPA) to examine the link between severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) and the modulation of APP expression upon virus binding the Angiotensin-converting enzyme-2 (ACE2) receptor. A Core Analysis was run on the infection by severe acute respiratory syndrome (SARS) coronavirus node, which included molecules affected by SARS-CoV-2, revealing its upstream regulators. Intermediary molecules were found between the upstream regulators and ACE2 and between ACE2 and APP. Activation of the upstream regulators downregulated the expression of ACE2 with a Z-score of -1.719 (p-value = 0.086) and upregulated APP with a Z-score of 1.898 (p-value = 0.058), showing a less than 10% chance of the results occurring by chance and pointing to an inverse relationship between ACE2 and APP expression. The neuroinflammation signaling pathway was the fifth top canonical pathway involved in APP upregulation. The study results suggest that ACE2 could be downregulated by SARS-CoV-2, resulting in APP upregulation, and potentially exacerbating the onset and progression of AD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Amyloid beta-Protein Precursor/*genetics/metabolism
Angiotensin-Converting Enzyme 2/genetics/*metabolism
COVID-19/*genetics/metabolism/pathology
Gene Expression Regulation
Humans
Plaque, Amyloid/genetics/metabolism/pathology
SARS-CoV-2/metabolism/pathogenicity/*physiology
Signal Transduction/genetics
RevDate: 2025-05-30
CmpDate: 2022-06-20
Mendelian Randomization Analysis Reveals No Causal Relationship Between Nonalcoholic Fatty Liver Disease and Severe COVID-19.
Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association, 20(7):1553-1560.e78.
BACKGROUND & AIMS: The coronavirus disease 2019 (COVID-19) pandemic has witnessed more than 4.5 million deaths as of the time of writing. Whether nonalcoholic fatty liver disease (NAFLD) increases the risk for severe COVID-19 remains unclear. We sought to address this question using 2-sample Mendelian randomization (TSMR) analysis approaches in large cohorts.
METHODS: We performed large-scale TSMR analyses to examine whether there is a causal relationship between NAFLD, serum alanine aminotransferase, grade of steatosis, NAFLD Activity Score, or fibrosis stage and severe COVID-19. To maximize the power of this analysis, we performed a genome-wide meta-analysis to identify single nucleotide polymorphisms associated with NAFLD. We also examined the impact of 20 major comorbid factors of NAFLD on severe COVID-19.
RESULTS: Univariate analysis of the UK Biobank data demonstrated a significant association between NAFLD and severe COVID-19 (odds ratio [OR], 3.06; P = 1.07 × 10[-6]). However, this association disappeared after demographic and comorbid factors were adjusted (OR, 1.57; P = .09). TSMR study indicated that NAFLD (OR, 0.97; P = .61), alanine aminotransferase level (OR, 1.03; P = .47), grade of steatosis (OR, 1.08; P = .41), NAFLD Activity Score (OR, 1.02; P = .39), and fibrosis stage (OR, 1.01; P = .87) were not associated with severe COVID-19. Among all NAFLD-related comorbid factors, body mass index (OR, 1.73; P = 7.65 × 10[-9]), waist circumference (OR, 1.76; P = 2.58 × 10[-5]), and hip circumference (OR, 1.33; P = 7.26 × 10[-3]) were the only ones demonstrated a causal impact on severe COVID-19.
CONCLUSIONS: There is no evidence supporting that NAFLD is a causal risk factor for severe COVID-19. Previous observational associations between NAFLD and COVID-19 are likely attributed to the correlation between NAFLD and obesity.
Additional Links: PMID-35124268
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35124268,
year = {2022},
author = {Li, J and Tian, A and Zhu, H and Chen, L and Wen, J and Liu, W and Chen, P},
title = {Mendelian Randomization Analysis Reveals No Causal Relationship Between Nonalcoholic Fatty Liver Disease and Severe COVID-19.},
journal = {Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association},
volume = {20},
number = {7},
pages = {1553-1560.e78},
pmid = {35124268},
issn = {1542-7714},
support = {MC_PC_17228/MRC_/Medical Research Council/United Kingdom ; MC_QA137853/MRC_/Medical Research Council/United Kingdom ; R01 DK106540/DK/NIDDK NIH HHS/United States ; R01 DK124612/DK/NIDDK NIH HHS/United States ; },
mesh = {Alanine Transaminase ; Body Mass Index ; *COVID-19/complications ; Fibrosis ; Humans ; Mendelian Randomization Analysis ; *Non-alcoholic Fatty Liver Disease/complications/genetics ; },
abstract = {BACKGROUND & AIMS: The coronavirus disease 2019 (COVID-19) pandemic has witnessed more than 4.5 million deaths as of the time of writing. Whether nonalcoholic fatty liver disease (NAFLD) increases the risk for severe COVID-19 remains unclear. We sought to address this question using 2-sample Mendelian randomization (TSMR) analysis approaches in large cohorts.
METHODS: We performed large-scale TSMR analyses to examine whether there is a causal relationship between NAFLD, serum alanine aminotransferase, grade of steatosis, NAFLD Activity Score, or fibrosis stage and severe COVID-19. To maximize the power of this analysis, we performed a genome-wide meta-analysis to identify single nucleotide polymorphisms associated with NAFLD. We also examined the impact of 20 major comorbid factors of NAFLD on severe COVID-19.
RESULTS: Univariate analysis of the UK Biobank data demonstrated a significant association between NAFLD and severe COVID-19 (odds ratio [OR], 3.06; P = 1.07 × 10[-6]). However, this association disappeared after demographic and comorbid factors were adjusted (OR, 1.57; P = .09). TSMR study indicated that NAFLD (OR, 0.97; P = .61), alanine aminotransferase level (OR, 1.03; P = .47), grade of steatosis (OR, 1.08; P = .41), NAFLD Activity Score (OR, 1.02; P = .39), and fibrosis stage (OR, 1.01; P = .87) were not associated with severe COVID-19. Among all NAFLD-related comorbid factors, body mass index (OR, 1.73; P = 7.65 × 10[-9]), waist circumference (OR, 1.76; P = 2.58 × 10[-5]), and hip circumference (OR, 1.33; P = 7.26 × 10[-3]) were the only ones demonstrated a causal impact on severe COVID-19.
CONCLUSIONS: There is no evidence supporting that NAFLD is a causal risk factor for severe COVID-19. Previous observational associations between NAFLD and COVID-19 are likely attributed to the correlation between NAFLD and obesity.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Alanine Transaminase
Body Mass Index
*COVID-19/complications
Fibrosis
Humans
Mendelian Randomization Analysis
*Non-alcoholic Fatty Liver Disease/complications/genetics
RevDate: 2025-07-20
CmpDate: 2022-02-03
Association of Convalescent Plasma Treatment With Clinical Status in Patients Hospitalized With COVID-19: A Meta-analysis.
JAMA network open, 5(1):e2147331.
IMPORTANCE: COVID-19 convalescent plasma (CCP) is a potentially beneficial treatment for COVID-19 that requires rigorous testing.
OBJECTIVE: To compile individual patient data from randomized clinical trials of CCP and to monitor the data until completion or until accumulated evidence enables reliable conclusions regarding the clinical outcomes associated with CCP.
DATA SOURCES: From May to August 2020, a systematic search was performed for trials of CCP in the literature, clinical trial registry sites, and medRxiv. Domain experts at local, national, and international organizations were consulted regularly.
STUDY SELECTION: Eligible trials enrolled hospitalized patients with confirmed COVID-19, not receiving mechanical ventilation, and randomized them to CCP or control. The administered CCP was required to have measurable antibodies assessed locally.
DATA EXTRACTION AND SYNTHESIS: A minimal data set was submitted regularly via a secure portal, analyzed using a prespecified bayesian statistical plan, and reviewed frequently by a collective data and safety monitoring board.
MAIN OUTCOMES AND MEASURES: Prespecified coprimary end points-the World Health Organization (WHO) 11-point ordinal scale analyzed using a proportional odds model and a binary indicator of WHO score of 7 or higher capturing the most severe outcomes including mechanical ventilation through death and analyzed using a logistic model-were assessed clinically at 14 days after randomization.
RESULTS: Eight international trials collectively enrolled 2369 participants (1138 randomized to control and 1231 randomized to CCP). A total of 2341 participants (median [IQR] age, 60 [50-72] years; 845 women [35.7%]) had primary outcome data as of April 2021. The median (IQR) of the ordinal WHO scale was 3 (3-6); the cumulative OR was 0.94 (95% credible interval [CrI], 0.74-1.19; posterior probability of OR <1 of 71%). A total of 352 patients (15%) had WHO score greater than or equal to 7; the OR was 0.94 (95% CrI, 0.69-1.30; posterior probability of OR <1 of 65%). Adjusted for baseline covariates, the ORs for mortality were 0.88 at day 14 (95% CrI, 0.61-1.26; posterior probability of OR <1 of 77%) and 0.85 at day 28 (95% CrI, 0.62-1.18; posterior probability of OR <1 of 84%). Heterogeneity of treatment effect sizes was observed across an array of baseline characteristics.
CONCLUSIONS AND RELEVANCE: This meta-analysis found no association of CCP with better clinical outcomes for the typical patient. These findings suggest that real-time individual patient data pooling and meta-analysis during a pandemic are feasible, offering a model for future research and providing a rich data resource.
Additional Links: PMID-35076699
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35076699,
year = {2022},
author = {Troxel, AB and Petkova, E and Goldfeld, K and Liu, M and Tarpey, T and Wu, Y and Wu, D and Agarwal, A and Avendaño-Solá, C and Bainbridge, E and Bar, KJ and Devos, T and Duarte, RF and Gharbharan, A and Hsue, PY and Kumar, G and Luetkemeyer, AF and Meyfroidt, G and Nicola, AM and Mukherjee, A and Ortigoza, MB and Pirofski, LA and Rijnders, BJA and Rokx, C and Sancho-Lopez, A and Shaw, P and Tebas, P and Yoon, HA and Grudzen, C and Hochman, J and Antman, EM},
title = {Association of Convalescent Plasma Treatment With Clinical Status in Patients Hospitalized With COVID-19: A Meta-analysis.},
journal = {JAMA network open},
volume = {5},
number = {1},
pages = {e2147331},
pmid = {35076699},
issn = {2574-3805},
support = {T32 AI060530/AI/NIAID NIH HHS/United States ; UL1 TR001445/TR/NCATS NIH HHS/United States ; UL1 TR002556/TR/NCATS NIH HHS/United States ; },
mesh = {Aged ; Bayes Theorem ; COVID-19/*therapy ; Female ; *Hospitalization ; Humans ; Immunization, Passive ; Male ; Middle Aged ; *Pandemics ; *Patient Selection ; *Plasma ; Respiration, Artificial ; SARS-CoV-2 ; Severity of Illness Index ; Treatment Outcome ; World Health Organization ; COVID-19 Serotherapy ; },
abstract = {IMPORTANCE: COVID-19 convalescent plasma (CCP) is a potentially beneficial treatment for COVID-19 that requires rigorous testing.
OBJECTIVE: To compile individual patient data from randomized clinical trials of CCP and to monitor the data until completion or until accumulated evidence enables reliable conclusions regarding the clinical outcomes associated with CCP.
DATA SOURCES: From May to August 2020, a systematic search was performed for trials of CCP in the literature, clinical trial registry sites, and medRxiv. Domain experts at local, national, and international organizations were consulted regularly.
STUDY SELECTION: Eligible trials enrolled hospitalized patients with confirmed COVID-19, not receiving mechanical ventilation, and randomized them to CCP or control. The administered CCP was required to have measurable antibodies assessed locally.
DATA EXTRACTION AND SYNTHESIS: A minimal data set was submitted regularly via a secure portal, analyzed using a prespecified bayesian statistical plan, and reviewed frequently by a collective data and safety monitoring board.
MAIN OUTCOMES AND MEASURES: Prespecified coprimary end points-the World Health Organization (WHO) 11-point ordinal scale analyzed using a proportional odds model and a binary indicator of WHO score of 7 or higher capturing the most severe outcomes including mechanical ventilation through death and analyzed using a logistic model-were assessed clinically at 14 days after randomization.
RESULTS: Eight international trials collectively enrolled 2369 participants (1138 randomized to control and 1231 randomized to CCP). A total of 2341 participants (median [IQR] age, 60 [50-72] years; 845 women [35.7%]) had primary outcome data as of April 2021. The median (IQR) of the ordinal WHO scale was 3 (3-6); the cumulative OR was 0.94 (95% credible interval [CrI], 0.74-1.19; posterior probability of OR <1 of 71%). A total of 352 patients (15%) had WHO score greater than or equal to 7; the OR was 0.94 (95% CrI, 0.69-1.30; posterior probability of OR <1 of 65%). Adjusted for baseline covariates, the ORs for mortality were 0.88 at day 14 (95% CrI, 0.61-1.26; posterior probability of OR <1 of 77%) and 0.85 at day 28 (95% CrI, 0.62-1.18; posterior probability of OR <1 of 84%). Heterogeneity of treatment effect sizes was observed across an array of baseline characteristics.
CONCLUSIONS AND RELEVANCE: This meta-analysis found no association of CCP with better clinical outcomes for the typical patient. These findings suggest that real-time individual patient data pooling and meta-analysis during a pandemic are feasible, offering a model for future research and providing a rich data resource.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Aged
Bayes Theorem
COVID-19/*therapy
Female
*Hospitalization
Humans
Immunization, Passive
Male
Middle Aged
*Pandemics
*Patient Selection
*Plasma
Respiration, Artificial
SARS-CoV-2
Severity of Illness Index
Treatment Outcome
World Health Organization
COVID-19 Serotherapy
RevDate: 2025-01-04
CmpDate: 2022-03-14
Ambient air pollution and COVID-19 incidence during four 2020-2021 case surges.
Environmental research, 208:112758.
BACKGROUND: Air pollution exposure may make people more vulnerable to COVID-19 infection. However, previous studies in this area mostly focused on infection before May 2020 and long-term exposure.
OBJECTIVE: To assess both long-term and short-term exposure to air pollution and COVID-19 incidence across four case surges from 03/1/2020 to 02/28/2021.
METHODS: The cohort included 4.6 million members from a large integrated health care system in southern California with comprehensive electronic medical records (EMR). COVID-19 cases were identified from EMR. Incidence of COVID-19 was computed at the census tract-level among members. Prior 1-month and 1-year averaged air pollutant levels (PM2.5, NO2, and O3) at the census tract-level were estimated based on hourly and daily air quality data. Data analyses were conducted by each wave: 3/1/2020-5/31/2020, 6/1/202-9/30/2020, 10/1/2020-12/31/2020, and 1/1/2021-2/28/2021 and pooled across waves using meta-analysis. Generalized linear mixed effects models with Poisson distribution and spatial autocorrelation were used with adjustment for meteorological factors and census tract-level social and health characteristics. Results were expressed as relative risk (RR) per 1 standard deviation.
RESULTS: The cohort included 446,440 COVID-19 cases covering 4609 census tracts. The pooled RRs (95% CI) of COVID-19 incidence associated with 1-year exposures to PM2.5, NO2, and O3 were 1.11 (1.04, 1.18) per 2.3 μg/m[3],1.09 (1.02, 1.17) per 3.2 ppb, and 1.06 (1.00, 1.12) per 5.5 ppb respectively. The corresponding RRs (95% CI) associated with prior 1-month exposures were 1.11 (1.03, 1.20) per 5.2 μg/m[3] for PM2.5, 1.09 (1.01, 1.17) per 6.0 ppb for NO2 and 0.96 (0.85, 1.08) per 12.0 ppb for O3.
CONCLUSION: Long-term PM2.5 and NO2 exposures were associated with increased risk of COVID-19 incidence across all case surges before February 2021. Short-term PM2.5 and NO2 exposures were also associated. Our findings suggest that air pollution may play a role in increasing the risk of COVID-19 infection.
Additional Links: PMID-35063430
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid35063430,
year = {2022},
author = {Sidell, MA and Chen, Z and Huang, BZ and Chow, T and Eckel, SP and Martinez, MP and Lurmann, F and Thomas, DC and Gilliland, FD and Xiang, AH},
title = {Ambient air pollution and COVID-19 incidence during four 2020-2021 case surges.},
journal = {Environmental research},
volume = {208},
number = {},
pages = {112758},
pmid = {35063430},
issn = {1096-0953},
support = {P30 ES007048/ES/NIEHS NIH HHS/United States ; R01 ES029963/ES/NIEHS NIH HHS/United States ; T32 CA229110/CA/NCI NIH HHS/United States ; },
mesh = {*Air Pollutants/analysis/toxicity ; *Air Pollution/analysis ; *COVID-19/epidemiology ; Environmental Exposure/analysis ; Humans ; Incidence ; Particulate Matter/analysis/toxicity ; SARS-CoV-2 ; },
abstract = {BACKGROUND: Air pollution exposure may make people more vulnerable to COVID-19 infection. However, previous studies in this area mostly focused on infection before May 2020 and long-term exposure.
OBJECTIVE: To assess both long-term and short-term exposure to air pollution and COVID-19 incidence across four case surges from 03/1/2020 to 02/28/2021.
METHODS: The cohort included 4.6 million members from a large integrated health care system in southern California with comprehensive electronic medical records (EMR). COVID-19 cases were identified from EMR. Incidence of COVID-19 was computed at the census tract-level among members. Prior 1-month and 1-year averaged air pollutant levels (PM2.5, NO2, and O3) at the census tract-level were estimated based on hourly and daily air quality data. Data analyses were conducted by each wave: 3/1/2020-5/31/2020, 6/1/202-9/30/2020, 10/1/2020-12/31/2020, and 1/1/2021-2/28/2021 and pooled across waves using meta-analysis. Generalized linear mixed effects models with Poisson distribution and spatial autocorrelation were used with adjustment for meteorological factors and census tract-level social and health characteristics. Results were expressed as relative risk (RR) per 1 standard deviation.
RESULTS: The cohort included 446,440 COVID-19 cases covering 4609 census tracts. The pooled RRs (95% CI) of COVID-19 incidence associated with 1-year exposures to PM2.5, NO2, and O3 were 1.11 (1.04, 1.18) per 2.3 μg/m[3],1.09 (1.02, 1.17) per 3.2 ppb, and 1.06 (1.00, 1.12) per 5.5 ppb respectively. The corresponding RRs (95% CI) associated with prior 1-month exposures were 1.11 (1.03, 1.20) per 5.2 μg/m[3] for PM2.5, 1.09 (1.01, 1.17) per 6.0 ppb for NO2 and 0.96 (0.85, 1.08) per 12.0 ppb for O3.
CONCLUSION: Long-term PM2.5 and NO2 exposures were associated with increased risk of COVID-19 incidence across all case surges before February 2021. Short-term PM2.5 and NO2 exposures were also associated. Our findings suggest that air pollution may play a role in increasing the risk of COVID-19 infection.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Air Pollutants/analysis/toxicity
*Air Pollution/analysis
*COVID-19/epidemiology
Environmental Exposure/analysis
Humans
Incidence
Particulate Matter/analysis/toxicity
SARS-CoV-2
RevDate: 2023-12-13
CmpDate: 2022-04-05
Comparative Immunogenicity and Effectiveness of mRNA-1273, BNT162b2, and Ad26.COV2.S COVID-19 Vaccines.
The Journal of infectious diseases, 225(7):1141-1150.
BACKGROUND: Understanding immunogenicity and effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is critical to guide rational use.
METHODS: We compared the immunogenicity of mRNA-1273, BNT-162b2, and Ad26.COV2.S in healthy ambulatory adults. We performed an inverse-variance meta-analysis of population-level effectiveness from public health reports in > 40 million individuals.
RESULTS: A single dose of either mRNA vaccine yielded comparable antibody and neutralization titers to convalescent individuals. Ad26.COV2.S yielded lower antibody concentrations and frequently undetectable neutralization titers. Bulk and cytotoxic T-cell responses were higher in mRNA1273 and BNT162b2 than Ad26.COV2.S recipients. Regardless of vaccine, <50% of vaccinees demonstrated CD8+ T-cell responses. Antibody concentrations and neutralization titers increased comparably after the first dose of either vaccine, and further in recipients of a second dose. Prior infection was associated with high antibody concentrations and neutralization even after a single dose and regardless of vaccine. Neutralization of Beta, Gamma, and Delta strains were poorer regardless of vaccine. In meta-analysis, relative to mRNA1273 the effectiveness of BNT162b2 was lower against infection and hospitalization, and Ad26COV2.S was lower against infection, hospitalization, and death.
CONCLUSIONS: Variation in the immunogenicity correlates with variable effectiveness of the 3 vaccines deployed in the United States.
Additional Links: PMID-34888672
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34888672,
year = {2022},
author = {Naranbhai, V and Garcia-Beltran, WF and Chang, CC and Berrios Mairena, C and Thierauf, JC and Kirkpatrick, G and Onozato, ML and Cheng, J and St Denis, KJ and Lam, EC and Kaseke, C and Tano-Menka, R and Yang, D and Pavlovic, M and Yang, W and Kui, A and Miller, TE and Astudillo, MG and Cahill, JE and Dighe, AS and Gregory, DJ and Poznansky, MC and Gaiha, GD and Balazs, AB and Iafrate, AJ},
title = {Comparative Immunogenicity and Effectiveness of mRNA-1273, BNT162b2, and Ad26.COV2.S COVID-19 Vaccines.},
journal = {The Journal of infectious diseases},
volume = {225},
number = {7},
pages = {1141-1150},
pmid = {34888672},
issn = {1537-6613},
support = {DP2 DA040254/DA/NIDA NIH HHS/United States ; },
mesh = {2019-nCoV Vaccine mRNA-1273 ; *Ad26COVS1 ; Adult ; BNT162 Vaccine ; *COVID-19/prevention & control ; COVID-19 Vaccines ; Humans ; Immunogenicity, Vaccine ; SARS-CoV-2/genetics ; Vaccines, Synthetic ; mRNA Vaccines ; },
abstract = {BACKGROUND: Understanding immunogenicity and effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is critical to guide rational use.
METHODS: We compared the immunogenicity of mRNA-1273, BNT-162b2, and Ad26.COV2.S in healthy ambulatory adults. We performed an inverse-variance meta-analysis of population-level effectiveness from public health reports in > 40 million individuals.
RESULTS: A single dose of either mRNA vaccine yielded comparable antibody and neutralization titers to convalescent individuals. Ad26.COV2.S yielded lower antibody concentrations and frequently undetectable neutralization titers. Bulk and cytotoxic T-cell responses were higher in mRNA1273 and BNT162b2 than Ad26.COV2.S recipients. Regardless of vaccine, <50% of vaccinees demonstrated CD8+ T-cell responses. Antibody concentrations and neutralization titers increased comparably after the first dose of either vaccine, and further in recipients of a second dose. Prior infection was associated with high antibody concentrations and neutralization even after a single dose and regardless of vaccine. Neutralization of Beta, Gamma, and Delta strains were poorer regardless of vaccine. In meta-analysis, relative to mRNA1273 the effectiveness of BNT162b2 was lower against infection and hospitalization, and Ad26COV2.S was lower against infection, hospitalization, and death.
CONCLUSIONS: Variation in the immunogenicity correlates with variable effectiveness of the 3 vaccines deployed in the United States.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
2019-nCoV Vaccine mRNA-1273
*Ad26COVS1
Adult
BNT162 Vaccine
*COVID-19/prevention & control
COVID-19 Vaccines
Humans
Immunogenicity, Vaccine
SARS-CoV-2/genetics
Vaccines, Synthetic
mRNA Vaccines
RevDate: 2022-11-19
CmpDate: 2022-01-25
Growing Public Health Concern of COVID-19 Chronic Olfactory Dysfunction.
JAMA otolaryngology-- head & neck surgery, 148(1):81-82.
This meta-analysis examines the scale of the public health concern of COVID-19–related chronic olfactory dysfunction.
Additional Links: PMID-34792577
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34792577,
year = {2022},
author = {Khan, AM and Kallogjeri, D and Piccirillo, JF},
title = {Growing Public Health Concern of COVID-19 Chronic Olfactory Dysfunction.},
journal = {JAMA otolaryngology-- head & neck surgery},
volume = {148},
number = {1},
pages = {81-82},
pmid = {34792577},
issn = {2168-619X},
support = {TL1 TR002344/TR/NCATS NIH HHS/United States ; },
mesh = {COVID-19/*epidemiology ; Chronic Disease/*epidemiology ; Humans ; Olfaction Disorders/*epidemiology/*virology ; Public Health ; United States/epidemiology ; },
abstract = {This meta-analysis examines the scale of the public health concern of COVID-19–related chronic olfactory dysfunction.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*epidemiology
Chronic Disease/*epidemiology
Humans
Olfaction Disorders/*epidemiology/*virology
Public Health
United States/epidemiology
RevDate: 2022-05-31
CmpDate: 2022-01-31
American Society of Hematology living guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19: July 2021 update on postdischarge thromboprophylaxis.
Blood advances, 6(2):664-671.
BACKGROUND: COVID-19-related acute illness is associated with an increased risk of venous thromboembolism (VTE).
OBJECTIVE: These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in decisions about the use of anticoagulation for thromboprophylaxis in patients with COVID-19 who do not have confirmed or suspected VTE.
METHODS: ASH formed a multidisciplinary guideline panel, including 3 patient representatives, and applied strategies to minimize potential bias from conflicts of interest. The McMaster University GRADE Centre supported the guideline development process, including performing systematic evidence reviews (up to March 2021). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the grading of recommendations assessment, development, and evaluation (GRADE) approach to assess evidence and make recommendations, which were subject to public comment.
RESULTS: The panel agreed on 1 additional recommendation. The panel issued a conditional recommendation against the use of outpatient anticoagulant prophylaxis in patients with COVID-19 who are discharged from the hospital and who do not have suspected or confirmed VTE or another indication for anticoagulation.
CONCLUSIONS: This recommendation was based on very low certainty in the evidence, underscoring the need for high-quality randomized controlled trials assessing the role of postdischarge thromboprophylaxis. Other key research priorities include better evidence on assessing risk of thrombosis and bleeding outcomes in patients with COVID-19 after hospital discharge.
Additional Links: PMID-34727173
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34727173,
year = {2022},
author = {Cuker, A and Tseng, EK and Nieuwlaat, R and Angchaisuksiri, P and Blair, C and Dane, K and Davila, J and DeSancho, MT and Diuguid, D and Griffin, DO and Kahn, SR and Klok, FA and Lee, AI and Neumann, I and Pai, A and Righini, M and Sanfilippo, KM and Siegal, D and Skara, M and Terrell, DR and Touri, K and Akl, EA and Al Jabiri, RN and Al Jabiri, YN and Barbara, AM and Bognanni, A and Bou Akl, I and Boulos, M and Brignardello-Petersen, R and Charide, R and Chan, M and Colunga-Lozano, LE and Dearness, K and Darzi, AJ and Hussein, H and Karam, SG and Kolb, P and Mansour, R and Morgano, GP and Morsi, RZ and Muti-Schünemann, G and Nadim, MK and Noori, A and Philip, BA and Piggott, T and Qiu, Y and Benitez, YR and Schünemann, F and Stevens, A and Solo, K and Wiercioch, W and Mustafa, RA and Schünemann, HJ},
title = {American Society of Hematology living guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19: July 2021 update on postdischarge thromboprophylaxis.},
journal = {Blood advances},
volume = {6},
number = {2},
pages = {664-671},
pmid = {34727173},
issn = {2473-9537},
support = {K01 HL135466/HL/NHLBI NIH HHS/United States ; UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Aftercare ; Anticoagulants/adverse effects ; *COVID-19 ; Evidence-Based Medicine ; *Hematology ; Humans ; Patient Discharge ; SARS-CoV-2 ; United States ; *Venous Thromboembolism/etiology/prevention & control ; },
abstract = {BACKGROUND: COVID-19-related acute illness is associated with an increased risk of venous thromboembolism (VTE).
OBJECTIVE: These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in decisions about the use of anticoagulation for thromboprophylaxis in patients with COVID-19 who do not have confirmed or suspected VTE.
METHODS: ASH formed a multidisciplinary guideline panel, including 3 patient representatives, and applied strategies to minimize potential bias from conflicts of interest. The McMaster University GRADE Centre supported the guideline development process, including performing systematic evidence reviews (up to March 2021). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the grading of recommendations assessment, development, and evaluation (GRADE) approach to assess evidence and make recommendations, which were subject to public comment.
RESULTS: The panel agreed on 1 additional recommendation. The panel issued a conditional recommendation against the use of outpatient anticoagulant prophylaxis in patients with COVID-19 who are discharged from the hospital and who do not have suspected or confirmed VTE or another indication for anticoagulation.
CONCLUSIONS: This recommendation was based on very low certainty in the evidence, underscoring the need for high-quality randomized controlled trials assessing the role of postdischarge thromboprophylaxis. Other key research priorities include better evidence on assessing risk of thrombosis and bleeding outcomes in patients with COVID-19 after hospital discharge.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Aftercare
Anticoagulants/adverse effects
*COVID-19
Evidence-Based Medicine
*Hematology
Humans
Patient Discharge
SARS-CoV-2
United States
*Venous Thromboembolism/etiology/prevention & control
RevDate: 2023-11-08
CmpDate: 2021-11-08
Genome-scale metabolic modeling reveals SARS-CoV-2-induced metabolic changes and antiviral targets.
Molecular systems biology, 17(11):e10260.
Tremendous progress has been made to control the COVID-19 pandemic caused by the SARS-CoV-2 virus. However, effective therapeutic options are still rare. Drug repurposing and combination represent practical strategies to address this urgent unmet medical need. Viruses, including coronaviruses, are known to hijack host metabolism to facilitate viral proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM), revealing complicated host metabolism reprogramming during SARS-CoV-2 infection. We next applied the GEM-based metabolic transformation algorithm to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. We successfully validated these targets using published drug and genetic screen data and by performing an siRNA assay in Caco-2 cells. Further generating and analyzing RNA-sequencing data of remdesivir-treated Vero E6 cell samples, we predicted metabolic targets acting in combination with remdesivir, an approved anti-SARS-CoV-2 drug. Our study provides clinical data-supported candidate anti-SARS-CoV-2 targets for future evaluation, demonstrating host metabolism targeting as a promising antiviral strategy.
Additional Links: PMID-34709707
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34709707,
year = {2021},
author = {Cheng, K and Martin-Sancho, L and Pal, LR and Pu, Y and Riva, L and Yin, X and Sinha, S and Nair, NU and Chanda, SK and Ruppin, E},
title = {Genome-scale metabolic modeling reveals SARS-CoV-2-induced metabolic changes and antiviral targets.},
journal = {Molecular systems biology},
volume = {17},
number = {11},
pages = {e10260},
pmid = {34709707},
issn = {1744-4292},
support = {U19 AI118610/AI/NIAID NIH HHS/United States ; U19 AI135972/AI/NIAID NIH HHS/United States ; W81XWH-20-1-0270//U.S. Department of Defense (DOD)/ ; //HHS | National Institutes of Health (NIH)/ ; },
mesh = {Adenosine Monophosphate/*analogs & derivatives/therapeutic use ; Alanine/*analogs & derivatives/therapeutic use ; Animals ; Antiviral Agents/*therapeutic use ; COVID-19/*metabolism/virology ; Caco-2 Cells ; Chlorocebus aethiops ; Datasets as Topic ; Drug Development ; Drug Repositioning ; Host-Pathogen Interactions ; Humans ; Metabolic Networks and Pathways/*genetics ; *Pandemics ; RNA, Small Interfering ; SARS-CoV-2/*physiology ; Sequence Analysis, RNA ; Vero Cells ; COVID-19 Drug Treatment ; },
abstract = {Tremendous progress has been made to control the COVID-19 pandemic caused by the SARS-CoV-2 virus. However, effective therapeutic options are still rare. Drug repurposing and combination represent practical strategies to address this urgent unmet medical need. Viruses, including coronaviruses, are known to hijack host metabolism to facilitate viral proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM), revealing complicated host metabolism reprogramming during SARS-CoV-2 infection. We next applied the GEM-based metabolic transformation algorithm to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. We successfully validated these targets using published drug and genetic screen data and by performing an siRNA assay in Caco-2 cells. Further generating and analyzing RNA-sequencing data of remdesivir-treated Vero E6 cell samples, we predicted metabolic targets acting in combination with remdesivir, an approved anti-SARS-CoV-2 drug. Our study provides clinical data-supported candidate anti-SARS-CoV-2 targets for future evaluation, demonstrating host metabolism targeting as a promising antiviral strategy.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adenosine Monophosphate/*analogs & derivatives/therapeutic use
Alanine/*analogs & derivatives/therapeutic use
Animals
Antiviral Agents/*therapeutic use
COVID-19/*metabolism/virology
Caco-2 Cells
Chlorocebus aethiops
Datasets as Topic
Drug Development
Drug Repositioning
Host-Pathogen Interactions
Humans
Metabolic Networks and Pathways/*genetics
*Pandemics
RNA, Small Interfering
SARS-CoV-2/*physiology
Sequence Analysis, RNA
Vero Cells
COVID-19 Drug Treatment
RevDate: 2024-08-29
CmpDate: 2021-10-12
American College of Rheumatology Guidance for COVID-19 Vaccination in Patients With Rheumatic and Musculoskeletal Diseases: Version 3.
Arthritis & rheumatology (Hoboken, N.J.), 73(10):e60-e75.
OBJECTIVE: To provide guidance to rheumatology providers on the use of coronavirus disease 2019 (COVID-19) vaccines for patients with rheumatic and musculoskeletal diseases (RMDs).
METHODS: A task force was assembled that included 9 rheumatologists/immunologists, 2 infectious disease specialists, and 2 public health physicians. After agreeing on scoping questions, an evidence report was created that summarized the published literature and publicly available data regarding COVID-19 vaccine efficacy and safety, as well as literature for other vaccines in RMD patients. Task force members rated their agreement with draft consensus statements on a 9-point numerical scoring system, using a modified Delphi process and the RAND/University of California Los Angeles Appropriateness Method, with refinement and iteration over 2 sessions. Consensus was determined based on the distribution of ratings.
RESULTS: Despite a paucity of direct evidence, 74 draft guidance statements were developed by the task force and agreed upon with consensus to provide guidance for use of the COVID-19 vaccines in RMD patients and to offer recommendations regarding the use and timing of immunomodulatory therapies around the time of vaccination.
CONCLUSION: These guidance statements, made in the context of limited clinical data, are intended to provide direction to rheumatology health care providers on how to best use COVID-19 vaccines and to facilitate implementation of vaccination strategies for RMD patients.
Additional Links: PMID-34346564
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34346564,
year = {2021},
author = {Curtis, JR and Johnson, SR and Anthony, DD and Arasaratnam, RJ and Baden, LR and Bass, AR and Calabrese, C and Gravallese, EM and Harpaz, R and Kroger, A and Sadun, RE and Turner, AS and Williams, EA and Mikuls, TR},
title = {American College of Rheumatology Guidance for COVID-19 Vaccination in Patients With Rheumatic and Musculoskeletal Diseases: Version 3.},
journal = {Arthritis & rheumatology (Hoboken, N.J.)},
volume = {73},
number = {10},
pages = {e60-e75},
pmid = {34346564},
issn = {2326-5205},
support = {I01 CX001791/CX/CSRD VA/United States ; },
mesh = {COVID-19/*prevention & control ; COVID-19 Vaccines/*therapeutic use ; Humans ; *Musculoskeletal Diseases ; *Rheumatic Diseases ; *Rheumatology ; United States ; },
abstract = {OBJECTIVE: To provide guidance to rheumatology providers on the use of coronavirus disease 2019 (COVID-19) vaccines for patients with rheumatic and musculoskeletal diseases (RMDs).
METHODS: A task force was assembled that included 9 rheumatologists/immunologists, 2 infectious disease specialists, and 2 public health physicians. After agreeing on scoping questions, an evidence report was created that summarized the published literature and publicly available data regarding COVID-19 vaccine efficacy and safety, as well as literature for other vaccines in RMD patients. Task force members rated their agreement with draft consensus statements on a 9-point numerical scoring system, using a modified Delphi process and the RAND/University of California Los Angeles Appropriateness Method, with refinement and iteration over 2 sessions. Consensus was determined based on the distribution of ratings.
RESULTS: Despite a paucity of direct evidence, 74 draft guidance statements were developed by the task force and agreed upon with consensus to provide guidance for use of the COVID-19 vaccines in RMD patients and to offer recommendations regarding the use and timing of immunomodulatory therapies around the time of vaccination.
CONCLUSION: These guidance statements, made in the context of limited clinical data, are intended to provide direction to rheumatology health care providers on how to best use COVID-19 vaccines and to facilitate implementation of vaccination strategies for RMD patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*prevention & control
COVID-19 Vaccines/*therapeutic use
Humans
*Musculoskeletal Diseases
*Rheumatic Diseases
*Rheumatology
United States
RevDate: 2025-05-30
CmpDate: 2022-03-23
Mapping the human genetic architecture of COVID-19.
Nature, 600(7889):472-477.
The genetic make-up of an individual contributes to the susceptibility and response to viral infection. Although environmental, clinical and social factors have a role in the chance of exposure to SARS-CoV-2 and the severity of COVID-19[1,2], host genetics may also be important. Identifying host-specific genetic factors may reveal biological mechanisms of therapeutic relevance and clarify causal relationships of modifiable environmental risk factors for SARS-CoV-2 infection and outcomes. We formed a global network of researchers to investigate the role of human genetics in SARS-CoV-2 infection and COVID-19 severity. Here we describe the results of three genome-wide association meta-analyses that consist of up to 49,562 patients with COVID-19 from 46 studies across 19 countries. We report 13 genome-wide significant loci that are associated with SARS-CoV-2 infection or severe manifestations of COVID-19. Several of these loci correspond to previously documented associations to lung or autoimmune and inflammatory diseases[3-7]. They also represent potentially actionable mechanisms in response to infection. Mendelian randomization analyses support a causal role for smoking and body-mass index for severe COVID-19 although not for type II diabetes. The identification of novel host genetic factors associated with COVID-19 was made possible by the community of human genetics researchers coming together to prioritize the sharing of data, results, resources and analytical frameworks. This working model of international collaboration underscores what is possible for future genetic discoveries in emerging pandemics, or indeed for any complex human disease.
Additional Links: PMID-34237774
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34237774,
year = {2021},
author = {, },
title = {Mapping the human genetic architecture of COVID-19.},
journal = {Nature},
volume = {600},
number = {7889},
pages = {472-477},
pmid = {34237774},
issn = {1476-4687},
support = {MR/L501542/1/MRC_/Medical Research Council/United Kingdom ; MR/R026408/1/MRC_/Medical Research Council/United Kingdom ; G1001253/MRC_/Medical Research Council/United Kingdom ; MR/M009203/1/MRC_/Medical Research Council/United Kingdom ; MC_PC_19025/MRC_/Medical Research Council/United Kingdom ; F31 MH124306/MH/NIMH NIH HHS/United States ; MR/S020039/1/MRC_/Medical Research Council/United Kingdom ; MC_UU_12014/12/MRC_/Medical Research Council/United Kingdom ; MC_UU_12014/10/MRC_/Medical Research Council/United Kingdom ; MR/P011705/2/MRC_/Medical Research Council/United Kingdom ; MR/S006753/1/MRC_/Medical Research Council/United Kingdom ; MC_UP_A090_1006/MRC_/Medical Research Council/United Kingdom ; G0901254/MRC_/Medical Research Council/United Kingdom ; MR/P011705/1/MRC_/Medical Research Council/United Kingdom ; MC_UU_00007/10/MRC_/Medical Research Council/United Kingdom ; MC_UU_12014/8/MRC_/Medical Research Council/United Kingdom ; K12 AR084232/AR/NIAMS NIH HHS/United States ; MC_UU_00007/15/MRC_/Medical Research Council/United Kingdom ; 210561/Z/18/Z/WT_/Wellcome Trust/United Kingdom ; MR/K01417X/1/MRC_/Medical Research Council/United Kingdom ; G-0907/PUK_/Parkinson's UK/United Kingdom ; MR/S032304/1/MRC_/Medical Research Council/United Kingdom ; R01 GM133169/GM/NIGMS NIH HHS/United States ; G0701075/MRC_/Medical Research Council/United Kingdom ; MC_UU_12008/1/MRC_/Medical Research Council/United Kingdom ; R01 HL142302/HL/NHLBI NIH HHS/United States ; G-1307/PUK_/Parkinson's UK/United Kingdom ; MC_PC_20004/MRC_/Medical Research Council/United Kingdom ; MC_UU_00011/1/MRC_/Medical Research Council/United Kingdom ; T32 HG000040/HG/NHGRI NIH HHS/United States ; MC_PC_14089/MRC_/Medical Research Council/United Kingdom ; MR/N02995X/1/MRC_/Medical Research Council/United Kingdom ; MC_EX_MR/M009203/1/MRC_/Medical Research Council/United Kingdom ; K01 MH121659/MH/NIMH NIH HHS/United States ; MR/J004758/1/MRC_/Medical Research Council/United Kingdom ; MC_PC_19026/MRC_/Medical Research Council/United Kingdom ; R01 HG009120/HG/NHGRI NIH HHS/United States ; MC_PC_19059/MRC_/Medical Research Council/United Kingdom ; },
mesh = {Female ; Humans ; Male ; Autoimmunity/genetics ; Body Mass Index ; *COVID-19/genetics/virology ; Critical Illness ; *Genetic Loci/genetics ; *Genetic Predisposition to Disease ; *Genome-Wide Association Study ; Geographic Mapping ; Hospitalization ; *Host-Pathogen Interactions/genetics ; Inflammation/complications ; Information Dissemination ; Multifactorial Inheritance ; Racial Groups/genetics ; SARS-CoV-2/pathogenicity ; Smoking ; },
abstract = {The genetic make-up of an individual contributes to the susceptibility and response to viral infection. Although environmental, clinical and social factors have a role in the chance of exposure to SARS-CoV-2 and the severity of COVID-19[1,2], host genetics may also be important. Identifying host-specific genetic factors may reveal biological mechanisms of therapeutic relevance and clarify causal relationships of modifiable environmental risk factors for SARS-CoV-2 infection and outcomes. We formed a global network of researchers to investigate the role of human genetics in SARS-CoV-2 infection and COVID-19 severity. Here we describe the results of three genome-wide association meta-analyses that consist of up to 49,562 patients with COVID-19 from 46 studies across 19 countries. We report 13 genome-wide significant loci that are associated with SARS-CoV-2 infection or severe manifestations of COVID-19. Several of these loci correspond to previously documented associations to lung or autoimmune and inflammatory diseases[3-7]. They also represent potentially actionable mechanisms in response to infection. Mendelian randomization analyses support a causal role for smoking and body-mass index for severe COVID-19 although not for type II diabetes. The identification of novel host genetic factors associated with COVID-19 was made possible by the community of human genetics researchers coming together to prioritize the sharing of data, results, resources and analytical frameworks. This working model of international collaboration underscores what is possible for future genetic discoveries in emerging pandemics, or indeed for any complex human disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Female
Humans
Male
Autoimmunity/genetics
Body Mass Index
*COVID-19/genetics/virology
Critical Illness
*Genetic Loci/genetics
*Genetic Predisposition to Disease
*Genome-Wide Association Study
Geographic Mapping
Hospitalization
*Host-Pathogen Interactions/genetics
Inflammation/complications
Information Dissemination
Multifactorial Inheritance
Racial Groups/genetics
SARS-CoV-2/pathogenicity
Smoking
RevDate: 2025-05-30
CmpDate: 2021-08-16
Association Between Administration of IL-6 Antagonists and Mortality Among Patients Hospitalized for COVID-19: A Meta-analysis.
JAMA, 326(6):499-518.
IMPORTANCE: Clinical trials assessing the efficacy of IL-6 antagonists in patients hospitalized for COVID-19 have variously reported benefit, no effect, and harm.
OBJECTIVE: To estimate the association between administration of IL-6 antagonists compared with usual care or placebo and 28-day all-cause mortality and other outcomes.
DATA SOURCES: Trials were identified through systematic searches of electronic databases between October 2020 and January 2021. Searches were not restricted by trial status or language. Additional trials were identified through contact with experts.
STUDY SELECTION: Eligible trials randomly assigned patients hospitalized for COVID-19 to a group in whom IL-6 antagonists were administered and to a group in whom neither IL-6 antagonists nor any other immunomodulators except corticosteroids were administered. Among 72 potentially eligible trials, 27 (37.5%) met study selection criteria.
DATA EXTRACTION AND SYNTHESIS: In this prospective meta-analysis, risk of bias was assessed using the Cochrane Risk of Bias Assessment Tool. Inconsistency among trial results was assessed using the I2 statistic. The primary analysis was an inverse variance-weighted fixed-effects meta-analysis of odds ratios (ORs) for 28-day all-cause mortality.
MAIN OUTCOMES AND MEASURES: The primary outcome measure was all-cause mortality at 28 days after randomization. There were 9 secondary outcomes including progression to invasive mechanical ventilation or death and risk of secondary infection by 28 days.
RESULTS: A total of 10 930 patients (median age, 61 years [range of medians, 52-68 years]; 3560 [33%] were women) participating in 27 trials were included. By 28 days, there were 1407 deaths among 6449 patients randomized to IL-6 antagonists and 1158 deaths among 4481 patients randomized to usual care or placebo (summary OR, 0.86 [95% CI, 0.79-0.95]; P = .003 based on a fixed-effects meta-analysis). This corresponds to an absolute mortality risk of 22% for IL-6 antagonists compared with an assumed mortality risk of 25% for usual care or placebo. The corresponding summary ORs were 0.83 (95% CI, 0.74-0.92; P < .001) for tocilizumab and 1.08 (95% CI, 0.86-1.36; P = .52) for sarilumab. The summary ORs for the association with mortality compared with usual care or placebo in those receiving corticosteroids were 0.77 (95% CI, 0.68-0.87) for tocilizumab and 0.92 (95% CI, 0.61-1.38) for sarilumab. The ORs for the association with progression to invasive mechanical ventilation or death, compared with usual care or placebo, were 0.77 (95% CI, 0.70-0.85) for all IL-6 antagonists, 0.74 (95% CI, 0.66-0.82) for tocilizumab, and 1.00 (95% CI, 0.74-1.34) for sarilumab. Secondary infections by 28 days occurred in 21.9% of patients treated with IL-6 antagonists vs 17.6% of patients treated with usual care or placebo (OR accounting for trial sample sizes, 0.99; 95% CI, 0.85-1.16).
CONCLUSIONS AND RELEVANCE: In this prospective meta-analysis of clinical trials of patients hospitalized for COVID-19, administration of IL-6 antagonists, compared with usual care or placebo, was associated with lower 28-day all-cause mortality.
TRIAL REGISTRATION: PROSPERO Identifier: CRD42021230155.
Additional Links: PMID-34228774
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34228774,
year = {2021},
author = {, and Shankar-Hari, M and Vale, CL and Godolphin, PJ and Fisher, D and Higgins, JPT and Spiga, F and Savovic, J and Tierney, J and Baron, G and Benbenishty, JS and Berry, LR and Broman, N and Cavalcanti, AB and Colman, R and De Buyser, SL and Derde, LPG and Domingo, P and Omar, SF and Fernandez-Cruz, A and Feuth, T and Garcia, F and Garcia-Vicuna, R and Gonzalez-Alvaro, I and Gordon, AC and Haynes, R and Hermine, O and Horby, PW and Horick, NK and Kumar, K and Lambrecht, BN and Landray, MJ and Leal, L and Lederer, DJ and Lorenzi, E and Mariette, X and Merchante, N and Misnan, NA and Mohan, SV and Nivens, MC and Oksi, J and Perez-Molina, JA and Pizov, R and Porcher, R and Postma, S and Rajasuriar, R and Ramanan, AV and Ravaud, P and Reid, PD and Rutgers, A and Sancho-Lopez, A and Seto, TB and Sivapalasingam, S and Soin, AS and Staplin, N and Stone, JH and Strohbehn, GW and Sunden-Cullberg, J and Torre-Cisneros, J and Tsai, LW and van Hoogstraten, H and van Meerten, T and Veiga, VC and Westerweel, PE and Murthy, S and Diaz, JV and Marshall, JC and Sterne, JAC},
title = {Association Between Administration of IL-6 Antagonists and Mortality Among Patients Hospitalized for COVID-19: A Meta-analysis.},
journal = {JAMA},
volume = {326},
number = {6},
pages = {499-518},
pmid = {34228774},
issn = {1538-3598},
support = {16896/CRUK_/Cancer Research UK/United Kingdom ; MC_UU_00004/06/MRC_/Medical Research Council/United Kingdom ; MC_U137686861/MRC_/Medical Research Council/United Kingdom ; UM1 AI144295/AI/NIAID NIH HHS/United States ; KL2 TR002387/TR/NCATS NIH HHS/United States ; MC_U137686860/MRC_/Medical Research Council/United Kingdom ; MC_UU_12026/4/MRC_/Medical Research Council/United Kingdom ; MC_PC_20062/MRC_/Medical Research Council/United Kingdom ; 001/WHO_/World Health Organization/International ; },
mesh = {Aged ; Antibodies, Monoclonal, Humanized/*therapeutic use ; COVID-19/complications/mortality/therapy ; Cause of Death ; Coinfection ; Disease Progression ; Drug Therapy, Combination ; Female ; Glucocorticoids/therapeutic use ; Hospitalization ; Humans ; Interleukin-6/*antagonists & inhibitors ; Male ; Middle Aged ; Prospective Studies ; Randomized Controlled Trials as Topic ; Respiration, Artificial ; *COVID-19 Drug Treatment ; },
abstract = {IMPORTANCE: Clinical trials assessing the efficacy of IL-6 antagonists in patients hospitalized for COVID-19 have variously reported benefit, no effect, and harm.
OBJECTIVE: To estimate the association between administration of IL-6 antagonists compared with usual care or placebo and 28-day all-cause mortality and other outcomes.
DATA SOURCES: Trials were identified through systematic searches of electronic databases between October 2020 and January 2021. Searches were not restricted by trial status or language. Additional trials were identified through contact with experts.
STUDY SELECTION: Eligible trials randomly assigned patients hospitalized for COVID-19 to a group in whom IL-6 antagonists were administered and to a group in whom neither IL-6 antagonists nor any other immunomodulators except corticosteroids were administered. Among 72 potentially eligible trials, 27 (37.5%) met study selection criteria.
DATA EXTRACTION AND SYNTHESIS: In this prospective meta-analysis, risk of bias was assessed using the Cochrane Risk of Bias Assessment Tool. Inconsistency among trial results was assessed using the I2 statistic. The primary analysis was an inverse variance-weighted fixed-effects meta-analysis of odds ratios (ORs) for 28-day all-cause mortality.
MAIN OUTCOMES AND MEASURES: The primary outcome measure was all-cause mortality at 28 days after randomization. There were 9 secondary outcomes including progression to invasive mechanical ventilation or death and risk of secondary infection by 28 days.
RESULTS: A total of 10 930 patients (median age, 61 years [range of medians, 52-68 years]; 3560 [33%] were women) participating in 27 trials were included. By 28 days, there were 1407 deaths among 6449 patients randomized to IL-6 antagonists and 1158 deaths among 4481 patients randomized to usual care or placebo (summary OR, 0.86 [95% CI, 0.79-0.95]; P = .003 based on a fixed-effects meta-analysis). This corresponds to an absolute mortality risk of 22% for IL-6 antagonists compared with an assumed mortality risk of 25% for usual care or placebo. The corresponding summary ORs were 0.83 (95% CI, 0.74-0.92; P < .001) for tocilizumab and 1.08 (95% CI, 0.86-1.36; P = .52) for sarilumab. The summary ORs for the association with mortality compared with usual care or placebo in those receiving corticosteroids were 0.77 (95% CI, 0.68-0.87) for tocilizumab and 0.92 (95% CI, 0.61-1.38) for sarilumab. The ORs for the association with progression to invasive mechanical ventilation or death, compared with usual care or placebo, were 0.77 (95% CI, 0.70-0.85) for all IL-6 antagonists, 0.74 (95% CI, 0.66-0.82) for tocilizumab, and 1.00 (95% CI, 0.74-1.34) for sarilumab. Secondary infections by 28 days occurred in 21.9% of patients treated with IL-6 antagonists vs 17.6% of patients treated with usual care or placebo (OR accounting for trial sample sizes, 0.99; 95% CI, 0.85-1.16).
CONCLUSIONS AND RELEVANCE: In this prospective meta-analysis of clinical trials of patients hospitalized for COVID-19, administration of IL-6 antagonists, compared with usual care or placebo, was associated with lower 28-day all-cause mortality.
TRIAL REGISTRATION: PROSPERO Identifier: CRD42021230155.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Aged
Antibodies, Monoclonal, Humanized/*therapeutic use
COVID-19/complications/mortality/therapy
Cause of Death
Coinfection
Disease Progression
Drug Therapy, Combination
Female
Glucocorticoids/therapeutic use
Hospitalization
Humans
Interleukin-6/*antagonists & inhibitors
Male
Middle Aged
Prospective Studies
Randomized Controlled Trials as Topic
Respiration, Artificial
*COVID-19 Drug Treatment
RevDate: 2022-12-07
CmpDate: 2021-10-20
Prospective individual patient data meta-analysis: Evaluating convalescent plasma for COVID-19.
Statistics in medicine, 40(24):5131-5151.
As the world faced the devastation of the COVID-19 pandemic in late 2019 and early 2020, numerous clinical trials were initiated in many locations in an effort to establish the efficacy (or lack thereof) of potential treatments. As the pandemic has been shifting locations rapidly, individual studies have been at risk of failing to meet recruitment targets because of declining numbers of eligible patients with COVID-19 encountered at participating sites. It has become clear that it might take several more COVID-19 surges at the same location to achieve full enrollment and to find answers about what treatments are effective for this disease. This paper proposes an innovative approach for pooling patient-level data from multiple ongoing randomized clinical trials (RCTs) that have not been configured as a network of sites. We present the statistical analysis plan of a prospective individual patient data (IPD) meta-analysis (MA) from ongoing RCTs of convalescent plasma (CP). We employ an adaptive Bayesian approach for continuously monitoring the accumulating pooled data via posterior probabilities for safety, efficacy, and harm. Although we focus on RCTs for CP and address specific challenges related to CP treatment for COVID-19, the proposed framework is generally applicable to pooling data from RCTs for other therapies and disease settings in order to find answers in weeks or months, rather than years.
Additional Links: PMID-34164838
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34164838,
year = {2021},
author = {Goldfeld, KS and Wu, D and Tarpey, T and Liu, M and Wu, Y and Troxel, AB and Petkova, E},
title = {Prospective individual patient data meta-analysis: Evaluating convalescent plasma for COVID-19.},
journal = {Statistics in medicine},
volume = {40},
number = {24},
pages = {5131-5151},
pmid = {34164838},
issn = {1097-0258},
support = {UL1 TR001445/TR/NCATS NIH HHS/United States ; 3UL1TR001445-05S3/TR/NCATS NIH HHS/United States ; 3UL1TR001445-05S3/TR/NCATS NIH HHS/United States ; 3UL1 TR001445-05S3/NH/NIH HHS/United States ; },
mesh = {*COVID-19/therapy ; *Coronavirus Infections ; Humans ; Immunization, Passive ; Pandemics ; SARS-CoV-2 ; COVID-19 Serotherapy ; },
abstract = {As the world faced the devastation of the COVID-19 pandemic in late 2019 and early 2020, numerous clinical trials were initiated in many locations in an effort to establish the efficacy (or lack thereof) of potential treatments. As the pandemic has been shifting locations rapidly, individual studies have been at risk of failing to meet recruitment targets because of declining numbers of eligible patients with COVID-19 encountered at participating sites. It has become clear that it might take several more COVID-19 surges at the same location to achieve full enrollment and to find answers about what treatments are effective for this disease. This paper proposes an innovative approach for pooling patient-level data from multiple ongoing randomized clinical trials (RCTs) that have not been configured as a network of sites. We present the statistical analysis plan of a prospective individual patient data (IPD) meta-analysis (MA) from ongoing RCTs of convalescent plasma (CP). We employ an adaptive Bayesian approach for continuously monitoring the accumulating pooled data via posterior probabilities for safety, efficacy, and harm. Although we focus on RCTs for CP and address specific challenges related to CP treatment for COVID-19, the proposed framework is generally applicable to pooling data from RCTs for other therapies and disease settings in order to find answers in weeks or months, rather than years.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/therapy
*Coronavirus Infections
Humans
Immunization, Passive
Pandemics
SARS-CoV-2
COVID-19 Serotherapy
RevDate: 2022-12-07
CmpDate: 2021-07-08
Integrative omics provide biological and clinical insights into acute respiratory distress syndrome.
Intensive care medicine, 47(7):761-771.
PURPOSE: Acute respiratory distress syndrome (ARDS) is accompanied by a dysfunctional immune-inflammatory response following lung injury, including during coronavirus disease 2019 (COVID-19). Limited causal biomarkers exist for ARDS development. We sought to identify novel genetic susceptibility targets for ARDS to focus further investigation on their biological mechanism and therapeutic potential.
METHODS: Meta-analyses of ARDS genome-wide association studies were performed with 1250 cases and 1583 controls in Europeans, and 387 cases and 387 controls in African Americans. The functionality of novel loci was determined in silico using multiple omics approaches. The causality of 114 factors potentially involved in ARDS development was assessed using Mendelian Randomization analysis.
RESULTS: There was distinct genetic heterogeneity in ARDS between Europeans and African Americans. rs7967111 at 12p13.2 was functionally associated with ARDS susceptibility in Europeans (odds ratio = 1.38; P = 2.15 × 10[-8]). Expression of two genes annotated at this locus, BORCS5 and DUSP16, was dynamic but ultimately decreased during ARDS development, as well as downregulated in immune cells alongside COVID-19 severity. Causal inference implied that comorbidity of inflammatory bowel disease and elevated levels of C-reactive protein and interleukin-10 causally increased ARDS risk, while vitamin D supplementation and vasodilator use ameliorated risk.
CONCLUSION: Our findings suggest a novel susceptibility locus in ARDS pathophysiology that implicates BORCS5 and DUSP16 as potentially acting in immune-inflammatory processes. This locus warrants further investigation to inform the development of therapeutic targets and clinical care strategies for ARDS, including those induced by COVID-19.
Additional Links: PMID-34032881
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34032881,
year = {2021},
author = {Du, M and Garcia, JGN and Christie, JD and Xin, J and Cai, G and Meyer, NJ and Zhu, Z and Yuan, Q and Zhang, Z and Su, L and Shen, S and Dong, X and Li, H and Hutchinson, JN and Tejera, P and Lin, X and Wang, M and Chen, F and Christiani, DC},
title = {Integrative omics provide biological and clinical insights into acute respiratory distress syndrome.},
journal = {Intensive care medicine},
volume = {47},
number = {7},
pages = {761-771},
pmid = {34032881},
issn = {1432-1238},
support = {P30 ES006694/ES/NIEHS NIH HHS/United States ; P30 ES000002/ES/NIEHS NIH HHS/United States ; ES000002//Foundation for the National Institutes of Health/ ; P30 ES013508/ES/NIEHS NIH HHS/United States ; 82041024//Special Program of the National Natural Science Foundation of China (NSFC) on tracing, pathogenesis, prevention, and treatment for COVID-19/ ; K01 AI153558/AI/NIAID NIH HHS/United States ; R01HL060710//Foundation for the National Institutes of Health/ ; R56HL134356//Foundation for the National Institutes of Health/ ; P01 HL126609/HL/NHLBI NIH HHS/United States ; },
mesh = {*COVID-19 ; Genome-Wide Association Study ; Humans ; *Respiratory Distress Syndrome/genetics ; SARS-CoV-2 ; White People/genetics ; },
abstract = {PURPOSE: Acute respiratory distress syndrome (ARDS) is accompanied by a dysfunctional immune-inflammatory response following lung injury, including during coronavirus disease 2019 (COVID-19). Limited causal biomarkers exist for ARDS development. We sought to identify novel genetic susceptibility targets for ARDS to focus further investigation on their biological mechanism and therapeutic potential.
METHODS: Meta-analyses of ARDS genome-wide association studies were performed with 1250 cases and 1583 controls in Europeans, and 387 cases and 387 controls in African Americans. The functionality of novel loci was determined in silico using multiple omics approaches. The causality of 114 factors potentially involved in ARDS development was assessed using Mendelian Randomization analysis.
RESULTS: There was distinct genetic heterogeneity in ARDS between Europeans and African Americans. rs7967111 at 12p13.2 was functionally associated with ARDS susceptibility in Europeans (odds ratio = 1.38; P = 2.15 × 10[-8]). Expression of two genes annotated at this locus, BORCS5 and DUSP16, was dynamic but ultimately decreased during ARDS development, as well as downregulated in immune cells alongside COVID-19 severity. Causal inference implied that comorbidity of inflammatory bowel disease and elevated levels of C-reactive protein and interleukin-10 causally increased ARDS risk, while vitamin D supplementation and vasodilator use ameliorated risk.
CONCLUSION: Our findings suggest a novel susceptibility locus in ARDS pathophysiology that implicates BORCS5 and DUSP16 as potentially acting in immune-inflammatory processes. This locus warrants further investigation to inform the development of therapeutic targets and clinical care strategies for ARDS, including those induced by COVID-19.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19
Genome-Wide Association Study
Humans
*Respiratory Distress Syndrome/genetics
SARS-CoV-2
White People/genetics
RevDate: 2022-02-21
CmpDate: 2022-02-21
Association Between Olfactory Dysfunction and Critical Illness and Mortality in COVID-19: A Meta-analysis.
Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery, 166(2):388-392.
4.
Additional Links: PMID-34030510
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34030510,
year = {2022},
author = {Goshtasbi, K and Pang, J and Lehrich, BM and Vasudev, M and Birkenbeuel, JL and Abiri, A and Kuan, EC},
title = {Association Between Olfactory Dysfunction and Critical Illness and Mortality in COVID-19: A Meta-analysis.},
journal = {Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery},
volume = {166},
number = {2},
pages = {388-392},
doi = {10.1177/01945998211017442},
pmid = {34030510},
issn = {1097-6817},
support = {T32 GM008208/GM/NIGMS NIH HHS/United States ; T32 GM008620/GM/NIGMS NIH HHS/United States ; },
mesh = {Anosmia/complications ; COVID-19/*complications/immunology ; Hospitalization/statistics & numerical data ; Humans ; Olfaction Disorders/*complications ; *Patient Acuity ; SARS-CoV-2/immunology ; },
abstract = {4.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Anosmia/complications
COVID-19/*complications/immunology
Hospitalization/statistics & numerical data
Humans
Olfaction Disorders/*complications
*Patient Acuity
SARS-CoV-2/immunology
RevDate: 2024-08-13
CmpDate: 2021-09-07
AGA Rapid Review and Guideline for SARS-CoV2 Testing and Endoscopy Post-Vaccination: 2021 Update.
Gastroenterology, 161(3):1011-1029.e11.
This guideline provides updated recommendations on the role of preprocedure testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in individuals undergoing endoscopy in the post-vaccination period and replaces the prior guideline from the American Gastroenterological Association (AGA) (released July 29, 2020). Since the start of the pandemic, our increased understanding of transmission has facilitated the implementation of practices to promote patient and health care worker (HCW) safety. Simultaneously, there has been increasing recognition of the potential harm associated with delays in patient care, as well as inefficiency of endoscopy units. With widespread vaccination of HCWs and the general population, a re-evaluation of AGA's prior recommendations was warranted. In order to update the role of preprocedure testing for SARS-CoV2, the AGA guideline panel reviewed the evidence on prevalence of asymptomatic SARS-CoV2 infections in individuals undergoing endoscopy; patient and HCW risk of infections that may be acquired immediately before, during, or after endoscopy; effectiveness of COVID-19 vaccine in reducing risk of infections and transmission; patient and HCW anxiety; patient delays in care and potential impact on cancer burden; and endoscopy volumes. The panel considered the certainty of the evidence, weighed the benefits and harms of routine preprocedure testing, and considered burden, equity, and cost using the Grading of Recommendations Assessment, Development and Evaluation framework. Based on very low certainty evidence, the panel made a conditional recommendation against routine preprocedure testing for SARS-CoV2 in patients scheduled to undergo endoscopy. The panel placed a high value on minimizing additional delays in patient care, acknowledging the reduced endoscopy volumes, downstream impact on delayed cancer diagnoses, and burden of testing on patients.
Additional Links: PMID-34029569
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid34029569,
year = {2021},
author = {Sultan, S and Siddique, SM and Singh, S and Altayar, O and Caliendo, AM and Davitkov, P and Feuerstein, JD and Kaul, V and Lim, JK and Mustafa, RA and Falck-Ytter, Y and Inadomi, JM and , },
title = {AGA Rapid Review and Guideline for SARS-CoV2 Testing and Endoscopy Post-Vaccination: 2021 Update.},
journal = {Gastroenterology},
volume = {161},
number = {3},
pages = {1011-1029.e11},
pmid = {34029569},
issn = {1528-0012},
support = {K23 DK117058/DK/NIDDK NIH HHS/United States ; UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {*COVID-19/diagnosis/therapy ; COVID-19 Vaccines/therapeutic use ; *Endoscopy/standards ; Gastroenterology/standards ; Humans ; Mass Screening/*standards ; *Pandemics ; SARS-CoV-2 ; Vaccination ; },
abstract = {This guideline provides updated recommendations on the role of preprocedure testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in individuals undergoing endoscopy in the post-vaccination period and replaces the prior guideline from the American Gastroenterological Association (AGA) (released July 29, 2020). Since the start of the pandemic, our increased understanding of transmission has facilitated the implementation of practices to promote patient and health care worker (HCW) safety. Simultaneously, there has been increasing recognition of the potential harm associated with delays in patient care, as well as inefficiency of endoscopy units. With widespread vaccination of HCWs and the general population, a re-evaluation of AGA's prior recommendations was warranted. In order to update the role of preprocedure testing for SARS-CoV2, the AGA guideline panel reviewed the evidence on prevalence of asymptomatic SARS-CoV2 infections in individuals undergoing endoscopy; patient and HCW risk of infections that may be acquired immediately before, during, or after endoscopy; effectiveness of COVID-19 vaccine in reducing risk of infections and transmission; patient and HCW anxiety; patient delays in care and potential impact on cancer burden; and endoscopy volumes. The panel considered the certainty of the evidence, weighed the benefits and harms of routine preprocedure testing, and considered burden, equity, and cost using the Grading of Recommendations Assessment, Development and Evaluation framework. Based on very low certainty evidence, the panel made a conditional recommendation against routine preprocedure testing for SARS-CoV2 in patients scheduled to undergo endoscopy. The panel placed a high value on minimizing additional delays in patient care, acknowledging the reduced endoscopy volumes, downstream impact on delayed cancer diagnoses, and burden of testing on patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19/diagnosis/therapy
COVID-19 Vaccines/therapeutic use
*Endoscopy/standards
Gastroenterology/standards
Humans
Mass Screening/*standards
*Pandemics
SARS-CoV-2
Vaccination
RevDate: 2024-08-31
CmpDate: 2021-09-24
Methodological assessment of systematic reviews and meta-analyses on COVID-19: A meta-epidemiological study.
Journal of evaluation in clinical practice, 27(5):1123-1133.
COVID-19 has caused an ongoing public health crisis. Many systematic reviews and meta-analyses have been performed to synthesize evidence for better understanding this new disease. However, some concerns have been raised about rapid COVID-19 research. This meta-epidemiological study aims to methodologically assess the current systematic reviews and meta-analyses on COVID-19.
METHODS: We searched in various databases for systematic reviews with meta-analyses published between 1 January 2020 and 31 October 2020. We extracted their basic characteristics, data analyses, evidence appraisal, and assessment of publication bias and heterogeneity.
RESULTS: We identified 295 systematic reviews on COVID-19. The median time from submission to acceptance was 33 days. Among these systematic reviews, 73.9% evaluated clinical manifestations or comorbidities of COVID-19. Stata was the most used software programme (43.39%). The odds ratio was the most used effect measure (34.24%). Moreover, 28.14% of the systematic reviews did not present evidence appraisal. Among those reporting the risk of bias results, 14.64% of studies had a high risk of bias. Egger's test was the most used method for assessing publication bias (38.31%), while 38.66% of the systematic reviews did not assess publication bias. The I[2] statistic was widely used for assessing heterogeneity (92.20%); many meta-analyses had high values of I[2] . Among the meta-analyses using the random-effects model, 75.82% did not report the methods for model implementation; among those meta-analyses reporting implementation methods, the DerSimonian-Laird method was the most used one.
CONCLUSIONS: The current systematic reviews and meta-analyses on COVID-19 might suffer from low transparency, high heterogeneity, and suboptimal statistical methods. It is recommended that future systematic reviews on COVID-19 strictly follow well-developed guidelines. Sensitivity analyses may be performed to examine how the synthesized evidence might depend on different methods for appraising evidence, assessing publication bias, and implementing meta-analysis models.
Additional Links: PMID-33955120
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33955120,
year = {2021},
author = {Rosenberger, KJ and Xu, C and Lin, L},
title = {Methodological assessment of systematic reviews and meta-analyses on COVID-19: A meta-epidemiological study.},
journal = {Journal of evaluation in clinical practice},
volume = {27},
number = {5},
pages = {1123-1133},
pmid = {33955120},
issn = {1365-2753},
support = {R01 LM012982/LM/NLM NIH HHS/United States ; UL1 TR001427/TR/NCATS NIH HHS/United States ; },
mesh = {*COVID-19 ; Epidemiologic Studies ; Humans ; Publication Bias ; SARS-CoV-2 ; Systematic Reviews as Topic ; },
abstract = {COVID-19 has caused an ongoing public health crisis. Many systematic reviews and meta-analyses have been performed to synthesize evidence for better understanding this new disease. However, some concerns have been raised about rapid COVID-19 research. This meta-epidemiological study aims to methodologically assess the current systematic reviews and meta-analyses on COVID-19.
METHODS: We searched in various databases for systematic reviews with meta-analyses published between 1 January 2020 and 31 October 2020. We extracted their basic characteristics, data analyses, evidence appraisal, and assessment of publication bias and heterogeneity.
RESULTS: We identified 295 systematic reviews on COVID-19. The median time from submission to acceptance was 33 days. Among these systematic reviews, 73.9% evaluated clinical manifestations or comorbidities of COVID-19. Stata was the most used software programme (43.39%). The odds ratio was the most used effect measure (34.24%). Moreover, 28.14% of the systematic reviews did not present evidence appraisal. Among those reporting the risk of bias results, 14.64% of studies had a high risk of bias. Egger's test was the most used method for assessing publication bias (38.31%), while 38.66% of the systematic reviews did not assess publication bias. The I[2] statistic was widely used for assessing heterogeneity (92.20%); many meta-analyses had high values of I[2] . Among the meta-analyses using the random-effects model, 75.82% did not report the methods for model implementation; among those meta-analyses reporting implementation methods, the DerSimonian-Laird method was the most used one.
CONCLUSIONS: The current systematic reviews and meta-analyses on COVID-19 might suffer from low transparency, high heterogeneity, and suboptimal statistical methods. It is recommended that future systematic reviews on COVID-19 strictly follow well-developed guidelines. Sensitivity analyses may be performed to examine how the synthesized evidence might depend on different methods for appraising evidence, assessing publication bias, and implementing meta-analysis models.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19
Epidemiologic Studies
Humans
Publication Bias
SARS-CoV-2
Systematic Reviews as Topic
RevDate: 2022-12-07
CmpDate: 2021-04-30
From nicotine to the cholinergic anti-inflammatory reflex - Can nicotine alleviate the dysregulated inflammation in COVID-19?.
Journal of immunotoxicology, 18(1):23-29.
The coronavirus SARS-CoV-2 of 2019 (COVID-19) causes a pandemic that has been diagnosed in more than 70 million people worldwide. Mild-to-moderate COVID-19 symptoms include coughing, fever, myalgia, shortness of breath, and acute inflammatory lung injury (ALI). In contrast, acute respiratory distress syndrome (ARDS) and respiratory failure occur in patients diagnosed with severe COVID-19. ARDS is mediated, at least in part, by a dysregulated inflammatory response due to excessive levels of circulating cytokines, a condition known as the "cytokine-storm syndrome." Currently, there are FDA-approved therapies that attenuate the dysregulated inflammation that occurs in COVID-19 patients, such as dexamethasone or other corticosteroids and IL-6 inhibitors, including sarilumab, tocilizumab, and siltuximab. However, the efficacy of these treatments have been shown to be inconsistent. Compounds that activate the vagus nerve-mediated cholinergic anti-inflammatory reflex, such as the α7 nicotinic acetylcholine receptor agonist, GTS-21, attenuate ARDS/inflammatory lung injury by decreasing the extracellular levels of high mobility group box-1 (HMGB1) in the airways and the circulation. It is possible that HMGB1 may be an important mediator of the "cytokine-storm syndrome." Notably, high plasma levels of HMGB1 have been reported in patients diagnosed with severe COVID-19, and there is a significant negative correlation between HMGB1 plasma levels and clinical outcomes. Nicotine can activate the cholinergic anti-inflammatory reflex, which attenuates the up-regulation and the excessive release of pro-inflammatory cytokines/chemokines. Therefore, we hypothesize that low molecular weight compounds that activate the cholinergic anti-inflammatory reflex, such as nicotine or GTS-21, may represent a potential therapeutic approach to attenuate the dysregulated inflammatory responses in patients with severe COVID-19.
Additional Links: PMID-33860730
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33860730,
year = {2021},
author = {Gauthier, AG and Lin, M and Wu, J and Kennedy, TP and Daley, LA and Ashby, CR and Mantell, LL},
title = {From nicotine to the cholinergic anti-inflammatory reflex - Can nicotine alleviate the dysregulated inflammation in COVID-19?.},
journal = {Journal of immunotoxicology},
volume = {18},
number = {1},
pages = {23-29},
doi = {10.1080/1547691X.2021.1875085},
pmid = {33860730},
issn = {1547-6901},
support = {R15 HL093708/HL/NHLBI NIH HHS/United States ; },
mesh = {Antibodies, Monoclonal, Humanized/therapeutic use ; Benzylidene Compounds/*pharmacology ; Cholinergic Agents/*pharmacology ; Cigarette Smoking/adverse effects ; Dexamethasone/therapeutic use ; HMGB1 Protein/blood ; Humans ; Inflammation/*drug therapy ; Nicotine/*metabolism ; Pandemics ; Pyridines/*pharmacology ; SARS-CoV-2/*physiology ; Tobacco Use Disorder/*drug therapy ; alpha7 Nicotinic Acetylcholine Receptor/agonists ; *COVID-19 Drug Treatment ; },
abstract = {The coronavirus SARS-CoV-2 of 2019 (COVID-19) causes a pandemic that has been diagnosed in more than 70 million people worldwide. Mild-to-moderate COVID-19 symptoms include coughing, fever, myalgia, shortness of breath, and acute inflammatory lung injury (ALI). In contrast, acute respiratory distress syndrome (ARDS) and respiratory failure occur in patients diagnosed with severe COVID-19. ARDS is mediated, at least in part, by a dysregulated inflammatory response due to excessive levels of circulating cytokines, a condition known as the "cytokine-storm syndrome." Currently, there are FDA-approved therapies that attenuate the dysregulated inflammation that occurs in COVID-19 patients, such as dexamethasone or other corticosteroids and IL-6 inhibitors, including sarilumab, tocilizumab, and siltuximab. However, the efficacy of these treatments have been shown to be inconsistent. Compounds that activate the vagus nerve-mediated cholinergic anti-inflammatory reflex, such as the α7 nicotinic acetylcholine receptor agonist, GTS-21, attenuate ARDS/inflammatory lung injury by decreasing the extracellular levels of high mobility group box-1 (HMGB1) in the airways and the circulation. It is possible that HMGB1 may be an important mediator of the "cytokine-storm syndrome." Notably, high plasma levels of HMGB1 have been reported in patients diagnosed with severe COVID-19, and there is a significant negative correlation between HMGB1 plasma levels and clinical outcomes. Nicotine can activate the cholinergic anti-inflammatory reflex, which attenuates the up-regulation and the excessive release of pro-inflammatory cytokines/chemokines. Therefore, we hypothesize that low molecular weight compounds that activate the cholinergic anti-inflammatory reflex, such as nicotine or GTS-21, may represent a potential therapeutic approach to attenuate the dysregulated inflammatory responses in patients with severe COVID-19.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Antibodies, Monoclonal, Humanized/therapeutic use
Benzylidene Compounds/*pharmacology
Cholinergic Agents/*pharmacology
Cigarette Smoking/adverse effects
Dexamethasone/therapeutic use
HMGB1 Protein/blood
Humans
Inflammation/*drug therapy
Nicotine/*metabolism
Pandemics
Pyridines/*pharmacology
SARS-CoV-2/*physiology
Tobacco Use Disorder/*drug therapy
alpha7 Nicotinic Acetylcholine Receptor/agonists
*COVID-19 Drug Treatment
RevDate: 2024-08-08
CmpDate: 2021-04-16
Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.
Radiology. Imaging cancer, 2(3):e204013.
BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
MATERIALS AND METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small cell lung cancer.
CONCLUSION: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.© 2020 RSNA; The American College of Chest Physicians, published by Elsevier Inc; and The American College of Radiology, published by Elsevier Inc.
Additional Links: PMID-33778716
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33778716,
year = {2020},
author = {Mazzone, PJ and Gould, MK and Arenberg, DA and Chen, AC and Choi, HK and Detterbeck, FC and Farjah, F and Fong, KM and Iaccarino, JM and Janes, SM and Kanne, JP and Kazerooni, EA and MacMahon, H and Naidich, DP and Powell, CA and Raoof, S and Rivera, MP and Tanner, NT and Tanoue, LK and Tremblay, A and Vachani, A and White, CS and Wiener, RS and Silvestri, GA},
title = {Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.},
journal = {Radiology. Imaging cancer},
volume = {2},
number = {3},
pages = {e204013},
pmid = {33778716},
issn = {2638-616X},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {COVID-19/*prevention & control ; Diagnostic Imaging/*methods ; Humans ; Lung/diagnostic imaging ; Lung Neoplasms/*diagnostic imaging ; Pandemics ; SARS-CoV-2 ; },
abstract = {BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
MATERIALS AND METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small cell lung cancer.
CONCLUSION: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.© 2020 RSNA; The American College of Chest Physicians, published by Elsevier Inc; and The American College of Radiology, published by Elsevier Inc.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*prevention & control
Diagnostic Imaging/*methods
Humans
Lung/diagnostic imaging
Lung Neoplasms/*diagnostic imaging
Pandemics
SARS-CoV-2
RevDate: 2023-08-27
CmpDate: 2021-03-17
Roadmap for Conducting Neuroscience Research in the COVID-19 Era and Beyond: Recommendations From the SNACC Research Committee.
Journal of neurosurgical anesthesiology, 33(2):100-106.
The coronavirus disease 2019 (COVID-19) pandemic has impacted many aspects of neuroscience research. At the 2020 Society of Neuroscience in Anesthesiology and Critical Care (SNACC) Annual Meeting, the SNACC Research Committee met virtually to discuss research challenges encountered during the COVID-19 pandemic along with possible strategies for facilitating research activities. These challenges and recommendations are included in this Consensus Statement. The objectives are to: (1) provide an overview of the disruptions and challenges to neuroscience research caused by the COVID-19 pandemic, and; (2) put forth a set of consensus recommendations for strengthening research sustainability during and beyond the current pandemic. Specific recommendations are highlighted for adapting laboratory and human subject study activities to optimize safety. Complementary research activities are also outlined for both laboratory and clinical researchers if specific investigations are impossible because of regulatory or societal changes. The role of virtual platforms is discussed with respect to fostering new collaborations, scheduling research meetings, and holding conferences such that scientific collaboration and exchange of ideas can continue. Our hope is for these recommendations to serve as a valuable resource for investigators in the neurosciences and other research disciplines for current and future research disruptions.
Additional Links: PMID-33660699
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33660699,
year = {2021},
author = {Vlisides, PE and Vogt, KM and Pal, D and Schnell, E and Armstead, WM and Brambrink, AM and Kuo, P and Nelson, P and Vacas, S and Goettel, N and Aglio, LS and Farag, E and Gorji, R and GarcÃa, PS and Koerner, IP},
title = {Roadmap for Conducting Neuroscience Research in the COVID-19 Era and Beyond: Recommendations From the SNACC Research Committee.},
journal = {Journal of neurosurgical anesthesiology},
volume = {33},
number = {2},
pages = {100-106},
pmid = {33660699},
issn = {1537-1921},
support = {K23 GM132755/GM/NIGMS NIH HHS/United States ; K23 GM132795/GM/NIGMS NIH HHS/United States ; L30 GM120759/GM/NIGMS NIH HHS/United States ; R21 AG070269/AG/NIA NIH HHS/United States ; },
mesh = {COVID-19/*prevention & control ; Consensus ; Humans ; Neurosciences/*methods ; Pandemics ; *Research ; SARS-CoV-2 ; Societies, Medical ; },
abstract = {The coronavirus disease 2019 (COVID-19) pandemic has impacted many aspects of neuroscience research. At the 2020 Society of Neuroscience in Anesthesiology and Critical Care (SNACC) Annual Meeting, the SNACC Research Committee met virtually to discuss research challenges encountered during the COVID-19 pandemic along with possible strategies for facilitating research activities. These challenges and recommendations are included in this Consensus Statement. The objectives are to: (1) provide an overview of the disruptions and challenges to neuroscience research caused by the COVID-19 pandemic, and; (2) put forth a set of consensus recommendations for strengthening research sustainability during and beyond the current pandemic. Specific recommendations are highlighted for adapting laboratory and human subject study activities to optimize safety. Complementary research activities are also outlined for both laboratory and clinical researchers if specific investigations are impossible because of regulatory or societal changes. The role of virtual platforms is discussed with respect to fostering new collaborations, scheduling research meetings, and holding conferences such that scientific collaboration and exchange of ideas can continue. Our hope is for these recommendations to serve as a valuable resource for investigators in the neurosciences and other research disciplines for current and future research disruptions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*prevention & control
Consensus
Humans
Neurosciences/*methods
Pandemics
*Research
SARS-CoV-2
Societies, Medical
RevDate: 2025-05-30
CmpDate: 2021-03-26
Single-cell meta-analysis of SARS-CoV-2 entry genes across tissues and demographics.
Nature medicine, 27(3):546-559.
Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2[+]TMPRSS2[+] cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.
Additional Links: PMID-33654293
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33654293,
year = {2021},
author = {Muus, C and Luecken, MD and Eraslan, G and Sikkema, L and Waghray, A and Heimberg, G and Kobayashi, Y and Vaishnav, ED and Subramanian, A and Smillie, C and Jagadeesh, KA and Duong, ET and Fiskin, E and Torlai Triglia, E and Ansari, M and Cai, P and Lin, B and Buchanan, J and Chen, S and Shu, J and Haber, AL and Chung, H and Montoro, DT and Adams, T and Aliee, H and Allon, SJ and Andrusivova, Z and Angelidis, I and Ashenberg, O and Bassler, K and Bécavin, C and Benhar, I and Bergenstråhle, J and Bergenstråhle, L and Bolt, L and Braun, E and Bui, LT and Callori, S and Chaffin, M and Chichelnitskiy, E and Chiou, J and Conlon, TM and Cuoco, MS and Cuomo, ASE and Deprez, M and Duclos, G and Fine, D and Fischer, DS and Ghazanfar, S and Gillich, A and Giotti, B and Gould, J and Guo, M and Gutierrez, AJ and Habermann, AC and Harvey, T and He, P and Hou, X and Hu, L and Hu, Y and Jaiswal, A and Ji, L and Jiang, P and Kapellos, TS and Kuo, CS and Larsson, L and Leney-Greene, MA and Lim, K and Litviňuková, M and Ludwig, LS and Lukassen, S and Luo, W and Maatz, H and Madissoon, E and Mamanova, L and Manakongtreecheep, K and Leroy, S and Mayr, CH and Mbano, IM and McAdams, AM and Nabhan, AN and Nyquist, SK and Penland, L and Poirion, OB and Poli, S and Qi, C and Queen, R and Reichart, D and Rosas, I and Schupp, JC and Shea, CV and Shi, X and Sinha, R and Sit, RV and Slowikowski, K and Slyper, M and Smith, NP and Sountoulidis, A and Strunz, M and Sullivan, TB and Sun, D and Talavera-López, C and Tan, P and Tantivit, J and Travaglini, KJ and Tucker, NR and Vernon, KA and Wadsworth, MH and Waldman, J and Wang, X and Xu, K and Yan, W and Zhao, W and Ziegler, CGK and , and , },
title = {Single-cell meta-analysis of SARS-CoV-2 entry genes across tissues and demographics.},
journal = {Nature medicine},
volume = {27},
number = {3},
pages = {546-559},
pmid = {33654293},
issn = {1546-170X},
support = {K08 HL130595/HL/NHLBI NIH HHS/United States ; MR/S005579/1/MRC_/Medical Research Council/United Kingdom ; U01 HL148867/HL/NHLBI NIH HHS/United States ; MR/S035826/1/MRC_/Medical Research Council/United Kingdom ; MR/S035907/1/MRC_/Medical Research Council/United Kingdom ; R01 HL146519/HL/NHLBI NIH HHS/United States ; R01 HL146557/HL/NHLBI NIH HHS/United States ; R01 HL130938/HL/NHLBI NIH HHS/United States ; U01 HL145567/HL/NHLBI NIH HHS/United States ; U01 HL145550/HL/NHLBI NIH HHS/United States ; U01 HL148861/HL/NHLBI NIH HHS/United States ; F32 HL149290/HL/NHLBI NIH HHS/United States ; MR/P009581/1/MRC_/Medical Research Council/United Kingdom ; R01 HL141380/HL/NHLBI NIH HHS/United States ; R01 HL119215/HL/NHLBI NIH HHS/United States ; SP/19/1/34461/BHF_/British Heart Foundation/United Kingdom ; R01 HL127349/HL/NHLBI NIH HHS/United States ; U54 HL165443/HL/NHLBI NIH HHS/United States ; P30 DK043351/DK/NIDDK NIH HHS/United States ; U01 HL148856/HL/NHLBI NIH HHS/United States ; RC2 DK114784/DK/NIDDK NIH HHS/United States ; U19 AI116482/AI/NIAID NIH HHS/United States ; R01 HL145372/HL/NHLBI NIH HHS/United States ; U01 HL122700/HL/NHLBI NIH HHS/United States ; P01 AG049665/AG/NIA NIH HHS/United States ; T32 GM007753/GM/NIGMS NIH HHS/United States ; R01 DK117263/DK/NIDDK NIH HHS/United States ; K08 HL146943/HL/NHLBI NIH HHS/United States ; U19 AI135964/AI/NIAID NIH HHS/United States ; R01 HL080494/HL/NHLBI NIH HHS/United States ; R24 HD000836/HD/NICHD NIH HHS/United States ; R35 HL140039/HL/NHLBI NIH HHS/United States ; /WT_/Wellcome Trust/United Kingdom ; U24 AI118672/AI/NIAID NIH HHS/United States ; R01 HL133153/HL/NHLBI NIH HHS/United States ; U19 AI142784/AI/NIAID NIH HHS/United States ; R56 HL135124/HL/NHLBI NIH HHS/United States ; },
mesh = {Adult ; Aged ; Aged, 80 and over ; Alveolar Epithelial Cells/metabolism/virology ; Angiotensin-Converting Enzyme 2/genetics/metabolism ; COVID-19/*epidemiology/*genetics/pathology/virology ; Cathepsin L/genetics/metabolism ; Datasets as Topic/statistics & numerical data ; Demography ; Female ; Gene Expression Profiling/statistics & numerical data ; Host-Pathogen Interactions/*genetics ; Humans ; Lung/metabolism/virology ; Male ; Middle Aged ; Organ Specificity/genetics ; Respiratory System/metabolism/virology ; SARS-CoV-2/*physiology ; Sequence Analysis, RNA/methods/*statistics & numerical data ; Serine Endopeptidases/genetics/metabolism ; Single-Cell Analysis/methods/*statistics & numerical data ; *Virus Internalization ; },
abstract = {Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2[+]TMPRSS2[+] cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Aged
Aged, 80 and over
Alveolar Epithelial Cells/metabolism/virology
Angiotensin-Converting Enzyme 2/genetics/metabolism
COVID-19/*epidemiology/*genetics/pathology/virology
Cathepsin L/genetics/metabolism
Datasets as Topic/statistics & numerical data
Demography
Female
Gene Expression Profiling/statistics & numerical data
Host-Pathogen Interactions/*genetics
Humans
Lung/metabolism/virology
Male
Middle Aged
Organ Specificity/genetics
Respiratory System/metabolism/virology
SARS-CoV-2/*physiology
Sequence Analysis, RNA/methods/*statistics & numerical data
Serine Endopeptidases/genetics/metabolism
Single-Cell Analysis/methods/*statistics & numerical data
*Virus Internalization
RevDate: 2025-05-30
CmpDate: 2021-03-08
A living WHO guideline on drugs to prevent covid-19.
BMJ (Clinical research ed.), 372:n526.
CLINICAL QUESTION: What is the role of drugs in preventing covid-19? WHY DOES THIS MATTER?: There is widespread interest in whether drug interventions can be used for the prevention of covid-19, but there is uncertainty about which drugs, if any, are effective. The first version of this living guideline focuses on the evidence for hydroxychloroquine. Subsequent updates will cover other drugs being investigated for their role in the prevention of covid-19.
RECOMMENDATION: The guideline development panel made a strong recommendation against the use of hydroxychloroquine for individuals who do not have covid-19 (high certainty).
This living guideline is from the World Health Organization (WHO) and provides up to date covid-19 guidance to inform policy and practice worldwide. Magic Evidence Ecosystem Foundation (MAGIC) provided methodological support. A living systematic review with network analysis informed the recommendations. An international guideline development panel of content experts, clinicians, patients, an ethicist and methodologists produced recommendations following standards for trustworthy guideline development using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach.
The linked systematic review and network meta-analysis (6 trials and 6059 participants) found that hydroxychloroquine had a small or no effect on mortality and admission to hospital (high certainty evidence). There was a small or no effect on laboratory confirmed SARS-CoV-2 infection (moderate certainty evidence) but probably increased adverse events leading to discontinuation (moderate certainty evidence). The panel judged that almost all people would not consider this drug worthwhile. In addition, the panel decided that contextual factors such as resources, feasibility, acceptability, and equity for countries and healthcare systems were unlikely to alter the recommendation. The panel considers that this drug is no longer a research priority and that resources should rather be oriented to evaluate other more promising drugs to prevent covid-19.
UPDATES: This is a living guideline. New recommendations will be published in this article and signposted by update notices to this guideline.
READERS NOTE: This is the first version of the living guideline for drugs to prevent covid-19. It complements the WHO living guideline on drugs to treat covid-19. When citing this article, please consider adding the update number and date of access for clarity.
Additional Links: PMID-33649077
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33649077,
year = {2021},
author = {Lamontagne, F and Stegemann, M and Agarwal, A and Agoritsas, T and Siemieniuk, R and Rochwerg, B and Bartoszko, J and Askie, L and Macdonald, H and Al-Maslamani, M and Amin, W and Da Silva, ARA and Barragan, FAJ and Bausch, FJ and Burhan, E and Cecconi, M and Chacko, B and Chanda, D and Dat, VQ and Du, B and Geduld, H and Gee, P and Haider, M and Nerina, H and Hashimi, M and Jehan, F and Hui, D and Hunt, BJ and Ismail, M and Kabra, S and Kanda, S and Kawano-Dourado, L and Kim, YJ and Kissoon, N and Krishna, S and Kwizera, A and Lisboa, T and Leo, YS and Mahaka, I and Hela, M and Migliori, GB and Mino, G and Nsutebu, E and Pshenichnaya, N and Qadir, N and Ranganathan, SS and Sabzwari, S and Sarin, R and Shankar-Hari, M and Sharland, M and Shen, Y and Souza, JP and Tshokey, T and Ugarte, S and Uyeki, T and Venkatapuram, S and Wachinou, AP and Wijewickrama, A and Vuyiseka, D and Preller, J and Brignardello-Petersen, R and Kum, E and Qasim, A and Zeraatkar, D and Owen, A and Guyatt, G and Lytvyn, L and Jacobs, M and Vandvik, PO and Diaz, J},
title = {A living WHO guideline on drugs to prevent covid-19.},
journal = {BMJ (Clinical research ed.)},
volume = {372},
number = {},
pages = {n526},
doi = {10.1136/bmj.n526},
pmid = {33649077},
issn = {1756-1833},
support = {001/WHO_/World Health Organization/International ; },
mesh = {COVID-19/epidemiology/*prevention & control ; *Chemoprevention/methods/standards ; Clinical Decision-Making/methods ; Humans ; Hydroxychloroquine/*pharmacology ; Immunologic Factors/pharmacology ; *Risk Assessment ; SARS-CoV-2/drug effects ; Uncertainty ; World Health Organization ; },
abstract = {CLINICAL QUESTION: What is the role of drugs in preventing covid-19? WHY DOES THIS MATTER?: There is widespread interest in whether drug interventions can be used for the prevention of covid-19, but there is uncertainty about which drugs, if any, are effective. The first version of this living guideline focuses on the evidence for hydroxychloroquine. Subsequent updates will cover other drugs being investigated for their role in the prevention of covid-19.
RECOMMENDATION: The guideline development panel made a strong recommendation against the use of hydroxychloroquine for individuals who do not have covid-19 (high certainty).
This living guideline is from the World Health Organization (WHO) and provides up to date covid-19 guidance to inform policy and practice worldwide. Magic Evidence Ecosystem Foundation (MAGIC) provided methodological support. A living systematic review with network analysis informed the recommendations. An international guideline development panel of content experts, clinicians, patients, an ethicist and methodologists produced recommendations following standards for trustworthy guideline development using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach.
The linked systematic review and network meta-analysis (6 trials and 6059 participants) found that hydroxychloroquine had a small or no effect on mortality and admission to hospital (high certainty evidence). There was a small or no effect on laboratory confirmed SARS-CoV-2 infection (moderate certainty evidence) but probably increased adverse events leading to discontinuation (moderate certainty evidence). The panel judged that almost all people would not consider this drug worthwhile. In addition, the panel decided that contextual factors such as resources, feasibility, acceptability, and equity for countries and healthcare systems were unlikely to alter the recommendation. The panel considers that this drug is no longer a research priority and that resources should rather be oriented to evaluate other more promising drugs to prevent covid-19.
UPDATES: This is a living guideline. New recommendations will be published in this article and signposted by update notices to this guideline.
READERS NOTE: This is the first version of the living guideline for drugs to prevent covid-19. It complements the WHO living guideline on drugs to treat covid-19. When citing this article, please consider adding the update number and date of access for clarity.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/epidemiology/*prevention & control
*Chemoprevention/methods/standards
Clinical Decision-Making/methods
Humans
Hydroxychloroquine/*pharmacology
Immunologic Factors/pharmacology
*Risk Assessment
SARS-CoV-2/drug effects
Uncertainty
World Health Organization
RevDate: 2021-04-07
CmpDate: 2021-04-07
Guidelines for Conducting Virtual Cognitive Interviews During a Pandemic.
Journal of medical Internet research, 23(3):e25173.
The COVID-19 pandemic has challenged researchers working in physical contact with research participants. Cognitive interviews examine whether study components (most often questionnaire items) are worded or structured in a manner that allows study participants to interpret the items in a way intended by the researcher. We developed guidelines to conduct cognitive interviews virtually to accommodate interviewees who have limited access to the internet. The guidelines describe the essential communication and safety equipment requirements and outline a procedure for collecting responses while maintaining the safety of the participants and researchers. Furthermore, the guidelines provide suggestions regarding training of participants to use the technology, encouraging them to respond aloud (a potential challenge given that the researcher is not physically present with the participant), and testing and deploying the equipment prior to the interview. Finally, the guidelines emphasize the need to adapt the interview to the circumstances and anticipate potential problems that might arise.
Additional Links: PMID-33577464
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33577464,
year = {2021},
author = {Shepperd, JA and Pogge, G and Hunleth, JM and Ruiz, S and Waters, EA},
title = {Guidelines for Conducting Virtual Cognitive Interviews During a Pandemic.},
journal = {Journal of medical Internet research},
volume = {23},
number = {3},
pages = {e25173},
pmid = {33577464},
issn = {1438-8871},
support = {R01 HL137680/HL/NHLBI NIH HHS/United States ; },
mesh = {COVID-19/*epidemiology/psychology ; Guidelines as Topic ; Humans ; Pandemics ; Psychometrics ; SARS-CoV-2/isolation & purification ; Surveys and Questionnaires/*standards ; Telemedicine/*methods ; Virtual Reality Exposure Therapy/*methods ; },
abstract = {The COVID-19 pandemic has challenged researchers working in physical contact with research participants. Cognitive interviews examine whether study components (most often questionnaire items) are worded or structured in a manner that allows study participants to interpret the items in a way intended by the researcher. We developed guidelines to conduct cognitive interviews virtually to accommodate interviewees who have limited access to the internet. The guidelines describe the essential communication and safety equipment requirements and outline a procedure for collecting responses while maintaining the safety of the participants and researchers. Furthermore, the guidelines provide suggestions regarding training of participants to use the technology, encouraging them to respond aloud (a potential challenge given that the researcher is not physically present with the participant), and testing and deploying the equipment prior to the interview. Finally, the guidelines emphasize the need to adapt the interview to the circumstances and anticipate potential problems that might arise.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*epidemiology/psychology
Guidelines as Topic
Humans
Pandemics
Psychometrics
SARS-CoV-2/isolation & purification
Surveys and Questionnaires/*standards
Telemedicine/*methods
Virtual Reality Exposure Therapy/*methods
RevDate: 2022-10-05
CmpDate: 2022-01-11
Outcomes of in-hospital cardiac arrest in COVID-19 patients: A proportional prevalence meta-analysis.
Catheterization and cardiovascular interventions : official journal of the Society for Cardiac Angiography & Interventions, 99(1):1-8.
BACKGROUND: Limited epidemiological data are available on the outcomes of in-hospital cardiac arrest (CA) in COVID-19 patients.
METHODS: We performed literature search of PubMed, EMBASE, Cochrane, and Ovid to identify research articles that studied outcomes of in-hospital cardiac arrest in COVID-19 patients. The primary outcome was survival at discharge. Secondary outcomes included return of spontaneous circulation (ROSC) and types of cardiac arrest. Pooled percentages with a 95% confidence interval (CI) were calculated for the prevalence of outcomes.
RESULTS: A total of 7,891 COVID patients were included in the study. There were 621 (pooled prevalence 8%, 95% CI 4-13%) cardiac arrest patients. There were 52 (pooled prevalence 3.0%; 95% CI 0.0-10.0%) patients that survived at the time of discharge. ROSC was achieved in 202 (pooled prevalence 39%;95% CI 21.0-59.0%) patients. Mean time to ROSC was 7.74 (95% CI 7.51-7.98) min. The commonest rhythm at the time of cardiac arrest was pulseless electrical activity (pooled prevalence 46%; 95% 13-80%), followed by asystole (pooled prevalence 40%; 95% CI 6-80%). Unstable ventricular arrhythmia occurred in a minority of patients (pooled prevalence 8%; 95% CI 4-13%).
CONCLUSION: This pooled analysis of studies showed that the survival post in-hospital cardiac arrest in COVID patients is dismal despite adequate ROSC obtained at the time of resuscitation. Nonshockable rhythm cardiac arrest is commoner suggesting a non-cardiac cause while cardiac related etiology is uncommon. Future studies are needed to improve the survival in these patients.
Additional Links: PMID-33543564
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33543564,
year = {2022},
author = {Mir, T and Sattar, Y and Ahmad, J and Ullah, W and Shanah, L and Alraies, MC and Qureshi, WT},
title = {Outcomes of in-hospital cardiac arrest in COVID-19 patients: A proportional prevalence meta-analysis.},
journal = {Catheterization and cardiovascular interventions : official journal of the Society for Cardiac Angiography & Interventions},
volume = {99},
number = {1},
pages = {1-8},
pmid = {33543564},
issn = {1522-726X},
mesh = {*COVID-19 ; *Cardiopulmonary Resuscitation ; *Heart Arrest/diagnosis/epidemiology ; Hospitals ; Humans ; Prevalence ; SARS-CoV-2 ; Treatment Outcome ; },
abstract = {BACKGROUND: Limited epidemiological data are available on the outcomes of in-hospital cardiac arrest (CA) in COVID-19 patients.
METHODS: We performed literature search of PubMed, EMBASE, Cochrane, and Ovid to identify research articles that studied outcomes of in-hospital cardiac arrest in COVID-19 patients. The primary outcome was survival at discharge. Secondary outcomes included return of spontaneous circulation (ROSC) and types of cardiac arrest. Pooled percentages with a 95% confidence interval (CI) were calculated for the prevalence of outcomes.
RESULTS: A total of 7,891 COVID patients were included in the study. There were 621 (pooled prevalence 8%, 95% CI 4-13%) cardiac arrest patients. There were 52 (pooled prevalence 3.0%; 95% CI 0.0-10.0%) patients that survived at the time of discharge. ROSC was achieved in 202 (pooled prevalence 39%;95% CI 21.0-59.0%) patients. Mean time to ROSC was 7.74 (95% CI 7.51-7.98) min. The commonest rhythm at the time of cardiac arrest was pulseless electrical activity (pooled prevalence 46%; 95% 13-80%), followed by asystole (pooled prevalence 40%; 95% CI 6-80%). Unstable ventricular arrhythmia occurred in a minority of patients (pooled prevalence 8%; 95% CI 4-13%).
CONCLUSION: This pooled analysis of studies showed that the survival post in-hospital cardiac arrest in COVID patients is dismal despite adequate ROSC obtained at the time of resuscitation. Nonshockable rhythm cardiac arrest is commoner suggesting a non-cardiac cause while cardiac related etiology is uncommon. Future studies are needed to improve the survival in these patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19
*Cardiopulmonary Resuscitation
*Heart Arrest/diagnosis/epidemiology
Hospitals
Humans
Prevalence
SARS-CoV-2
Treatment Outcome
RevDate: 2024-03-30
CmpDate: 2021-02-25
Meta-analysis of host transcriptional responses to SARS-CoV-2 infection reveals their manifestation in human tumors.
Scientific reports, 11(1):2459.
A deeper understanding of the molecular biology of SARS-CoV-2 infection, including the host response to the virus, is urgently needed. Commonalities exist between the host immune response to viral infections and cancer. Here, we defined transcriptional signatures of SARS-CoV-2 infection involving hundreds of genes common across lung adenocarcinoma cell lines (A549, Calu-3) and normal human bronchial epithelial cells (NHBE), with additional signatures being specific to one or both adenocarcinoma lines. Cross-examining eight transcriptomic databases, we found that host transcriptional responses of lung adenocarcinoma cells to SARS-CoV-2 infection shared broad similarities with host responses to multiple viruses across different model systems and patient samples. Furthermore, these SARS-CoV-2 transcriptional signatures were manifested within specific subsets of human cancer, involving ~ 20% of cases across a wide range of histopathological types. These cancer subsets show immune cell infiltration and inflammation and involve pathways linked to the SARS-CoV-2 response, such as immune checkpoint, IL-6, type II interferon signaling, and NF-κB. The cell line data represented immune responses activated specifically within the cancer cells of the tumor. Common genes and pathways implicated as part of the viral host response point to therapeutic strategies that may apply to both SARS-CoV-2 and cancer.
Additional Links: PMID-33510359
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33510359,
year = {2021},
author = {Chen, F and Zhang, Y and Sucgang, R and Ramani, S and Corry, D and Kheradmand, F and Creighton, CJ},
title = {Meta-analysis of host transcriptional responses to SARS-CoV-2 infection reveals their manifestation in human tumors.},
journal = {Scientific reports},
volume = {11},
number = {1},
pages = {2459},
pmid = {33510359},
issn = {2045-2322},
support = {R01AI135803/NH/NIH HHS/United States ; R01 ES029442/ES/NIEHS NIH HHS/United States ; P30 CA125123/CA/NCI NIH HHS/United States ; R01 AI135803/AI/NIAID NIH HHS/United States ; CA125123/NH/NIH HHS/United States ; R01ES029442-01/NH/NIH HHS/United States ; U19 AI144297/AI/NIAID NIH HHS/United States ; U19AI144297/NH/NIH HHS/United States ; },
mesh = {A549 Cells ; Bronchi/metabolism ; COVID-19/*genetics/metabolism ; Epithelial Cells/metabolism/virology ; Host Microbial Interactions/*physiology ; Humans ; Immunity ; Lung Neoplasms/pathology/virology ; SARS-CoV-2/genetics/metabolism/*physiology ; Transcription, Genetic ; Transcriptome ; Virus Replication/genetics ; },
abstract = {A deeper understanding of the molecular biology of SARS-CoV-2 infection, including the host response to the virus, is urgently needed. Commonalities exist between the host immune response to viral infections and cancer. Here, we defined transcriptional signatures of SARS-CoV-2 infection involving hundreds of genes common across lung adenocarcinoma cell lines (A549, Calu-3) and normal human bronchial epithelial cells (NHBE), with additional signatures being specific to one or both adenocarcinoma lines. Cross-examining eight transcriptomic databases, we found that host transcriptional responses of lung adenocarcinoma cells to SARS-CoV-2 infection shared broad similarities with host responses to multiple viruses across different model systems and patient samples. Furthermore, these SARS-CoV-2 transcriptional signatures were manifested within specific subsets of human cancer, involving ~ 20% of cases across a wide range of histopathological types. These cancer subsets show immune cell infiltration and inflammation and involve pathways linked to the SARS-CoV-2 response, such as immune checkpoint, IL-6, type II interferon signaling, and NF-κB. The cell line data represented immune responses activated specifically within the cancer cells of the tumor. Common genes and pathways implicated as part of the viral host response point to therapeutic strategies that may apply to both SARS-CoV-2 and cancer.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
A549 Cells
Bronchi/metabolism
COVID-19/*genetics/metabolism
Epithelial Cells/metabolism/virology
Host Microbial Interactions/*physiology
Humans
Immunity
Lung Neoplasms/pathology/virology
SARS-CoV-2/genetics/metabolism/*physiology
Transcription, Genetic
Transcriptome
Virus Replication/genetics
RevDate: 2024-09-01
CmpDate: 2021-03-15
Associations between COVID-19 and skin conditions identified through epidemiology and genomic studies.
The Journal of allergy and clinical immunology, 147(3):857-869.e7.
BACKGROUND: Coronavirus disease 2019 (COVID-19) is commonly associated with skin manifestations, and may also exacerbate existing skin diseases, yet the relationship between COVID-19 and skin diseases remains unclear.
OBJECTIVE: By investigating this relationship through a multiomics approach, we sought to ascertain whether patients with skin conditions are more susceptible to COVID-19.
METHODS: We conducted an epidemiological study and then compared gene expression across 9 different inflammatory skin conditions and severe acute respiratory syndrome coronavirus 2-infected bronchial epithelial cell lines, and then performed a genome-wide association study transdisease meta-analysis between COVID-19 susceptibility and 2 skin diseases (psoriasis and atopic dermatitis).
RESULTS: Skin conditions, including psoriasis and atopic dermatitis, increase the risk of COVID-19 (odds ratio, 1.55; P = 1.4 × 10[-9]) but decrease the risk of mechanical ventilation (odds ratio, 0.22; P = 8.5 × 10[-5]). We observed significant overlap in gene expression between the infected normal bronchial epithelial cells and inflammatory skin diseases, such as psoriasis and atopic dermatitis. For genes that are commonly induced in both the severe acute respiratory syndrome coronavirus 2 infection and skin diseases, there are 4 S100 family members located in the epidermal differentiation complex, and we also identified the "IL-17 signaling pathway" (P = 4.9 × 10[-77]) as one of the most significantly enriched pathways. Furthermore, a shared genome-wide significant locus in the epidermal differentiation complex was identified between psoriasis and severe acute respiratory syndrome coronavirus 2 infection, with the lead marker being a significant expression quantitative trait locus for S100A12 (P = 3.3 × 10[-7]).
CONCLUSIONS: Together our findings suggest association between inflammatory skin conditions and higher risk of COVID-19, but with less severe course, and highlight shared components involved in anti-COVID-19 immune response.
Additional Links: PMID-33485957
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33485957,
year = {2021},
author = {Patrick, MT and Zhang, H and Wasikowski, R and Prens, EP and Weidinger, S and Gudjonsson, JE and Elder, JT and He, K and Tsoi, LC},
title = {Associations between COVID-19 and skin conditions identified through epidemiology and genomic studies.},
journal = {The Journal of allergy and clinical immunology},
volume = {147},
number = {3},
pages = {857-869.e7},
pmid = {33485957},
issn = {1097-6825},
support = {K08 AR060802/AR/NIAMS NIH HHS/United States ; R01 AR042742/AR/NIAMS NIH HHS/United States ; P30 AR075043/AR/NIAMS NIH HHS/United States ; K01 AR072129/AR/NIAMS NIH HHS/United States ; R01 AR054966/AR/NIAMS NIH HHS/United States ; R01 AR050511/AR/NIAMS NIH HHS/United States ; R01 AR065183/AR/NIAMS NIH HHS/United States ; R01 AR063611/AR/NIAMS NIH HHS/United States ; },
mesh = {Adolescent ; Adult ; Aged ; Aged, 80 and over ; *COVID-19/epidemiology/genetics/metabolism ; Cell Line ; *Dermatitis, Atopic/epidemiology/genetics/metabolism ; Female ; *Gene Expression Regulation ; *Genetic Predisposition to Disease ; Genome-Wide Association Study ; Genomics ; Humans ; Male ; Middle Aged ; *Psoriasis/epidemiology/genetics/metabolism ; *Quantitative Trait Loci ; Risk Factors ; *S100A12 Protein/biosynthesis/genetics ; SARS-CoV-2/genetics/*metabolism ; Skin/metabolism/virology ; },
abstract = {BACKGROUND: Coronavirus disease 2019 (COVID-19) is commonly associated with skin manifestations, and may also exacerbate existing skin diseases, yet the relationship between COVID-19 and skin diseases remains unclear.
OBJECTIVE: By investigating this relationship through a multiomics approach, we sought to ascertain whether patients with skin conditions are more susceptible to COVID-19.
METHODS: We conducted an epidemiological study and then compared gene expression across 9 different inflammatory skin conditions and severe acute respiratory syndrome coronavirus 2-infected bronchial epithelial cell lines, and then performed a genome-wide association study transdisease meta-analysis between COVID-19 susceptibility and 2 skin diseases (psoriasis and atopic dermatitis).
RESULTS: Skin conditions, including psoriasis and atopic dermatitis, increase the risk of COVID-19 (odds ratio, 1.55; P = 1.4 × 10[-9]) but decrease the risk of mechanical ventilation (odds ratio, 0.22; P = 8.5 × 10[-5]). We observed significant overlap in gene expression between the infected normal bronchial epithelial cells and inflammatory skin diseases, such as psoriasis and atopic dermatitis. For genes that are commonly induced in both the severe acute respiratory syndrome coronavirus 2 infection and skin diseases, there are 4 S100 family members located in the epidermal differentiation complex, and we also identified the "IL-17 signaling pathway" (P = 4.9 × 10[-77]) as one of the most significantly enriched pathways. Furthermore, a shared genome-wide significant locus in the epidermal differentiation complex was identified between psoriasis and severe acute respiratory syndrome coronavirus 2 infection, with the lead marker being a significant expression quantitative trait locus for S100A12 (P = 3.3 × 10[-7]).
CONCLUSIONS: Together our findings suggest association between inflammatory skin conditions and higher risk of COVID-19, but with less severe course, and highlight shared components involved in anti-COVID-19 immune response.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adolescent
Adult
Aged
Aged, 80 and over
*COVID-19/epidemiology/genetics/metabolism
Cell Line
*Dermatitis, Atopic/epidemiology/genetics/metabolism
Female
*Gene Expression Regulation
*Genetic Predisposition to Disease
Genome-Wide Association Study
Genomics
Humans
Male
Middle Aged
*Psoriasis/epidemiology/genetics/metabolism
*Quantitative Trait Loci
Risk Factors
*S100A12 Protein/biosynthesis/genetics
SARS-CoV-2/genetics/*metabolism
Skin/metabolism/virology
RevDate: 2025-04-01
CmpDate: 2021-04-26
National Psoriasis Foundation COVID-19 Task Force guidance for management of psoriatic disease during the pandemic: Version 2-Advances in psoriatic disease management, COVID-19 vaccines, and COVID-19 treatments.
Journal of the American Academy of Dermatology, 84(5):1254-1268.
OBJECTIVE: To update guidance regarding the management of psoriatic disease during the COVID-19 pandemic.
STUDY DESIGN: The task force (TF) includes 18 physician voting members with expertise in dermatology, rheumatology, epidemiology, infectious diseases, and critical care. The TF was supplemented by nonvoting members, which included fellows and National Psoriasis Foundation staff. Clinical questions relevant to the psoriatic disease community were informed by inquiries received by the National Psoriasis Foundation. A Delphi process was conducted.
RESULTS: The TF updated evidence for the original 22 statements and added 5 new recommendations. The average of the votes was within the category of agreement for all statements, 13 with high consensus and 14 with moderate consensus.
LIMITATIONS: The evidence behind many guidance statements is variable in quality and/or quantity.
CONCLUSIONS: These statements provide guidance for the treatment of patients with psoriatic disease on topics including how the disease and its treatments affect COVID-19 risk, how medical care can be optimized during the pandemic, what patients should do to lower their risk of getting infected with severe acute respiratory syndrome coronavirus 2 (including novel vaccination), and what they should do if they develop COVID-19. The guidance is a living document that is continuously updated by the TF as data emerge.
Additional Links: PMID-33422626
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33422626,
year = {2021},
author = {Gelfand, JM and Armstrong, AW and Bell, S and Anesi, GL and Blauvelt, A and Calabrese, C and Dommasch, ED and Feldman, SR and Gladman, D and Kircik, L and Lebwohl, M and Lo Re, V and Martin, G and Merola, JF and Scher, JU and Schwartzman, S and Treat, JR and Van Voorhees, AS and Ellebrecht, CT and Fenner, J and Ocon, A and Syed, MN and Weinstein, EJ and Gondo, G and Heydon, S and Koons, S and Ritchlin, CT},
title = {National Psoriasis Foundation COVID-19 Task Force guidance for management of psoriatic disease during the pandemic: Version 2-Advances in psoriatic disease management, COVID-19 vaccines, and COVID-19 treatments.},
journal = {Journal of the American Academy of Dermatology},
volume = {84},
number = {5},
pages = {1254-1268},
pmid = {33422626},
issn = {1097-6787},
support = {L30 HL154200/HL/NHLBI NIH HHS/United States ; T32 AI055435/AI/NIAID NIH HHS/United States ; },
mesh = {Biological Products/therapeutic use ; COVID-19/complications/epidemiology/*prevention & control ; *COVID-19 Vaccines ; Decision Making, Shared ; Evidence-Based Medicine ; Humans ; Immunologic Factors/therapeutic use ; Pandemics ; Psoriasis/complications/*drug therapy ; Risk Factors ; United States/epidemiology ; COVID-19 Drug Treatment ; },
abstract = {OBJECTIVE: To update guidance regarding the management of psoriatic disease during the COVID-19 pandemic.
STUDY DESIGN: The task force (TF) includes 18 physician voting members with expertise in dermatology, rheumatology, epidemiology, infectious diseases, and critical care. The TF was supplemented by nonvoting members, which included fellows and National Psoriasis Foundation staff. Clinical questions relevant to the psoriatic disease community were informed by inquiries received by the National Psoriasis Foundation. A Delphi process was conducted.
RESULTS: The TF updated evidence for the original 22 statements and added 5 new recommendations. The average of the votes was within the category of agreement for all statements, 13 with high consensus and 14 with moderate consensus.
LIMITATIONS: The evidence behind many guidance statements is variable in quality and/or quantity.
CONCLUSIONS: These statements provide guidance for the treatment of patients with psoriatic disease on topics including how the disease and its treatments affect COVID-19 risk, how medical care can be optimized during the pandemic, what patients should do to lower their risk of getting infected with severe acute respiratory syndrome coronavirus 2 (including novel vaccination), and what they should do if they develop COVID-19. The guidance is a living document that is continuously updated by the TF as data emerge.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Biological Products/therapeutic use
COVID-19/complications/epidemiology/*prevention & control
*COVID-19 Vaccines
Decision Making, Shared
Evidence-Based Medicine
Humans
Immunologic Factors/therapeutic use
Pandemics
Psoriasis/complications/*drug therapy
Risk Factors
United States/epidemiology
COVID-19 Drug Treatment
RevDate: 2023-11-10
CmpDate: 2021-11-17
Impact of colchicine on mortality in patients with COVID-19: A meta-analysis.
Hellenic journal of cardiology : HJC = Hellenike kardiologike epitheorese, 62(5):374-377.
Additional Links: PMID-33421583
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33421583,
year = {2021},
author = {Vrachatis, DA and Giannopoulos, GV and Giotaki, SG and Raisakis, K and Kossyvakis, C and Iliodromitis, KE and Reimers, B and Tousoulis, D and Cleman, M and Stefanadis, C and Lansky, A and Deftereos, SG},
title = {Impact of colchicine on mortality in patients with COVID-19: A meta-analysis.},
journal = {Hellenic journal of cardiology : HJC = Hellenike kardiologike epitheorese},
volume = {62},
number = {5},
pages = {374-377},
pmid = {33421583},
issn = {2241-5955},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {*COVID-19 ; *Colchicine/therapeutic use ; Humans ; SARS-CoV-2 ; Treatment Outcome ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*COVID-19
*Colchicine/therapeutic use
Humans
SARS-CoV-2
Treatment Outcome
RevDate: 2023-11-12
CmpDate: 2021-04-16
Lack of association of antihypertensive drugs with the risk and severity of COVID-19: A meta-analysis.
Journal of cardiology, 77(5):482-491.
BACKGROUND: The association of antihypertensive drugs with the risk and severity of COVID-19 remains unknown.
METHODS AND RESULTS: We systematically searched PubMed, MEDLINE, The Cochrane Library, Cochrane Central Register of Controlled Trials (CENTRAL), ClinicalTrials.gov, and medRxiv for publications before July 13, 2020. Cohort studies and case-control studies that contain information on the association of antihypertensive agents including angiotensin-converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), calcium-channel blockers (CCBs), β-blockers, and diuretics with the risk and severity of COVID-19 were selected. The random or fixed-effects models were used to pool the odds ratio (OR) with 95% confidence interval (CI) for the outcomes. The literature search yielded 53 studies that satisfied our inclusion criteria, which comprised 39 cohort studies and 14 case-control studies. These studies included a total of 2,100,587 participants. We observed no association between prior usage of antihypertensive medications including ACEIs/ARBs, CCBs, β-blockers, or diuretics and the risk and severity of COVID-19. Additionally, when only hypertensive patients were included, the severity and mortality were lower with prior usage of ACEIs/ARBs (overall OR of 0.81, 95% CI 0.66-0.99, p < 0.05 and overall OR of 0.77, 95% CI 0.66-0.91, p < 0.01).
CONCLUSIONS: Taken together, usage of antihypertensive drugs is not associated with the risk and severity of COVID-19. Based on the current available literature, it is not recommended to abstain from the usage of these drugs in COVID-19 patients.
REGISTRATION: The meta-analysis was registered on OSF (https://osf.io/ynd5g).
Additional Links: PMID-33168337
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33168337,
year = {2021},
author = {Ren, L and Yu, S and Xu, W and Overton, JL and Chiamvimonvat, N and Thai, PN},
title = {Lack of association of antihypertensive drugs with the risk and severity of COVID-19: A meta-analysis.},
journal = {Journal of cardiology},
volume = {77},
number = {5},
pages = {482-491},
pmid = {33168337},
issn = {1876-4738},
support = {T32 HL086350/HL/NHLBI NIH HHS/United States ; R01 HL085727/HL/NHLBI NIH HHS/United States ; I01 CX001490/CX/CSRD VA/United States ; R01 HL137228/HL/NHLBI NIH HHS/United States ; I01 BX000576/BX/BLRD VA/United States ; R01 HL085844/HL/NHLBI NIH HHS/United States ; },
mesh = {Adrenergic beta-Antagonists/therapeutic use ; Angiotensin Receptor Antagonists/*therapeutic use ; Angiotensin-Converting Enzyme Inhibitors/*therapeutic use ; Antihypertensive Agents/*adverse effects/*therapeutic use ; COVID-19/*epidemiology/prevention & control ; Calcium Channel Blockers/therapeutic use ; Case-Control Studies ; Diuretics/therapeutic use ; Humans ; Hypertension/complications ; SARS-CoV-2 ; *COVID-19 Drug Treatment ; },
abstract = {BACKGROUND: The association of antihypertensive drugs with the risk and severity of COVID-19 remains unknown.
METHODS AND RESULTS: We systematically searched PubMed, MEDLINE, The Cochrane Library, Cochrane Central Register of Controlled Trials (CENTRAL), ClinicalTrials.gov, and medRxiv for publications before July 13, 2020. Cohort studies and case-control studies that contain information on the association of antihypertensive agents including angiotensin-converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), calcium-channel blockers (CCBs), β-blockers, and diuretics with the risk and severity of COVID-19 were selected. The random or fixed-effects models were used to pool the odds ratio (OR) with 95% confidence interval (CI) for the outcomes. The literature search yielded 53 studies that satisfied our inclusion criteria, which comprised 39 cohort studies and 14 case-control studies. These studies included a total of 2,100,587 participants. We observed no association between prior usage of antihypertensive medications including ACEIs/ARBs, CCBs, β-blockers, or diuretics and the risk and severity of COVID-19. Additionally, when only hypertensive patients were included, the severity and mortality were lower with prior usage of ACEIs/ARBs (overall OR of 0.81, 95% CI 0.66-0.99, p < 0.05 and overall OR of 0.77, 95% CI 0.66-0.91, p < 0.01).
CONCLUSIONS: Taken together, usage of antihypertensive drugs is not associated with the risk and severity of COVID-19. Based on the current available literature, it is not recommended to abstain from the usage of these drugs in COVID-19 patients.
REGISTRATION: The meta-analysis was registered on OSF (https://osf.io/ynd5g).},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adrenergic beta-Antagonists/therapeutic use
Angiotensin Receptor Antagonists/*therapeutic use
Angiotensin-Converting Enzyme Inhibitors/*therapeutic use
Antihypertensive Agents/*adverse effects/*therapeutic use
COVID-19/*epidemiology/prevention & control
Calcium Channel Blockers/therapeutic use
Case-Control Studies
Diuretics/therapeutic use
Humans
Hypertension/complications
SARS-CoV-2
*COVID-19 Drug Treatment
RevDate: 2024-04-25
CmpDate: 2020-11-04
Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2020 Recommendations of the International Antiviral Society-USA Panel.
JAMA, 324(16):1651-1669.
IMPORTANCE: Data on the use of antiretroviral drugs, including new drugs and formulations, for the treatment and prevention of HIV infection continue to guide optimal practices.
OBJECTIVE: To evaluate new data and incorporate them into current recommendations for initiating HIV therapy, monitoring individuals starting on therapy, changing regimens, preventing HIV infection for those at risk, and special considerations for older people with HIV.
EVIDENCE REVIEW: New evidence was collected since the previous International Antiviral (formerly AIDS) Society-USA recommendations in 2018, including data published or presented at peer-reviewed scientific conferences through August 22, 2020. A volunteer panel of 15 experts in HIV research and patient care considered these data and updated previous recommendations.
FINDINGS: From 5316 citations about antiretroviral drugs identified, 549 were included to form the evidence basis for these recommendations. Antiretroviral therapy is recommended as soon as possible for all individuals with HIV who have detectable viremia. Most patients can start with a 3-drug regimen or now a 2-drug regimen, which includes an integrase strand transfer inhibitor. Effective options are available for patients who may be pregnant, those who have specific clinical conditions, such as kidney, liver, or cardiovascular disease, those who have opportunistic diseases, or those who have health care access issues. Recommended for the first time, a long-acting antiretroviral regimen injected once every 4 weeks for treatment or every 8 weeks pending approval by regulatory bodies and availability. For individuals at risk for HIV, preexposure prophylaxis with an oral regimen is recommended or, pending approval by regulatory bodies and availability, with a long-acting injection given every 8 weeks. Monitoring before and during therapy for effectiveness and safety is recommended. Switching therapy for virological failure is relatively rare at this time, and the recommendations for switching therapies for convenience and for other reasons are included. With the survival benefits provided by therapy, recommendations are made for older individuals with HIV. The current coronavirus disease 2019 pandemic poses particular challenges for HIV research, care, and efforts to end the HIV epidemic.
CONCLUSION AND RELEVANCE: Advances in HIV prevention and management with antiretroviral drugs continue to improve clinical care and outcomes among individuals at risk for and with HIV.
Additional Links: PMID-33052386
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid33052386,
year = {2020},
author = {Saag, MS and Gandhi, RT and Hoy, JF and Landovitz, RJ and Thompson, MA and Sax, PE and Smith, DM and Benson, CA and Buchbinder, SP and Del Rio, C and Eron, JJ and Fätkenheuer, G and Günthard, HF and Molina, JM and Jacobsen, DM and Volberding, PA},
title = {Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2020 Recommendations of the International Antiviral Society-USA Panel.},
journal = {JAMA},
volume = {324},
number = {16},
pages = {1651-1669},
pmid = {33052386},
issn = {1538-3598},
support = {P30 AI027763/AI/NIAID NIH HHS/United States ; },
mesh = {AIDS-Related Opportunistic Infections/drug therapy ; Age Factors ; Anti-Retroviral Agents/economics/*therapeutic use ; Betacoronavirus ; COVID-19 ; Comorbidity ; Coronavirus Infections/epidemiology ; Drug Administration Schedule ; Drug Costs ; Drug Resistance, Viral/genetics ; Drug Substitution/standards ; Drug Therapy, Combination/methods ; Female ; HIV Infections/blood/diagnosis/*drug therapy/*prevention & control ; Humans ; International Agencies ; Male ; Pandemics ; Pneumonia, Viral/epidemiology ; Polypharmacy ; Pre-Exposure Prophylaxis/methods ; Pregnancy ; Pregnancy Complications, Infectious/drug therapy ; RNA, Viral/blood ; SARS-CoV-2 ; Societies, Medical ; United States ; Viral Load/genetics ; },
abstract = {IMPORTANCE: Data on the use of antiretroviral drugs, including new drugs and formulations, for the treatment and prevention of HIV infection continue to guide optimal practices.
OBJECTIVE: To evaluate new data and incorporate them into current recommendations for initiating HIV therapy, monitoring individuals starting on therapy, changing regimens, preventing HIV infection for those at risk, and special considerations for older people with HIV.
EVIDENCE REVIEW: New evidence was collected since the previous International Antiviral (formerly AIDS) Society-USA recommendations in 2018, including data published or presented at peer-reviewed scientific conferences through August 22, 2020. A volunteer panel of 15 experts in HIV research and patient care considered these data and updated previous recommendations.
FINDINGS: From 5316 citations about antiretroviral drugs identified, 549 were included to form the evidence basis for these recommendations. Antiretroviral therapy is recommended as soon as possible for all individuals with HIV who have detectable viremia. Most patients can start with a 3-drug regimen or now a 2-drug regimen, which includes an integrase strand transfer inhibitor. Effective options are available for patients who may be pregnant, those who have specific clinical conditions, such as kidney, liver, or cardiovascular disease, those who have opportunistic diseases, or those who have health care access issues. Recommended for the first time, a long-acting antiretroviral regimen injected once every 4 weeks for treatment or every 8 weeks pending approval by regulatory bodies and availability. For individuals at risk for HIV, preexposure prophylaxis with an oral regimen is recommended or, pending approval by regulatory bodies and availability, with a long-acting injection given every 8 weeks. Monitoring before and during therapy for effectiveness and safety is recommended. Switching therapy for virological failure is relatively rare at this time, and the recommendations for switching therapies for convenience and for other reasons are included. With the survival benefits provided by therapy, recommendations are made for older individuals with HIV. The current coronavirus disease 2019 pandemic poses particular challenges for HIV research, care, and efforts to end the HIV epidemic.
CONCLUSION AND RELEVANCE: Advances in HIV prevention and management with antiretroviral drugs continue to improve clinical care and outcomes among individuals at risk for and with HIV.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
AIDS-Related Opportunistic Infections/drug therapy
Age Factors
Anti-Retroviral Agents/economics/*therapeutic use
Betacoronavirus
COVID-19
Comorbidity
Coronavirus Infections/epidemiology
Drug Administration Schedule
Drug Costs
Drug Resistance, Viral/genetics
Drug Substitution/standards
Drug Therapy, Combination/methods
Female
HIV Infections/blood/diagnosis/*drug therapy/*prevention & control
Humans
International Agencies
Male
Pandemics
Pneumonia, Viral/epidemiology
Polypharmacy
Pre-Exposure Prophylaxis/methods
Pregnancy
Pregnancy Complications, Infectious/drug therapy
RNA, Viral/blood
SARS-CoV-2
Societies, Medical
United States
Viral Load/genetics
RevDate: 2021-09-29
CmpDate: 2020-11-23
Continuous Glucose Monitors and Automated Insulin Dosing Systems in the Hospital Consensus Guideline.
Journal of diabetes science and technology, 14(6):1035-1064.
This article is the work product of the Continuous Glucose Monitor and Automated Insulin Dosing Systems in the Hospital Consensus Guideline Panel, which was organized by Diabetes Technology Society and met virtually on April 23, 2020. The guideline panel consisted of 24 international experts in the use of continuous glucose monitors (CGMs) and automated insulin dosing (AID) systems representing adult endocrinology, pediatric endocrinology, obstetrics and gynecology, advanced practice nursing, diabetes care and education, clinical chemistry, bioengineering, and product liability law. The panelists reviewed the medical literature pertaining to five topics: (1) continuation of home CGMs after hospitalization, (2) initiation of CGMs in the hospital, (3) continuation of AID systems in the hospital, (4) logistics and hands-on care of hospitalized patients using CGMs and AID systems, and (5) data management of CGMs and AID systems in the hospital. The panelists then developed three types of recommendations for each topic, including clinical practice (to use the technology optimally), research (to improve the safety and effectiveness of the technology), and hospital policies (to build an environment for facilitating use of these devices) for each of the five topics. The panelists voted on 78 proposed recommendations. Based on the panel vote, 77 recommendations were classified as either strong or mild. One recommendation failed to reach consensus. Additional research is needed on CGMs and AID systems in the hospital setting regarding device accuracy, practices for deployment, data management, and achievable outcomes. This guideline is intended to support these technologies for the management of hospitalized patients with diabetes.
Additional Links: PMID-32985262
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32985262,
year = {2020},
author = {Galindo, RJ and Umpierrez, GE and Rushakoff, RJ and Basu, A and Lohnes, S and Nichols, JH and Spanakis, EK and Espinoza, J and Palermo, NE and Awadjie, DG and Bak, L and Buckingham, B and Cook, CB and Freckmann, G and Heinemann, L and Hovorka, R and Mathioudakis, N and Newman, T and O'Neal, DN and Rickert, M and Sacks, DB and Seley, JJ and Wallia, A and Shang, T and Zhang, JY and Han, J and Klonoff, DC},
title = {Continuous Glucose Monitors and Automated Insulin Dosing Systems in the Hospital Consensus Guideline.},
journal = {Journal of diabetes science and technology},
volume = {14},
number = {6},
pages = {1035-1064},
pmid = {32985262},
issn = {1932-2968},
support = {I01 CX001825/CX/CSRD VA/United States ; K23 DK123384/DK/NIDDK NIH HHS/United States ; UL1 TR002378/TR/NCATS NIH HHS/United States ; P30 DK111024/DK/NIDDK NIH HHS/United States ; P50 FD006425/FD/FDA HHS/United States ; },
mesh = {Adult ; Blood Glucose/*analysis/drug effects/metabolism ; Blood Glucose Self-Monitoring/instrumentation/methods/standards ; COVID-19 ; Child ; Consensus ; Coronavirus Infections/blood/complications/epidemiology/therapy ; Diabetes Complications/blood/epidemiology/therapy ; Diabetes Mellitus/blood/epidemiology/therapy ; Drug Dosage Calculations ; *Equipment and Supplies/standards ; Female ; *Hospitalization ; Hospitals/standards ; Humans ; Insulin/*administration & dosage ; *Insulin Infusion Systems/standards ; Monitoring, Physiologic/*instrumentation/methods/standards ; Pandemics ; Pneumonia, Viral/blood/complications/epidemiology/therapy ; Pregnancy ; },
abstract = {This article is the work product of the Continuous Glucose Monitor and Automated Insulin Dosing Systems in the Hospital Consensus Guideline Panel, which was organized by Diabetes Technology Society and met virtually on April 23, 2020. The guideline panel consisted of 24 international experts in the use of continuous glucose monitors (CGMs) and automated insulin dosing (AID) systems representing adult endocrinology, pediatric endocrinology, obstetrics and gynecology, advanced practice nursing, diabetes care and education, clinical chemistry, bioengineering, and product liability law. The panelists reviewed the medical literature pertaining to five topics: (1) continuation of home CGMs after hospitalization, (2) initiation of CGMs in the hospital, (3) continuation of AID systems in the hospital, (4) logistics and hands-on care of hospitalized patients using CGMs and AID systems, and (5) data management of CGMs and AID systems in the hospital. The panelists then developed three types of recommendations for each topic, including clinical practice (to use the technology optimally), research (to improve the safety and effectiveness of the technology), and hospital policies (to build an environment for facilitating use of these devices) for each of the five topics. The panelists voted on 78 proposed recommendations. Based on the panel vote, 77 recommendations were classified as either strong or mild. One recommendation failed to reach consensus. Additional research is needed on CGMs and AID systems in the hospital setting regarding device accuracy, practices for deployment, data management, and achievable outcomes. This guideline is intended to support these technologies for the management of hospitalized patients with diabetes.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Blood Glucose/*analysis/drug effects/metabolism
Blood Glucose Self-Monitoring/instrumentation/methods/standards
COVID-19
Child
Consensus
Coronavirus Infections/blood/complications/epidemiology/therapy
Diabetes Complications/blood/epidemiology/therapy
Diabetes Mellitus/blood/epidemiology/therapy
Drug Dosage Calculations
*Equipment and Supplies/standards
Female
*Hospitalization
Hospitals/standards
Humans
Insulin/*administration & dosage
*Insulin Infusion Systems/standards
Monitoring, Physiologic/*instrumentation/methods/standards
Pandemics
Pneumonia, Viral/blood/complications/epidemiology/therapy
Pregnancy
RevDate: 2025-05-30
CmpDate: 2021-01-08
Effect of Acid Suppressants on the Risk of COVID-19: A Propensity Score-Matched Study Using UK Biobank.
Gastroenterology, 160(1):455-458.e5.
Additional Links: PMID-32980342
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32980342,
year = {2021},
author = {Fan, X and Liu, Z and Miyata, T and Dasarathy, S and Rotroff, DM and Wu, X and Poulsen, KL and Nagy, LE},
title = {Effect of Acid Suppressants on the Risk of COVID-19: A Propensity Score-Matched Study Using UK Biobank.},
journal = {Gastroenterology},
volume = {160},
number = {1},
pages = {455-458.e5},
pmid = {32980342},
issn = {1528-0012},
support = {U01 AA026938/AA/NIAAA NIH HHS/United States ; U01 DK061732/DK/NIDDK NIH HHS/United States ; MC_QA137853/MRC_/Medical Research Council/United Kingdom ; U01 AA021890/AA/NIAAA NIH HHS/United States ; R00 AA026648/AA/NIAAA NIH HHS/United States ; R01 AA027456/AA/NIAAA NIH HHS/United States ; R01 DK113196/DK/NIDDK NIH HHS/United States ; P50 AA024333/AA/NIAAA NIH HHS/United States ; MC_PC_17228/MRC_/Medical Research Council/United Kingdom ; KL2 TR002547/TR/NCATS NIH HHS/United States ; R01 GM119174/GM/NIGMS NIH HHS/United States ; K99 AA026648/AA/NIAAA NIH HHS/United States ; R21 AR071046/AR/NIAMS NIH HHS/United States ; },
mesh = {Adult ; Aged ; COVID-19/diagnosis/*epidemiology/mortality ; Female ; Histamine H2 Antagonists/adverse effects/*therapeutic use ; Humans ; Male ; Middle Aged ; Prevalence ; Prognosis ; Propensity Score ; Proton Pump Inhibitors/adverse effects/*therapeutic use ; Risk Assessment ; Risk Factors ; Time Factors ; United Kingdom/epidemiology ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adult
Aged
COVID-19/diagnosis/*epidemiology/mortality
Female
Histamine H2 Antagonists/adverse effects/*therapeutic use
Humans
Male
Middle Aged
Prevalence
Prognosis
Propensity Score
Proton Pump Inhibitors/adverse effects/*therapeutic use
Risk Assessment
Risk Factors
Time Factors
United Kingdom/epidemiology
RevDate: 2022-12-07
CmpDate: 2021-01-28
Convalescent Plasma for the Treatment of COVID-19: Perspectives of the National Institutes of Health COVID-19 Treatment Guidelines Panel.
Annals of internal medicine, 174(1):93-95.
In the United States, the efficacy and safety of convalescent plasma for treating coronavirus disease 2019 (COVID-19) is currently being tested in randomized placebo-controlled clinical trials. Treatment of individual patients with COVID-19 with convalescent plasma outside such trials is also now permitted through U.S. Food and Drug Administration Emergency Use Authorization. Here, members of the National Institutes of Health COVID-19 Treatment Guidelines Panel provide their views regarding use of convalescent plasma for treating COVID-19.
Additional Links: PMID-32976026
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32976026,
year = {2021},
author = {Pau, AK and Aberg, J and Baker, J and Belperio, PS and Coopersmith, C and Crew, P and Grund, B and Gulick, RM and Harrison, C and Kim, A and Lane, HC and Masur, H and Sheikh, V and Singh, K and Yazdany, J and Tebas, P and , },
title = {Convalescent Plasma for the Treatment of COVID-19: Perspectives of the National Institutes of Health COVID-19 Treatment Guidelines Panel.},
journal = {Annals of internal medicine},
volume = {174},
number = {1},
pages = {93-95},
pmid = {32976026},
issn = {1539-3704},
support = {T32 DA023356/DA/NIDA NIH HHS/United States ; },
mesh = {COVID-19/*immunology/*therapy ; Humans ; Immunization, Passive/methods ; National Institutes of Health (U.S.) ; Plasma/*immunology ; SARS-CoV-2 ; United States ; COVID-19 Serotherapy ; },
abstract = {In the United States, the efficacy and safety of convalescent plasma for treating coronavirus disease 2019 (COVID-19) is currently being tested in randomized placebo-controlled clinical trials. Treatment of individual patients with COVID-19 with convalescent plasma outside such trials is also now permitted through U.S. Food and Drug Administration Emergency Use Authorization. Here, members of the National Institutes of Health COVID-19 Treatment Guidelines Panel provide their views regarding use of convalescent plasma for treating COVID-19.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*immunology/*therapy
Humans
Immunization, Passive/methods
National Institutes of Health (U.S.)
Plasma/*immunology
SARS-CoV-2
United States
COVID-19 Serotherapy
RevDate: 2023-06-15
CmpDate: 2020-12-01
National Psoriasis Foundation COVID-19 Task Force Guidance for Management of Psoriatic Disease During the Pandemic: Version 1.
Journal of the American Academy of Dermatology, 83(6):1704-1716.
OBJECTIVE: To provide guidance about management of psoriatic disease during the coronavirus disease 2019 (COVID-19) pandemic.
STUDY DESIGN: A task force (TF) of 18 physician voting members with expertise in dermatology, rheumatology, epidemiology, infectious diseases, and critical care was convened. The TF was supplemented by nonvoting members, which included fellows and National Psoriasis Foundation (NPF) staff. Clinical questions relevant to the psoriatic disease community were informed by questions received by the NPF. A Delphi process was conducted.
RESULTS: The TF approved 22 guidance statements. The average of the votes was within the category of agreement for all statements. All guidance statements proposed were recommended, 9 with high consensus and 13 with moderate consensus.
LIMITATIONS: The evidence behind many guidance statements is limited in quality.
CONCLUSION: These statements provide guidance for the management of patients with psoriatic disease on topics ranging from how the disease and its treatments impact COVID-19 risk and outcome, how medical care can be optimized during the pandemic, what patients should do to lower their risk of getting infected with severe acute respiratory syndrome coronavirus 2 and what they should do if they develop COVID-19. The guidance is intended to be a living document that will be updated by the TF as data emerge.
Additional Links: PMID-32891785
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32891785,
year = {2020},
author = {Gelfand, JM and Armstrong, AW and Bell, S and Anesi, GL and Blauvelt, A and Calabrese, C and Dommasch, ED and Feldman, SR and Gladman, D and Kircik, L and Lebwohl, M and Lo Re, V and Martin, G and Merola, JF and Scher, JU and Schwartzman, S and Treat, JR and Van Voorhees, AS and Ellebrecht, CT and Fenner, J and Ocon, A and Syed, MN and Weinstein, EJ and Smith, J and Gondo, G and Heydon, S and Koons, S and Ritchlin, CT},
title = {National Psoriasis Foundation COVID-19 Task Force Guidance for Management of Psoriatic Disease During the Pandemic: Version 1.},
journal = {Journal of the American Academy of Dermatology},
volume = {83},
number = {6},
pages = {1704-1716},
pmid = {32891785},
issn = {1097-6787},
support = {K12 HS026372/HS/AHRQ HHS/United States ; L30 HL154200/HL/NHLBI NIH HHS/United States ; T32 AR007465/AR/NIAMS NIH HHS/United States ; },
mesh = {Advisory Committees/standards ; Betacoronavirus/immunology/pathogenicity ; COVID-19 ; Consensus ; Coronavirus Infections/*epidemiology/immunology/prevention & control/virology ; Critical Care/standards ; Delphi Technique ; Dermatology/standards ; Epidemiology/standards ; Humans ; Immunosuppressive Agents/*adverse effects ; Infectious Disease Medicine/standards ; Organizations, Nonprofit/organization & administration/*standards ; Pandemics/prevention & control ; Pneumonia, Viral/*epidemiology/immunology/prevention & control/virology ; Psoriasis/complications/*drug therapy/immunology ; Rheumatology/standards ; SARS-CoV-2 ; United States/epidemiology ; },
abstract = {OBJECTIVE: To provide guidance about management of psoriatic disease during the coronavirus disease 2019 (COVID-19) pandemic.
STUDY DESIGN: A task force (TF) of 18 physician voting members with expertise in dermatology, rheumatology, epidemiology, infectious diseases, and critical care was convened. The TF was supplemented by nonvoting members, which included fellows and National Psoriasis Foundation (NPF) staff. Clinical questions relevant to the psoriatic disease community were informed by questions received by the NPF. A Delphi process was conducted.
RESULTS: The TF approved 22 guidance statements. The average of the votes was within the category of agreement for all statements. All guidance statements proposed were recommended, 9 with high consensus and 13 with moderate consensus.
LIMITATIONS: The evidence behind many guidance statements is limited in quality.
CONCLUSION: These statements provide guidance for the management of patients with psoriatic disease on topics ranging from how the disease and its treatments impact COVID-19 risk and outcome, how medical care can be optimized during the pandemic, what patients should do to lower their risk of getting infected with severe acute respiratory syndrome coronavirus 2 and what they should do if they develop COVID-19. The guidance is intended to be a living document that will be updated by the TF as data emerge.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Advisory Committees/standards
Betacoronavirus/immunology/pathogenicity
COVID-19
Consensus
Coronavirus Infections/*epidemiology/immunology/prevention & control/virology
Critical Care/standards
Delphi Technique
Dermatology/standards
Epidemiology/standards
Humans
Immunosuppressive Agents/*adverse effects
Infectious Disease Medicine/standards
Organizations, Nonprofit/organization & administration/*standards
Pandemics/prevention & control
Pneumonia, Viral/*epidemiology/immunology/prevention & control/virology
Psoriasis/complications/*drug therapy/immunology
Rheumatology/standards
SARS-CoV-2
United States/epidemiology
RevDate: 2025-05-30
CmpDate: 2020-09-16
A living WHO guideline on drugs for covid-19.
BMJ (Clinical research ed.), 370:m3379.
UPDATES: This is the fourteenth version (thirteenth update) of the living guideline, replacing earlier versions (available as data supplements). New recommendations will be published as updates to this guideline.
CLINICAL QUESTION: What is the role of drugs in the treatment of patients with covid-19?
CONTEXT: The evidence base for therapeutics for covid-19 is evolving with numerous randomised controlled trials (RCTs) recently completed and underway. Emerging SARS-CoV-2 variants and subvariants are changing the role of therapeutics.
WHAT IS NEW?: The guideline development group (GDG) defined 1.5% as a new threshold for an important reduction in risk of hospitalisation in patients with non-severe covid-19. Combined with updated baseline risk estimates, this resulted in stratification into patients at low, moderate, and high risk for hospitalisation. New recommendations were added for moderate risk of hospitalisation for nirmatrelvir/ritonavir, and for moderate and low risk of hospitalisation for molnupiravir and remdesivir. New pharmacokinetic evidence was included for nirmatrelvir/ritonavir and molnupiravir, supporting existing recommendations for patients at high risk of hospitalisation. The recommendation for ivermectin in patients with non-severe illness was updated in light of additional trial evidence which reduced the high degree of uncertainty informing previous guidance. A new recommendation was made against the antiviral agent VV116 for patients with non-severe and with severe or critical illness outside of randomised clinical trials based on one RCT comparing the drug with nirmatrelvir/ritonavir. The structure of the guideline publication has also been changed; recommendations are now ordered by severity of covid-19.
ABOUT THIS GUIDELINE: This living guideline from the World Health Organization (WHO) incorporates new evidence to dynamically update recommendations for covid-19 therapeutics. The GDG typically evaluates a therapy when the WHO judges sufficient evidence is available to make a recommendation. While the GDG takes an individual patient perspective in making recommendations, it also considers resource implications, acceptability, feasibility, equity, and human rights. This guideline was developed according to standards and methods for trustworthy guidelines, making use of an innovative process to achieve efficiency in dynamic updating of recommendations. The methods are aligned with the WHO Handbook for Guideline Development and according to a pre-approved protocol (planning proposal) by the Guideline Review Committee (GRC). A box at the end of the article outlines key methodological aspects of the guideline process. MAGIC Evidence Ecosystem Foundation provides methodological support, including the coordination of living systematic reviews with network meta-analyses to inform the recommendations. The full version of the guideline is available online in MAGICapp and in PDF on the WHO website, with a summary version here in The BMJ. These formats should facilitate adaptation, which is strongly encouraged by WHO to contextualise recommendations in a healthcare system to maximise impact.
FUTURE RECOMMENDATIONS: Recommendations on anticoagulation are planned for the next update to this guideline. Updated data regarding systemic corticosteroids, azithromycin, favipiravir and umefenovir for non-severe illness, and convalescent plasma and statin therapy for severe or critical illness, are planned for review in upcoming guideline iterations.
Additional Links: PMID-32887691
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32887691,
year = {2020},
author = {Agarwal, A and Hunt, B and Stegemann, M and Rochwerg, B and Lamontagne, F and Siemieniuk, RA and Agoritsas, T and Askie, L and Lytvyn, L and Leo, YS and Macdonald, H and Zeng, L and Alhadyan, A and Muna, A and Amin, W and da Silva, ARA and Aryal, D and Barragan, FAJ and Bausch, FJ and Burhan, E and Calfee, CS and Cecconi, M and Chacko, B and Chanda, D and Dat, VQ and De Sutter, A and Du, B and Freedman, S and Geduld, H and Gee, P and Haider, M and Gotte, M and Harley, N and Hashimi, M and Hui, D and Ismail, M and Jehan, F and Kabra, SK and Kanda, S and Kim, YJ and Kissoon, N and Krishna, S and Kuppalli, K and Kwizera, A and Lado Castro-Rial, M and Lisboa, T and Lodha, R and Mahaka, I and Manai, H and Mendelson, M and Migliori, GB and Mino, G and Nsutebu, E and Peter, J and Preller, J and Pshenichnaya, N and Qadir, N and Ranganathan, SS and Relan, P and Rylance, J and Sabzwari, S and Sarin, R and Shankar-Hari, M and Sharland, M and Shen, Y and Souza, JP and Swanstrom, R and Tshokey, T and Ugarte, S and Uyeki, T and Evangelina, VC and Venkatapuram, S and Vuyiseka, D and Wijewickrama, A and Tran, L and Zeraatkar, D and Bartoszko, JJ and Ge, L and Brignardello-Petersen, R and Owen, A and Guyatt, G and Diaz, J and Kawano-Dourado, L and Jacobs, M and Vandvik, PO},
title = {A living WHO guideline on drugs for covid-19.},
journal = {BMJ (Clinical research ed.)},
volume = {370},
number = {},
pages = {m3379},
doi = {10.1136/bmj.m3379},
pmid = {32887691},
issn = {1756-1833},
support = {001/WHO_/World Health Organization/International ; RP-2016-07-012/DH_/Department of Health/United Kingdom ; },
mesh = {Adrenal Cortex Hormones/*therapeutic use ; *Betacoronavirus ; COVID-19 ; Coronavirus Infections/*drug therapy ; Humans ; Pandemics ; Pneumonia, Viral/*drug therapy ; SARS-CoV-2 ; World Health Organization ; COVID-19 Drug Treatment ; },
abstract = {UPDATES: This is the fourteenth version (thirteenth update) of the living guideline, replacing earlier versions (available as data supplements). New recommendations will be published as updates to this guideline.
CLINICAL QUESTION: What is the role of drugs in the treatment of patients with covid-19?
CONTEXT: The evidence base for therapeutics for covid-19 is evolving with numerous randomised controlled trials (RCTs) recently completed and underway. Emerging SARS-CoV-2 variants and subvariants are changing the role of therapeutics.
WHAT IS NEW?: The guideline development group (GDG) defined 1.5% as a new threshold for an important reduction in risk of hospitalisation in patients with non-severe covid-19. Combined with updated baseline risk estimates, this resulted in stratification into patients at low, moderate, and high risk for hospitalisation. New recommendations were added for moderate risk of hospitalisation for nirmatrelvir/ritonavir, and for moderate and low risk of hospitalisation for molnupiravir and remdesivir. New pharmacokinetic evidence was included for nirmatrelvir/ritonavir and molnupiravir, supporting existing recommendations for patients at high risk of hospitalisation. The recommendation for ivermectin in patients with non-severe illness was updated in light of additional trial evidence which reduced the high degree of uncertainty informing previous guidance. A new recommendation was made against the antiviral agent VV116 for patients with non-severe and with severe or critical illness outside of randomised clinical trials based on one RCT comparing the drug with nirmatrelvir/ritonavir. The structure of the guideline publication has also been changed; recommendations are now ordered by severity of covid-19.
ABOUT THIS GUIDELINE: This living guideline from the World Health Organization (WHO) incorporates new evidence to dynamically update recommendations for covid-19 therapeutics. The GDG typically evaluates a therapy when the WHO judges sufficient evidence is available to make a recommendation. While the GDG takes an individual patient perspective in making recommendations, it also considers resource implications, acceptability, feasibility, equity, and human rights. This guideline was developed according to standards and methods for trustworthy guidelines, making use of an innovative process to achieve efficiency in dynamic updating of recommendations. The methods are aligned with the WHO Handbook for Guideline Development and according to a pre-approved protocol (planning proposal) by the Guideline Review Committee (GRC). A box at the end of the article outlines key methodological aspects of the guideline process. MAGIC Evidence Ecosystem Foundation provides methodological support, including the coordination of living systematic reviews with network meta-analyses to inform the recommendations. The full version of the guideline is available online in MAGICapp and in PDF on the WHO website, with a summary version here in The BMJ. These formats should facilitate adaptation, which is strongly encouraged by WHO to contextualise recommendations in a healthcare system to maximise impact.
FUTURE RECOMMENDATIONS: Recommendations on anticoagulation are planned for the next update to this guideline. Updated data regarding systemic corticosteroids, azithromycin, favipiravir and umefenovir for non-severe illness, and convalescent plasma and statin therapy for severe or critical illness, are planned for review in upcoming guideline iterations.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adrenal Cortex Hormones/*therapeutic use
*Betacoronavirus
COVID-19
Coronavirus Infections/*drug therapy
Humans
Pandemics
Pneumonia, Viral/*drug therapy
SARS-CoV-2
World Health Organization
COVID-19 Drug Treatment
RevDate: 2024-03-29
CmpDate: 2020-08-27
Guidance for Rebooting Electrophysiology Through the COVID-19 Pandemic From the Heart Rhythm Society and the American Heart Association Electrocardiography and Arrhythmias Committee of the Council on Clinical Cardiology: Endorsed by the American College of Cardiology.
JACC. Clinical electrophysiology, 6(8):1053-1066.
Coronavirus disease 2019 (COVID-19) has presented substantial challenges to patient care and impacted health care delivery, including cardiac electrophysiology practice throughout the globe. Based upon the undetermined course and regional variability of the pandemic, there is uncertainty as to how and when to resume and deliver electrophysiology services for arrhythmia patients. This joint document from representatives of the Heart Rhythm Society, American Heart Association, and American College of Cardiology seeks to provide guidance for clinicians and institutions reestablishing safe electrophysiological care. To achieve this aim, we address regional and local COVID-19 disease status, the role of viral screening and serologic testing, return-to-work considerations for exposed or infected health care workers, risk stratification and management strategies based on COVID-19 disease burden, institutional preparedness for resumption of elective procedures, patient preparation and communication, prioritization of procedures, and development of outpatient and periprocedural care pathways.
Additional Links: PMID-32819525
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32819525,
year = {2020},
author = {Lakkireddy, DR and Chung, MK and Deering, TF and Gopinathannair, R and Albert, CM and Epstein, LM and Harding, CV and Hurwitz, JL and Jeffery, CC and Krahn, AD and Kusumoto, FM and Lampert, R and Mansour, M and Natale, A and Patton, KK and Seiler, A and Shah, MJ and Wang, PJ and Russo, AM},
title = {Guidance for Rebooting Electrophysiology Through the COVID-19 Pandemic From the Heart Rhythm Society and the American Heart Association Electrocardiography and Arrhythmias Committee of the Council on Clinical Cardiology: Endorsed by the American College of Cardiology.},
journal = {JACC. Clinical electrophysiology},
volume = {6},
number = {8},
pages = {1053-1066},
pmid = {32819525},
issn = {2405-5018},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Ambulatory Care ; American Heart Association ; Arrhythmias, Cardiac/*diagnosis/*therapy ; Betacoronavirus ; COVID-19 ; COVID-19 Testing ; *Cardiology ; Clinical Laboratory Techniques ; Coronavirus Infections/diagnosis/*epidemiology/prevention & control ; Decision Making, Shared ; *Delivery of Health Care ; *Electrophysiologic Techniques, Cardiac ; Health Personnel ; Humans ; Mass Screening ; Organizational Policy ; Pandemics/prevention & control ; Patient Selection ; Personal Protective Equipment/supply & distribution ; Pneumonia, Viral/diagnosis/*epidemiology/prevention & control ; Return to Work ; Risk Assessment ; SARS-CoV-2 ; Telemedicine ; United States/epidemiology ; },
abstract = {Coronavirus disease 2019 (COVID-19) has presented substantial challenges to patient care and impacted health care delivery, including cardiac electrophysiology practice throughout the globe. Based upon the undetermined course and regional variability of the pandemic, there is uncertainty as to how and when to resume and deliver electrophysiology services for arrhythmia patients. This joint document from representatives of the Heart Rhythm Society, American Heart Association, and American College of Cardiology seeks to provide guidance for clinicians and institutions reestablishing safe electrophysiological care. To achieve this aim, we address regional and local COVID-19 disease status, the role of viral screening and serologic testing, return-to-work considerations for exposed or infected health care workers, risk stratification and management strategies based on COVID-19 disease burden, institutional preparedness for resumption of elective procedures, patient preparation and communication, prioritization of procedures, and development of outpatient and periprocedural care pathways.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Ambulatory Care
American Heart Association
Arrhythmias, Cardiac/*diagnosis/*therapy
Betacoronavirus
COVID-19
COVID-19 Testing
*Cardiology
Clinical Laboratory Techniques
Coronavirus Infections/diagnosis/*epidemiology/prevention & control
Decision Making, Shared
*Delivery of Health Care
*Electrophysiologic Techniques, Cardiac
Health Personnel
Humans
Mass Screening
Organizational Policy
Pandemics/prevention & control
Patient Selection
Personal Protective Equipment/supply & distribution
Pneumonia, Viral/diagnosis/*epidemiology/prevention & control
Return to Work
Risk Assessment
SARS-CoV-2
Telemedicine
United States/epidemiology
RevDate: 2024-09-06
CmpDate: 2020-12-17
Guidelines for COVID-19 Management in Hematopoietic Cell Transplantation and Cellular Therapy Recipients.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, 26(11):1983-1994.
There are currently limited data on the epidemiology, clinical manifestations, and optimal management of Coronavirus Disease 2019 (COVID-19) in hematopoietic cell transplantation and cellular therapy recipients. Given the experience with other respiratory viruses, we anticipate that patients may develop severe clinical disease and thus provide the following general principles for cancer centers across the nation. These guidelines were developed by members of the American Society for Transplantation and Cellular Therapy Infectious Diseases Special Interest Group. Specific practices may vary depending on local epidemiology and testing capacity, and the guidance provided in this document may change as new information becomes available.
Additional Links: PMID-32736007
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32736007,
year = {2020},
author = {Waghmare, A and Abidi, MZ and Boeckh, M and Chemaly, RF and Dadwal, S and El Boghdadly, Z and Kamboj, M and Papanicolaou, GA and Pergam, SA and Shahid, Z},
title = {Guidelines for COVID-19 Management in Hematopoietic Cell Transplantation and Cellular Therapy Recipients.},
journal = {Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation},
volume = {26},
number = {11},
pages = {1983-1994},
pmid = {32736007},
issn = {1523-6536},
support = {P30 CA016672/CA/NCI NIH HHS/United States ; U01 AI132004/AI/NIAID NIH HHS/United States ; },
mesh = {Adrenal Cortex Hormones/*therapeutic use ; Antiviral Agents/*therapeutic use ; COVID-19/*diagnosis/immunology/therapy/virology ; COVID-19 Testing ; Cell- and Tissue-Based Therapy/methods ; Clinical Decision-Making ; Disease Management ; Disinfection/methods ; Hematopoietic Stem Cell Transplantation/*methods ; Humans ; Immunization, Passive ; Immunologic Factors/*therapeutic use ; Neoplasms/immunology/*therapy/virology ; Physical Distancing ; Risk Assessment ; SARS-CoV-2/drug effects/pathogenicity ; Time Factors ; COVID-19 Serotherapy ; *COVID-19 Drug Treatment ; },
abstract = {There are currently limited data on the epidemiology, clinical manifestations, and optimal management of Coronavirus Disease 2019 (COVID-19) in hematopoietic cell transplantation and cellular therapy recipients. Given the experience with other respiratory viruses, we anticipate that patients may develop severe clinical disease and thus provide the following general principles for cancer centers across the nation. These guidelines were developed by members of the American Society for Transplantation and Cellular Therapy Infectious Diseases Special Interest Group. Specific practices may vary depending on local epidemiology and testing capacity, and the guidance provided in this document may change as new information becomes available.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adrenal Cortex Hormones/*therapeutic use
Antiviral Agents/*therapeutic use
COVID-19/*diagnosis/immunology/therapy/virology
COVID-19 Testing
Cell- and Tissue-Based Therapy/methods
Clinical Decision-Making
Disease Management
Disinfection/methods
Hematopoietic Stem Cell Transplantation/*methods
Humans
Immunization, Passive
Immunologic Factors/*therapeutic use
Neoplasms/immunology/*therapy/virology
Physical Distancing
Risk Assessment
SARS-CoV-2/drug effects/pathogenicity
Time Factors
COVID-19 Serotherapy
*COVID-19 Drug Treatment
RevDate: 2021-06-08
CmpDate: 2020-12-04
Adapting care for older cancer patients during the COVID-19 pandemic: Recommendations from the International Society of Geriatric Oncology (SIOG) COVID-19 Working Group.
Journal of geriatric oncology, 11(8):1190-1198.
The COVID-19 pandemic poses a barrier to equal and evidence-based management of cancer in older adults. The International Society of Geriatric Oncology (SIOG) formed a panel of experts to develop consensus recommendations on the implications of the pandemic on several aspects of cancer care in this age group including geriatric assessment (GA), surgery, radiotherapy, systemic treatment, palliative care and research. Age and cancer diagnosis are significant predictors of adverse outcomes of the COVID-19 infection. In this setting, GA is particularly valuable to drive decision-making. GA may aid estimating physiologic reserve and adaptive capability, assessing risk-benefits of either providing or temporarily withholding treatments, and determining patient preferences to help inform treatment decisions. In a resource-constrained setting, geriatric screening tools may be administered remotely to identify patients requiring comprehensive GA. Tele-health is also crucial to ensure adequate continuity of care and minimize the risk of infection exposure. In general, therapeutic decisions should favor the most effective and least invasive approach with the lowest risk of adverse outcomes. In selected cases, this might require deferring or omitting surgery, radiotherapy or systemic treatments especially where benefits are marginal and alternative safe therapeutic options are available. Ongoing research is necessary to expand knowledge of the management of cancer in older adults. However, the pandemic presents a significant barrier and efforts should be made to ensure equitable access to clinical trials and prospective data collection to elucidate the outcomes of COVID-19 in this population.
Additional Links: PMID-32709495
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32709495,
year = {2020},
author = {Battisti, NML and Mislang, AR and Cooper, L and O'Donovan, A and Audisio, RA and Cheung, KL and Sarrió, RG and Stauder, R and Soto-Perez-de-Celis, E and Jaklitsch, M and Williams, GR and O'Hanlon, S and Alam, M and Cairo, C and Colloca, G and Gil, LA and Sattar, S and Kantilal, K and Russo, C and Lichtman, SM and Brain, E and Kanesvaran, R and Wildiers, H},
title = {Adapting care for older cancer patients during the COVID-19 pandemic: Recommendations from the International Society of Geriatric Oncology (SIOG) COVID-19 Working Group.},
journal = {Journal of geriatric oncology},
volume = {11},
number = {8},
pages = {1190-1198},
pmid = {32709495},
issn = {1879-4076},
support = {K08 CA234225/CA/NCI NIH HHS/United States ; P30 CA008748/CA/NCI NIH HHS/United States ; },
mesh = {Aged ; COVID-19/*complications/epidemiology ; Consensus ; *Geriatric Assessment ; Geriatrics/standards ; Humans ; Medical Oncology/standards ; Neoplasms/*complications/radiotherapy/surgery/*therapy ; Palliative Care/methods ; Pandemics ; Risk Assessment ; Societies, Medical ; },
abstract = {The COVID-19 pandemic poses a barrier to equal and evidence-based management of cancer in older adults. The International Society of Geriatric Oncology (SIOG) formed a panel of experts to develop consensus recommendations on the implications of the pandemic on several aspects of cancer care in this age group including geriatric assessment (GA), surgery, radiotherapy, systemic treatment, palliative care and research. Age and cancer diagnosis are significant predictors of adverse outcomes of the COVID-19 infection. In this setting, GA is particularly valuable to drive decision-making. GA may aid estimating physiologic reserve and adaptive capability, assessing risk-benefits of either providing or temporarily withholding treatments, and determining patient preferences to help inform treatment decisions. In a resource-constrained setting, geriatric screening tools may be administered remotely to identify patients requiring comprehensive GA. Tele-health is also crucial to ensure adequate continuity of care and minimize the risk of infection exposure. In general, therapeutic decisions should favor the most effective and least invasive approach with the lowest risk of adverse outcomes. In selected cases, this might require deferring or omitting surgery, radiotherapy or systemic treatments especially where benefits are marginal and alternative safe therapeutic options are available. Ongoing research is necessary to expand knowledge of the management of cancer in older adults. However, the pandemic presents a significant barrier and efforts should be made to ensure equitable access to clinical trials and prospective data collection to elucidate the outcomes of COVID-19 in this population.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Aged
COVID-19/*complications/epidemiology
Consensus
*Geriatric Assessment
Geriatrics/standards
Humans
Medical Oncology/standards
Neoplasms/*complications/radiotherapy/surgery/*therapy
Palliative Care/methods
Pandemics
Risk Assessment
Societies, Medical
RevDate: 2024-08-29
CmpDate: 2020-12-22
American College of Rheumatology Clinical Guidance for Multisystem Inflammatory Syndrome in Children Associated With SARS-CoV-2 and Hyperinflammation in Pediatric COVID-19: Version 1.
Arthritis & rheumatology (Hoboken, N.J.), 72(11):1791-1805.
OBJECTIVE: To provide guidance on the management of multisystem inflammatory syndrome in children (MIS-C), a condition characterized by fever, inflammation, and multiorgan dysfunction that manifests late in the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and to provide recommendations for children with hyperinflammation during coronavirus disease 2019 (COVID-19), the acute, infectious phase of SARS-CoV-2 infection.
METHODS: A multidisciplinary task force was convened by the American College of Rheumatology (ACR) to provide guidance on the management of MIS-C associated with SARS-CoV-2 and hyperinflammation in COVID-19. The task force was composed of 9 pediatric rheumatologists, 2 adult rheumatologists, 2 pediatric cardiologists, 2 pediatric infectious disease specialists, and 1 pediatric critical care physician. Preliminary statements addressing clinical questions related to MIS-C and hyperinflammation in COVID-19 were developed based on evidence reports. Consensus was built through a modified Delphi process that involved 2 rounds of anonymous voting and 2 webinars. A 9-point scale was used to determine the appropriateness of each statement (median scores of 1-3 for inappropriate, 4-6 for uncertain, and 7-9 for appropriate), and consensus was rated as low, moderate, or high based on dispersion of the votes along the numeric scale. Approved guidance statements were those that were classified as appropriate with moderate or high levels of consensus, as prespecified prior to voting.
RESULTS: The ACR task force approved a total of 128 guidance statements addressing the management of MIS-C and hyperinflammation in pediatric COVID-19. These statements were refined into 40 final clinical guidance statements, accompanied by a flow diagram depicting the diagnostic pathway for MIS-C.
CONCLUSION: Our understanding of SARS-CoV-2-related syndromes in the pediatric population continues to evolve. The guidance provided in this "living document" reflects currently available evidence, coupled with expert opinion, and will be revised as further evidence becomes available.
Additional Links: PMID-32705809
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32705809,
year = {2020},
author = {Henderson, LA and Canna, SW and Friedman, KG and Gorelik, M and Lapidus, SK and Bassiri, H and Behrens, EM and Ferris, A and Kernan, KF and Schulert, GS and Seo, P and F Son, MB and Tremoulet, AH and Yeung, RSM and Mudano, AS and Turner, AS and Karp, DR and Mehta, JJ},
title = {American College of Rheumatology Clinical Guidance for Multisystem Inflammatory Syndrome in Children Associated With SARS-CoV-2 and Hyperinflammation in Pediatric COVID-19: Version 1.},
journal = {Arthritis & rheumatology (Hoboken, N.J.)},
volume = {72},
number = {11},
pages = {1791-1805},
pmid = {32705809},
issn = {2326-5205},
support = {R01 HD098428/HD/NICHD NIH HHS/United States ; },
mesh = {COVID-19/*complications/etiology/therapy ; Consensus ; Humans ; Systemic Inflammatory Response Syndrome/etiology/*therapy ; },
abstract = {OBJECTIVE: To provide guidance on the management of multisystem inflammatory syndrome in children (MIS-C), a condition characterized by fever, inflammation, and multiorgan dysfunction that manifests late in the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and to provide recommendations for children with hyperinflammation during coronavirus disease 2019 (COVID-19), the acute, infectious phase of SARS-CoV-2 infection.
METHODS: A multidisciplinary task force was convened by the American College of Rheumatology (ACR) to provide guidance on the management of MIS-C associated with SARS-CoV-2 and hyperinflammation in COVID-19. The task force was composed of 9 pediatric rheumatologists, 2 adult rheumatologists, 2 pediatric cardiologists, 2 pediatric infectious disease specialists, and 1 pediatric critical care physician. Preliminary statements addressing clinical questions related to MIS-C and hyperinflammation in COVID-19 were developed based on evidence reports. Consensus was built through a modified Delphi process that involved 2 rounds of anonymous voting and 2 webinars. A 9-point scale was used to determine the appropriateness of each statement (median scores of 1-3 for inappropriate, 4-6 for uncertain, and 7-9 for appropriate), and consensus was rated as low, moderate, or high based on dispersion of the votes along the numeric scale. Approved guidance statements were those that were classified as appropriate with moderate or high levels of consensus, as prespecified prior to voting.
RESULTS: The ACR task force approved a total of 128 guidance statements addressing the management of MIS-C and hyperinflammation in pediatric COVID-19. These statements were refined into 40 final clinical guidance statements, accompanied by a flow diagram depicting the diagnostic pathway for MIS-C.
CONCLUSION: Our understanding of SARS-CoV-2-related syndromes in the pediatric population continues to evolve. The guidance provided in this "living document" reflects currently available evidence, coupled with expert opinion, and will be revised as further evidence becomes available.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19/*complications/etiology/therapy
Consensus
Humans
Systemic Inflammatory Response Syndrome/etiology/*therapy
RevDate: 2022-07-16
CmpDate: 2020-12-22
American College of Rheumatology Guidance for the Management of Pediatric Rheumatic Disease During the COVID-19 Pandemic: Version 1.
Arthritis & rheumatology (Hoboken, N.J.), 72(11):1809-1819.
OBJECTIVE: To provide clinical guidance to rheumatology providers who treat children with pediatric rheumatic disease (PRD) in the context of the coronavirus disease 2019 (COVID-19) pandemic.
METHODS: The task force, consisting of 7 pediatric rheumatologists, 2 pediatric infectious disease physicians, 1 adult rheumatologist, and 1 pediatric nurse practitioner, was convened on May 21, 2020. Clinical questions and subsequent guidance statements were drafted based on a review of the queries posed by the patients as well as the families and healthcare providers of children with PRD. An evidence report was generated and disseminated to task force members to assist with 3 rounds of asynchronous, anonymous voting by email using a modified Delphi approach. Voting was completed using a 9-point numeric scoring system with predefined levels of agreement (categorized as disagreement, uncertainty, or agreement, with median scores of 1-3, 4-6, and 7-9, respectively) and consensus (categorized as low, moderate, or high). To be approved as a guidance statement, median vote ratings were required to fall into the highest tertile for agreement, with either moderate or high levels of consensus.
RESULTS: The task force drafted 33 guidance statements, which were voted upon during the second and third rounds of voting. Of these 33 statements, all received median vote ratings within the highest tertile of agreement and were associated with either moderate consensus (n = 6) or high consensus (n = 27). Statements with similar recommendations were combined, resulting in 27 final guidance statements.
CONCLUSION: These guidance statements have been generated based on review of the available literature, indicating that children with PRD do not appear to be at increased risk for susceptibility to SARS-CoV-2 infection. This guidance is presented as a "living document," recognizing that the literature on COVID-19 is rapidly evolving, with future updates anticipated.
Additional Links: PMID-32705780
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32705780,
year = {2020},
author = {Wahezi, DM and Lo, MS and Rubinstein, TB and Ringold, S and Ardoin, SP and Downes, KJ and Jones, KB and Laxer, RM and Pellet Madan, R and Mudano, AS and Turner, AS and Karp, DR and Mehta, JJ},
title = {American College of Rheumatology Guidance for the Management of Pediatric Rheumatic Disease During the COVID-19 Pandemic: Version 1.},
journal = {Arthritis & rheumatology (Hoboken, N.J.)},
volume = {72},
number = {11},
pages = {1809-1819},
pmid = {32705780},
issn = {2326-5205},
support = {K23 HD091365/HD/NICHD NIH HHS/United States ; },
mesh = {Antirheumatic Agents/*therapeutic use ; *COVID-19 ; Child ; Consensus ; Humans ; Pandemics ; Rheumatic Diseases/*drug therapy ; },
abstract = {OBJECTIVE: To provide clinical guidance to rheumatology providers who treat children with pediatric rheumatic disease (PRD) in the context of the coronavirus disease 2019 (COVID-19) pandemic.
METHODS: The task force, consisting of 7 pediatric rheumatologists, 2 pediatric infectious disease physicians, 1 adult rheumatologist, and 1 pediatric nurse practitioner, was convened on May 21, 2020. Clinical questions and subsequent guidance statements were drafted based on a review of the queries posed by the patients as well as the families and healthcare providers of children with PRD. An evidence report was generated and disseminated to task force members to assist with 3 rounds of asynchronous, anonymous voting by email using a modified Delphi approach. Voting was completed using a 9-point numeric scoring system with predefined levels of agreement (categorized as disagreement, uncertainty, or agreement, with median scores of 1-3, 4-6, and 7-9, respectively) and consensus (categorized as low, moderate, or high). To be approved as a guidance statement, median vote ratings were required to fall into the highest tertile for agreement, with either moderate or high levels of consensus.
RESULTS: The task force drafted 33 guidance statements, which were voted upon during the second and third rounds of voting. Of these 33 statements, all received median vote ratings within the highest tertile of agreement and were associated with either moderate consensus (n = 6) or high consensus (n = 27). Statements with similar recommendations were combined, resulting in 27 final guidance statements.
CONCLUSION: These guidance statements have been generated based on review of the available literature, indicating that children with PRD do not appear to be at increased risk for susceptibility to SARS-CoV-2 infection. This guidance is presented as a "living document," recognizing that the literature on COVID-19 is rapidly evolving, with future updates anticipated.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Antirheumatic Agents/*therapeutic use
*COVID-19
Child
Consensus
Humans
Pandemics
Rheumatic Diseases/*drug therapy
RevDate: 2026-01-15
CmpDate: 2020-07-29
COVID-19 guidance for triage of operations for thoracic malignancies: A consensus statement from Thoracic Surgery Outcomes Research Network.
The Journal of thoracic and cardiovascular surgery, 160(2):601-605.
The extraordinary demands of managing the COVID-19 pandemic has disrupted the world's ability to care for patients with thoracic malignancies. As a hospital's COVID-19 population increases and hospital resources are depleted, the ability to provide surgical care is progressively restricted, forcing surgeons to prioritize among their cancer populations. Representatives from multiple cancer, surgical, and research organizations have come together to provide a guide for triaging patients with thoracic malignancies as the impact of COVID-19 evolves as each hospital.
Additional Links: PMID-32689703
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32689703,
year = {2020},
author = {, and Antonoff, M and Backhus, L and Boffa, DJ and Broderick, SR and Brown, LM and Carrott, P and Clark, JM and Cooke, D and David, E and Facktor, M and Farjah, F and Grogan, E and Isbell, J and Jones, DR and Kidane, B and Kim, AW and Keshavjee, S and Krantz, S and Lui, N and Martin, L and Meguid, RA and Meyerson, SL and Mullett, T and Nelson, H and Odell, DD and Phillips, JD and Puri, V and Rusch, V and Shulman, L and Varghese, TK and Wakeam, E and Wood, DE},
title = {COVID-19 guidance for triage of operations for thoracic malignancies: A consensus statement from Thoracic Surgery Outcomes Research Network.},
journal = {The Journal of thoracic and cardiovascular surgery},
volume = {160},
number = {2},
pages = {601-605},
pmid = {32689703},
issn = {1097-685X},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Betacoronavirus/*pathogenicity ; COVID-19 ; Clinical Decision-Making ; Coronavirus Infections/epidemiology/*therapy/transmission/virology ; Delivery of Health Care, Integrated/*organization & administration ; Health Services Needs and Demand/organization & administration ; Host Microbial Interactions ; Humans ; Needs Assessment/organization & administration ; Occupational Health ; Pandemics ; Patient Safety ; Patient Selection ; Pneumonia, Viral/epidemiology/*therapy/transmission/virology ; Risk Assessment ; Risk Factors ; SARS-CoV-2 ; Thoracic Neoplasms/epidemiology/*surgery ; *Thoracic Surgical Procedures/adverse effects ; Time-to-Treatment ; Triage/*organization & administration ; },
abstract = {The extraordinary demands of managing the COVID-19 pandemic has disrupted the world's ability to care for patients with thoracic malignancies. As a hospital's COVID-19 population increases and hospital resources are depleted, the ability to provide surgical care is progressively restricted, forcing surgeons to prioritize among their cancer populations. Representatives from multiple cancer, surgical, and research organizations have come together to provide a guide for triaging patients with thoracic malignancies as the impact of COVID-19 evolves as each hospital.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus/*pathogenicity
COVID-19
Clinical Decision-Making
Coronavirus Infections/epidemiology/*therapy/transmission/virology
Delivery of Health Care, Integrated/*organization & administration
Health Services Needs and Demand/organization & administration
Host Microbial Interactions
Humans
Needs Assessment/organization & administration
Occupational Health
Pandemics
Patient Safety
Patient Selection
Pneumonia, Viral/epidemiology/*therapy/transmission/virology
Risk Assessment
Risk Factors
SARS-CoV-2
Thoracic Neoplasms/epidemiology/*surgery
*Thoracic Surgical Procedures/adverse effects
Time-to-Treatment
Triage/*organization & administration
RevDate: 2024-03-29
CmpDate: 2020-09-03
Inter Organizational Practice Committee Recommendations/Guidance for Teleneuropsychology in Response to the COVID-19 Pandemic†.
Archives of clinical neuropsychology : the official journal of the National Academy of Neuropsychologists, 35(6):647-659.
OBJECTIVE: The Inter Organizational Practice Committee convened a workgroup to provide rapid guidance about teleneuropsychology (TeleNP) in response to the COVID-19 pandemic.
METHOD: A collaborative panel of experts from major professional organizations developed provisional guidance for neuropsychological practice during the pandemic. The stakeholders included the American Academy of Clinical Neuropsychology/American Board of Clinical Neuropsychology, the National Academy of Neuropsychology, Division 40 of the American Psychological Association, the American Board of Professional Neuropsychology, and the American Psychological Association Services, Inc. The group reviewed literature; collated federal, regional, and state regulations and information from insurers; and surveyed practitioners to identify best practices.
RESULTS: Literature indicates that TeleNP may offer reliable and valid assessments, but clinicians need to consider limitations, develop new informed consent procedures, report modifications of standard procedures, and state limitations to diagnostic conclusions and recommendations. Specific limitations affect TeleNP assessments of older adults, younger children, individuals with limited access to technology, and individuals with other individual, cultural, and/or linguistic differences. TeleNP may be contraindicated or infeasible given specific patient characteristics, circumstances, and referral questions. Considerations for billing TeleNP services are offered with reservations that clinicians must verify procedures independently. Guidance about technical issues and "tips" for TeleNP procedures are provided.
CONCLUSION: This document provides provisional guidance with links to resources and established guidelines for telepsychology. Specific recommendations extend these practices to TeleNP. These recommendations may be revised as circumstances evolve, with updates posted continuously at IOPC.online.
Additional Links: PMID-32666093
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32666093,
year = {2020},
author = {Bilder, RM and Postal, KS and Barisa, M and Aase, DM and Cullum, CM and Gillaspy, SR and Harder, L and Kanter, G and Lanca, M and Lechuga, DM and Morgan, JM and Most, R and Puente, AE and Salinas, CM and Woodhouse, J},
title = {Inter Organizational Practice Committee Recommendations/Guidance for Teleneuropsychology in Response to the COVID-19 Pandemic†.},
journal = {Archives of clinical neuropsychology : the official journal of the National Academy of Neuropsychologists},
volume = {35},
number = {6},
pages = {647-659},
pmid = {32666093},
issn = {1873-5843},
support = {R01 MH118514/MH/NIMH NIH HHS/United States ; },
mesh = {Betacoronavirus ; COVID-19 ; Coronavirus Infections/epidemiology ; Humans ; Informed Consent ; Insurance, Health ; Licensure ; Medicaid ; Medicare ; Neuropsychological Tests ; Neuropsychology/*methods ; Pandemics ; Pneumonia, Viral/epidemiology ; Reimbursement Mechanisms ; SARS-CoV-2 ; Societies, Scientific ; Surveys and Questionnaires ; Telemedicine/*methods ; United States/epidemiology ; *Videoconferencing ; },
abstract = {OBJECTIVE: The Inter Organizational Practice Committee convened a workgroup to provide rapid guidance about teleneuropsychology (TeleNP) in response to the COVID-19 pandemic.
METHOD: A collaborative panel of experts from major professional organizations developed provisional guidance for neuropsychological practice during the pandemic. The stakeholders included the American Academy of Clinical Neuropsychology/American Board of Clinical Neuropsychology, the National Academy of Neuropsychology, Division 40 of the American Psychological Association, the American Board of Professional Neuropsychology, and the American Psychological Association Services, Inc. The group reviewed literature; collated federal, regional, and state regulations and information from insurers; and surveyed practitioners to identify best practices.
RESULTS: Literature indicates that TeleNP may offer reliable and valid assessments, but clinicians need to consider limitations, develop new informed consent procedures, report modifications of standard procedures, and state limitations to diagnostic conclusions and recommendations. Specific limitations affect TeleNP assessments of older adults, younger children, individuals with limited access to technology, and individuals with other individual, cultural, and/or linguistic differences. TeleNP may be contraindicated or infeasible given specific patient characteristics, circumstances, and referral questions. Considerations for billing TeleNP services are offered with reservations that clinicians must verify procedures independently. Guidance about technical issues and "tips" for TeleNP procedures are provided.
CONCLUSION: This document provides provisional guidance with links to resources and established guidelines for telepsychology. Specific recommendations extend these practices to TeleNP. These recommendations may be revised as circumstances evolve, with updates posted continuously at IOPC.online.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus
COVID-19
Coronavirus Infections/epidemiology
Humans
Informed Consent
Insurance, Health
Licensure
Medicaid
Medicare
Neuropsychological Tests
Neuropsychology/*methods
Pandemics
Pneumonia, Viral/epidemiology
Reimbursement Mechanisms
SARS-CoV-2
Societies, Scientific
Surveys and Questionnaires
Telemedicine/*methods
United States/epidemiology
*Videoconferencing
RevDate: 2023-11-12
CmpDate: 2020-10-30
COVID-19 PICU guidelines: for high- and limited-resource settings.
Pediatric research, 88(5):705-716.
BACKGROUND: Fewer children than adults have been affected by the COVID-19 pandemic, and the clinical manifestations are distinct from those of adults. Some children particularly those with acute or chronic co-morbidities are likely to develop critical illness. Recently, a multisystem inflammatory syndrome (MIS-C) has been described in children with some of these patients requiring care in the pediatric ICU.
METHODS: An international collaboration was formed to review the available evidence and develop evidence-based guidelines for the care of critically ill children with SARS-CoV-2 infection. Where the evidence was lacking, those gaps were replaced with consensus-based guidelines.
RESULTS: This process has generated 44 recommendations related to pediatric COVID-19 patients presenting with respiratory distress or failure, sepsis or septic shock, cardiopulmonary arrest, MIS-C, those requiring adjuvant therapies, or ECMO. Evidence to explain the milder disease patterns in children and the potential to use repurposed anti-viral drugs, anti-inflammatory or anti-thrombotic therapies are also described.
CONCLUSION: Brief summaries of pediatric SARS-CoV-2 infection in different regions of the world are included since few registries are capturing this data globally. These guidelines seek to harmonize the standards and strategies for intensive care that critically ill children with COVID-19 receive across the world.
IMPACT: At the time of publication, this is the latest evidence for managing critically ill children infected with SARS-CoV-2. Referring to these guidelines can decrease the morbidity and potentially the mortality of children effected by COVID-19 and its sequalae. These guidelines can be adapted to both high- and limited-resource settings.
Additional Links: PMID-32634818
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32634818,
year = {2020},
author = {Kache, S and Chisti, MJ and Gumbo, F and Mupere, E and Zhi, X and Nallasamy, K and Nakagawa, S and Lee, JH and Di Nardo, M and de la Oliva, P and Katyal, C and Anand, KJS and de Souza, DC and Lanziotti, VS and Carcillo, J},
title = {COVID-19 PICU guidelines: for high- and limited-resource settings.},
journal = {Pediatric research},
volume = {88},
number = {5},
pages = {705-716},
pmid = {32634818},
issn = {1530-0447},
support = {R01 GM108618/GM/NIGMS NIH HHS/United States ; },
mesh = {Adolescent ; Africa/epidemiology ; Americas/epidemiology ; Antiviral Agents/therapeutic use ; Asia/epidemiology ; *Betacoronavirus ; COVID-19 ; Cardiopulmonary Resuscitation/methods/standards ; Child ; Child, Preschool ; Combined Modality Therapy ; Comorbidity ; Coronavirus Infections/complications/drug therapy/epidemiology/*therapy ; Critical Care/methods/*standards ; Cross Infection/prevention & control ; Europe/epidemiology ; Extracorporeal Membrane Oxygenation/standards ; Female ; Humans ; Infant ; Infant, Newborn ; Infection Control/methods/standards ; Intensive Care Units, Pediatric/*standards ; Male ; *Pandemics ; Pneumonia, Viral/complications/epidemiology/*therapy ; Respiration, Artificial/standards ; Respiratory Distress Syndrome/etiology/therapy ; SARS-CoV-2 ; Shock/etiology/therapy ; Systemic Inflammatory Response Syndrome/epidemiology/therapy ; COVID-19 Drug Treatment ; },
abstract = {BACKGROUND: Fewer children than adults have been affected by the COVID-19 pandemic, and the clinical manifestations are distinct from those of adults. Some children particularly those with acute or chronic co-morbidities are likely to develop critical illness. Recently, a multisystem inflammatory syndrome (MIS-C) has been described in children with some of these patients requiring care in the pediatric ICU.
METHODS: An international collaboration was formed to review the available evidence and develop evidence-based guidelines for the care of critically ill children with SARS-CoV-2 infection. Where the evidence was lacking, those gaps were replaced with consensus-based guidelines.
RESULTS: This process has generated 44 recommendations related to pediatric COVID-19 patients presenting with respiratory distress or failure, sepsis or septic shock, cardiopulmonary arrest, MIS-C, those requiring adjuvant therapies, or ECMO. Evidence to explain the milder disease patterns in children and the potential to use repurposed anti-viral drugs, anti-inflammatory or anti-thrombotic therapies are also described.
CONCLUSION: Brief summaries of pediatric SARS-CoV-2 infection in different regions of the world are included since few registries are capturing this data globally. These guidelines seek to harmonize the standards and strategies for intensive care that critically ill children with COVID-19 receive across the world.
IMPACT: At the time of publication, this is the latest evidence for managing critically ill children infected with SARS-CoV-2. Referring to these guidelines can decrease the morbidity and potentially the mortality of children effected by COVID-19 and its sequalae. These guidelines can be adapted to both high- and limited-resource settings.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Adolescent
Africa/epidemiology
Americas/epidemiology
Antiviral Agents/therapeutic use
Asia/epidemiology
*Betacoronavirus
COVID-19
Cardiopulmonary Resuscitation/methods/standards
Child
Child, Preschool
Combined Modality Therapy
Comorbidity
Coronavirus Infections/complications/drug therapy/epidemiology/*therapy
Critical Care/methods/*standards
Cross Infection/prevention & control
Europe/epidemiology
Extracorporeal Membrane Oxygenation/standards
Female
Humans
Infant
Infant, Newborn
Infection Control/methods/standards
Intensive Care Units, Pediatric/*standards
Male
*Pandemics
Pneumonia, Viral/complications/epidemiology/*therapy
Respiration, Artificial/standards
Respiratory Distress Syndrome/etiology/therapy
SARS-CoV-2
Shock/etiology/therapy
Systemic Inflammatory Response Syndrome/epidemiology/therapy
COVID-19 Drug Treatment
RevDate: 2020-12-23
CmpDate: 2020-08-24
A Meta-analysis of the Relationship Between Renin-Angiotensin-Aldosterone System Inhibitors and COVID-19.
The American journal of cardiology, 130:159-161.
Additional Links: PMID-32624189
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32624189,
year = {2020},
author = {Usman, MS and Siddiqi, TJ and Khan, MS and Ahmed, A and Ali, SS and Michos, ED and Hall, ME and Krasuski, RA and Greene, SJ and Butler, J and Alkhouli, M},
title = {A Meta-analysis of the Relationship Between Renin-Angiotensin-Aldosterone System Inhibitors and COVID-19.},
journal = {The American journal of cardiology},
volume = {130},
number = {},
pages = {159-161},
pmid = {32624189},
issn = {1879-1913},
support = {K08 DK099415/DK/NIDDK NIH HHS/United States ; },
mesh = {Angiotensin Receptor Antagonists/*adverse effects ; Angiotensin-Converting Enzyme 2 ; Angiotensin-Converting Enzyme Inhibitors/*adverse effects ; Betacoronavirus ; COVID-19 ; Coronavirus Infections/*diagnosis/mortality ; Humans ; Pandemics ; Peptidyl-Dipeptidase A ; Pneumonia, Viral/*diagnosis/mortality ; Renin-Angiotensin System ; Risk Factors ; SARS-CoV-2 ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Angiotensin Receptor Antagonists/*adverse effects
Angiotensin-Converting Enzyme 2
Angiotensin-Converting Enzyme Inhibitors/*adverse effects
Betacoronavirus
COVID-19
Coronavirus Infections/*diagnosis/mortality
Humans
Pandemics
Peptidyl-Dipeptidase A
Pneumonia, Viral/*diagnosis/mortality
Renin-Angiotensin System
Risk Factors
SARS-CoV-2
RevDate: 2021-09-19
CmpDate: 2020-09-08
Spotlight: COVID-19 PPE and Endoscopy.
Gastroenterology, 159(2):759.
Additional Links: PMID-32590006
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32590006,
year = {2020},
author = {Siddique, SM and Sultan, S and Lim, JK and Altayar, O and Davitkov, P and Feuerstein, JD and Falck-Ytter, Y and El-Serag, HB and , },
title = {Spotlight: COVID-19 PPE and Endoscopy.},
journal = {Gastroenterology},
volume = {159},
number = {2},
pages = {759},
doi = {10.1053/j.gastro.2020.06.047},
pmid = {32590006},
issn = {1528-0012},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Betacoronavirus ; COVID-19 ; Coronavirus Infections/*prevention & control/transmission ; Disease Transmission, Infectious/*prevention & control ; Endoscopy/adverse effects/*standards ; Humans ; Pandemics/*prevention & control ; Personal Protective Equipment/*standards ; Pneumonia, Viral/*prevention & control/transmission ; *Practice Guidelines as Topic ; SARS-CoV-2 ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus
COVID-19
Coronavirus Infections/*prevention & control/transmission
Disease Transmission, Infectious/*prevention & control
Endoscopy/adverse effects/*standards
Humans
Pandemics/*prevention & control
Personal Protective Equipment/*standards
Pneumonia, Viral/*prevention & control/transmission
*Practice Guidelines as Topic
SARS-CoV-2
RevDate: 2022-10-22
CmpDate: 2020-09-09
HRS/EHRA/APHRS/LAHRS/ACC/AHA worldwide practice update for telehealth and arrhythmia monitoring during and after a pandemic.
Heart rhythm, 17(9):e255-e268.
Additional Links: PMID-32535140
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32535140,
year = {2020},
author = {Varma, N and Marrouche, NF and Aguinaga, L and Albert, CM and Arbelo, E and Choi, JI and Chung, MK and Conte, G and Dagher, L and Epstein, LM and Ghanbari, H and Han, JK and Heidbuchel, H and Huang, H and Lakkireddy, DR and Ngarmukos, T and Russo, AM and Saad, EB and Saenz Morales, LC and Sandau, KE and Sridhar, ARM and Stecker, EC and Varosy, PD},
title = {HRS/EHRA/APHRS/LAHRS/ACC/AHA worldwide practice update for telehealth and arrhythmia monitoring during and after a pandemic.},
journal = {Heart rhythm},
volume = {17},
number = {9},
pages = {e255-e268},
pmid = {32535140},
issn = {1556-3871},
support = {K23 HL135397/HL/NHLBI NIH HHS/United States ; R21 DC020005/DC/NIDCD NIH HHS/United States ; },
mesh = {Arrhythmias, Cardiac/*diagnosis/etiology/therapy ; *Betacoronavirus ; COVID-19 ; Coronavirus Infections/*complications/epidemiology/therapy ; *Electrocardiography ; Humans ; Pandemics ; Pneumonia, Viral/*complications/epidemiology/therapy ; SARS-CoV-2 ; Telemedicine/*organization & administration ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Arrhythmias, Cardiac/*diagnosis/etiology/therapy
*Betacoronavirus
COVID-19
Coronavirus Infections/*complications/epidemiology/therapy
*Electrocardiography
Humans
Pandemics
Pneumonia, Viral/*complications/epidemiology/therapy
SARS-CoV-2
Telemedicine/*organization & administration
RevDate: 2024-02-11
CmpDate: 2020-09-21
HRS/EHRA/APHRS/LAHRS/ACC/AHA Worldwide Practice Update for Telehealth and Arrhythmia Monitoring During and After a Pandemic.
Journal of the American College of Cardiology, 76(11):1363-1374.
Additional Links: PMID-32534936
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32534936,
year = {2020},
author = {Varma, N and Marrouche, NF and Aguinaga, L and Albert, CM and Arbelo, E and Choi, JI and Chung, MK and Conte, G and Dagher, L and Epstein, LM and Ghanbari, H and Han, JK and Heidbuchel, H and Huang, H and Lakkireddy, DR and Ngarmukos, T and Russo, AM and Saad, EB and Saenz Morales, LC and Sandau, KE and Sridhar, ARM and Stecker, EC and Varosy, PD},
title = {HRS/EHRA/APHRS/LAHRS/ACC/AHA Worldwide Practice Update for Telehealth and Arrhythmia Monitoring During and After a Pandemic.},
journal = {Journal of the American College of Cardiology},
volume = {76},
number = {11},
pages = {1363-1374},
pmid = {32534936},
issn = {1558-3597},
support = {K23 HL135397/HL/NHLBI NIH HHS/United States ; R21 DC020005/DC/NIDCD NIH HHS/United States ; },
mesh = {*Arrhythmias, Cardiac/diagnosis/epidemiology ; Betacoronavirus ; COVID-19 ; Communicable Disease Control/organization & administration ; *Coronavirus Infections/epidemiology/prevention & control ; Electrophysiologic Techniques, Cardiac/methods/trends ; Humans ; International Cooperation ; *Pandemics/prevention & control ; *Pneumonia, Viral/epidemiology/prevention & control ; Remote Consultation/*methods ; SARS-CoV-2 ; Telemedicine/*organization & administration ; *Telemetry/instrumentation/methods ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Arrhythmias, Cardiac/diagnosis/epidemiology
Betacoronavirus
COVID-19
Communicable Disease Control/organization & administration
*Coronavirus Infections/epidemiology/prevention & control
Electrophysiologic Techniques, Cardiac/methods/trends
Humans
International Cooperation
*Pandemics/prevention & control
*Pneumonia, Viral/epidemiology/prevention & control
Remote Consultation/*methods
SARS-CoV-2
Telemedicine/*organization & administration
*Telemetry/instrumentation/methods
RevDate: 2024-07-29
CmpDate: 2020-07-16
Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.
Journal of the American College of Radiology : JACR, 17(7):845-854.
BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small-cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small-cell lung cancer.
CONCLUSIONS: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.
Additional Links: PMID-32485147
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32485147,
year = {2020},
author = {Mazzone, PJ and Gould, MK and Arenberg, DA and Chen, AC and Choi, HK and Detterbeck, FC and Farjah, F and Fong, KM and Iaccarino, JM and Janes, SM and Kanne, JP and Kazerooni, EA and MacMahon, H and Naidich, DP and Powell, CA and Raoof, S and Rivera, MP and Tanner, NT and Tanoue, LK and Tremblay, A and Vachani, A and White, CS and Wiener, RS and Silvestri, GA},
title = {Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.},
journal = {Journal of the American College of Radiology : JACR},
volume = {17},
number = {7},
pages = {845-854},
pmid = {32485147},
issn = {1558-349X},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Betacoronavirus ; COVID-19 ; Consensus ; Coronavirus Infections/*prevention & control/transmission ; Diagnostic Imaging/*standards ; Early Detection of Cancer ; Humans ; Lung Neoplasms/*diagnostic imaging ; Multiple Pulmonary Nodules/*diagnostic imaging ; Pandemics/*prevention & control ; Pneumonia, Viral/*prevention & control/transmission ; SARS-CoV-2 ; Solitary Pulmonary Nodule/*diagnostic imaging ; },
abstract = {BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small-cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small-cell lung cancer.
CONCLUSIONS: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus
COVID-19
Consensus
Coronavirus Infections/*prevention & control/transmission
Diagnostic Imaging/*standards
Early Detection of Cancer
Humans
Lung Neoplasms/*diagnostic imaging
Multiple Pulmonary Nodules/*diagnostic imaging
Pandemics/*prevention & control
Pneumonia, Viral/*prevention & control/transmission
SARS-CoV-2
Solitary Pulmonary Nodule/*diagnostic imaging
RevDate: 2021-09-19
CmpDate: 2020-06-05
Advancing the NIH Strategic Plan and Health Care Needs During the COVID-19 Pandemic.
Nursing outlook, 68(3):371-373.
Additional Links: PMID-32482242
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32482242,
year = {2020},
author = {Redeker, NS},
title = {Advancing the NIH Strategic Plan and Health Care Needs During the COVID-19 Pandemic.},
journal = {Nursing outlook},
volume = {68},
number = {3},
pages = {371-373},
pmid = {32482242},
issn = {1528-3968},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {COVID-19 ; Coronavirus Infections/*epidemiology/nursing ; Health Services Needs and Demand ; Humans ; National Institutes of Health (U.S.)/economics/*organization & administration ; Nurse's Role ; Nursing Research ; Organizational Objectives ; *Pandemics ; Pneumonia, Viral/*epidemiology/nursing ; Public Health/*legislation & jurisprudence ; United States/epidemiology ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19
Coronavirus Infections/*epidemiology/nursing
Health Services Needs and Demand
Humans
National Institutes of Health (U.S.)/economics/*organization & administration
Nurse's Role
Nursing Research
Organizational Objectives
*Pandemics
Pneumonia, Viral/*epidemiology/nursing
Public Health/*legislation & jurisprudence
United States/epidemiology
RevDate: 2025-12-11
CmpDate: 2020-07-08
North American Neuroendocrine Tumor Society Guide for Neuroendocrine Tumor Patient Health Care Providers During COVID-19.
Pancreas, 49(6):723-728.
Additional Links: PMID-32433264
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32433264,
year = {2020},
author = {Bergsland, EK and Halperin, DM and Dillon, JS and Dasari, NA and Kunz, PL and Soares, HP and Pryma, D and Bodei, L and Hope, T and Soulen, MC and Mailman, J and Howe, JR},
title = {North American Neuroendocrine Tumor Society Guide for Neuroendocrine Tumor Patient Health Care Providers During COVID-19.},
journal = {Pancreas},
volume = {49},
number = {6},
pages = {723-728},
pmid = {32433264},
issn = {1536-4828},
support = {P50 CA174521/CA/NCI NIH HHS/United States ; UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {*Betacoronavirus ; COVID-19 ; Coronavirus Infections/complications/*prevention & control/transmission ; Health Personnel ; Humans ; Infectious Disease Transmission, Patient-to-Professional/*prevention & control ; Infectious Disease Transmission, Professional-to-Patient/*prevention & control ; Neuroendocrine Tumors/complications/immunology/*therapy ; North America ; Occupational Diseases/*prevention & control ; Occupational Exposure/*prevention & control ; Pandemics/*prevention & control ; Pneumonia, Viral/complications/*prevention & control/transmission ; SARS-CoV-2 ; Societies, Medical ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Betacoronavirus
COVID-19
Coronavirus Infections/complications/*prevention & control/transmission
Health Personnel
Humans
Infectious Disease Transmission, Patient-to-Professional/*prevention & control
Infectious Disease Transmission, Professional-to-Patient/*prevention & control
Neuroendocrine Tumors/complications/immunology/*therapy
North America
Occupational Diseases/*prevention & control
Occupational Exposure/*prevention & control
Pandemics/*prevention & control
Pneumonia, Viral/complications/*prevention & control/transmission
SARS-CoV-2
Societies, Medical
RevDate: 2020-12-18
CmpDate: 2020-07-29
Temporary Emergency Guidance to STEMI Systems of Care During the COVID-19 Pandemic: AHA's Mission: Lifeline.
Circulation, 142(3):199-202.
Additional Links: PMID-32363905
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32363905,
year = {2020},
author = {, },
title = {Temporary Emergency Guidance to STEMI Systems of Care During the COVID-19 Pandemic: AHA's Mission: Lifeline.},
journal = {Circulation},
volume = {142},
number = {3},
pages = {199-202},
pmid = {32363905},
issn = {1524-4539},
support = {K23 NR017896/NR/NINR NIH HHS/United States ; },
mesh = {American Heart Association ; Betacoronavirus/*pathogenicity ; COVID-19 ; Cardiology/*standards ; Cardiology Service, Hospital/*standards ; Consensus ; Coronavirus Infections/diagnosis/epidemiology/*therapy/virology ; Delivery of Health Care/*standards ; Emergency Service, Hospital/*standards ; Host Microbial Interactions ; Humans ; Occupational Health/standards ; Pandemics ; Patient Safety/standards ; Pneumonia, Viral/diagnosis/epidemiology/*therapy/virology ; Prognosis ; Risk Assessment ; Risk Factors ; SARS-CoV-2 ; ST Elevation Myocardial Infarction/diagnosis/epidemiology/*therapy ; United States/epidemiology ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
American Heart Association
Betacoronavirus/*pathogenicity
COVID-19
Cardiology/*standards
Cardiology Service, Hospital/*standards
Consensus
Coronavirus Infections/diagnosis/epidemiology/*therapy/virology
Delivery of Health Care/*standards
Emergency Service, Hospital/*standards
Host Microbial Interactions
Humans
Occupational Health/standards
Pandemics
Patient Safety/standards
Pneumonia, Viral/diagnosis/epidemiology/*therapy/virology
Prognosis
Risk Assessment
Risk Factors
SARS-CoV-2
ST Elevation Myocardial Infarction/diagnosis/epidemiology/*therapy
United States/epidemiology
RevDate: 2024-07-28
CmpDate: 2020-04-30
Society for Cardiovascular Magnetic Resonance (SCMR) guidance for the practice of cardiovascular magnetic resonance during the COVID-19 pandemic.
Journal of cardiovascular magnetic resonance : official journal of the Society for Cardiovascular Magnetic Resonance, 22(1):26.
The aim of this document is to provide general guidance and specific recommendations on the practice of cardiovascular magnetic resonance (CMR) in the era of the COVID-19 pandemic. There are two major considerations. First, continued urgent and semi-urgent care for the patients who have no known active COVID-19 should be provided in a safe manner for both patients and staff. Second, when necessary, CMR on patients with confirmed or suspected active COVID-19 should focus on the specific clinical question with an emphasis on myocardial function and tissue characterization while optimizing patient and staff safety.
Additional Links: PMID-32340614
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32340614,
year = {2020},
author = {Han, Y and Chen, T and Bryant, J and Bucciarelli-Ducci, C and Dyke, C and Elliott, MD and Ferrari, VA and Friedrich, MG and Lawton, C and Manning, WJ and Ordovas, K and Plein, S and Powell, AJ and Raman, SV and Carr, J},
title = {Society for Cardiovascular Magnetic Resonance (SCMR) guidance for the practice of cardiovascular magnetic resonance during the COVID-19 pandemic.},
journal = {Journal of cardiovascular magnetic resonance : official journal of the Society for Cardiovascular Magnetic Resonance},
volume = {22},
number = {1},
pages = {26},
pmid = {32340614},
issn = {1532-429X},
support = {R01 HL148103/HL/NHLBI NIH HHS/United States ; },
mesh = {Betacoronavirus ; COVID-19 ; Cardiovascular Diseases/*diagnostic imaging ; *Coronavirus Infections/complications/epidemiology ; Humans ; Infection Control/methods ; Infectious Disease Transmission, Patient-to-Professional/prevention & control ; Magnetic Resonance Imaging/*standards ; *Pandemics ; *Patient Safety ; *Pneumonia, Viral/complications/epidemiology ; SARS-CoV-2 ; },
abstract = {The aim of this document is to provide general guidance and specific recommendations on the practice of cardiovascular magnetic resonance (CMR) in the era of the COVID-19 pandemic. There are two major considerations. First, continued urgent and semi-urgent care for the patients who have no known active COVID-19 should be provided in a safe manner for both patients and staff. Second, when necessary, CMR on patients with confirmed or suspected active COVID-19 should focus on the specific clinical question with an emphasis on myocardial function and tissue characterization while optimizing patient and staff safety.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus
COVID-19
Cardiovascular Diseases/*diagnostic imaging
*Coronavirus Infections/complications/epidemiology
Humans
Infection Control/methods
Infectious Disease Transmission, Patient-to-Professional/prevention & control
Magnetic Resonance Imaging/*standards
*Pandemics
*Patient Safety
*Pneumonia, Viral/complications/epidemiology
SARS-CoV-2
RevDate: 2025-05-30
CmpDate: 2020-07-20
Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.
Chest, 158(1):406-415.
BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small cell lung cancer.
CONCLUSIONS: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.
Additional Links: PMID-32335067
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32335067,
year = {2020},
author = {Mazzone, PJ and Gould, MK and Arenberg, DA and Chen, AC and Choi, HK and Detterbeck, FC and Farjah, F and Fong, KM and Iaccarino, JM and Janes, SM and Kanne, JP and Kazerooni, EA and MacMahon, H and Naidich, DP and Powell, CA and Raoof, S and Rivera, MP and Tanner, NT and Tanoue, LK and Tremblay, A and Vachani, A and White, CS and Wiener, RS and Silvestri, GA},
title = {Management of Lung Nodules and Lung Cancer Screening During the COVID-19 Pandemic: CHEST Expert Panel Report.},
journal = {Chest},
volume = {158},
number = {1},
pages = {406-415},
pmid = {32335067},
issn = {1931-3543},
support = {28706/CRUK_/Cancer Research UK/United Kingdom ; UL1 TR001863/TR/NCATS NIH HHS/United States ; UM1 CA221939/CA/NCI NIH HHS/United States ; },
mesh = {Betacoronavirus/isolation & purification ; COVID-19 ; Carcinoma, Non-Small-Cell Lung/*diagnosis ; Consensus ; *Coronavirus Infections/diagnosis/epidemiology ; Early Detection of Cancer/methods/standards ; Humans ; *Lung Neoplasms/diagnostic imaging/pathology ; Multiple Pulmonary Nodules/*diagnosis ; Neoplasm Staging ; *Pandemics/prevention & control ; *Pneumonia, Viral/diagnosis/epidemiology ; Radiography, Thoracic/*methods ; Resource Allocation ; Risk Assessment/methods ; SARS-CoV-2 ; },
abstract = {BACKGROUND: The risks from potential exposure to coronavirus disease 2019 (COVID-19), and resource reallocation that has occurred to combat the pandemic, have altered the balance of benefits and harms that informed current (pre-COVID-19) guideline recommendations for lung cancer screening and lung nodule evaluation. Consensus statements were developed to guide clinicians managing lung cancer screening programs and patients with lung nodules during the COVID-19 pandemic.
METHODS: An expert panel of 24 members, including pulmonologists (n = 17), thoracic radiologists (n = 5), and thoracic surgeons (n = 2), was formed. The panel was provided with an overview of current evidence, summarized by recent guidelines related to lung cancer screening and lung nodule evaluation. The panel was convened by video teleconference to discuss and then vote on statements related to 12 common clinical scenarios. A predefined threshold of 70% of panel members voting agree or strongly agree was used to determine if there was a consensus for each statement. Items that may influence decisions were listed as notes to be considered for each scenario.
RESULTS: Twelve statements related to baseline and annual lung cancer screening (n = 2), surveillance of a previously detected lung nodule (n = 5), evaluation of intermediate and high-risk lung nodules (n = 4), and management of clinical stage I non-small cell lung cancer (n = 1) were developed and modified. All 12 statements were confirmed as consensus statements according to the voting results. The consensus statements provide guidance about situations in which it was believed to be appropriate to delay screening, defer surveillance imaging of lung nodules, and minimize nonurgent interventions during the evaluation of lung nodules and stage I non-small cell lung cancer.
CONCLUSIONS: There was consensus that during the COVID-19 pandemic, it is appropriate to defer enrollment in lung cancer screening and modify the evaluation of lung nodules due to the added risks from potential exposure and the need for resource reallocation. There are multiple local, regional, and patient-related factors that should be considered when applying these statements to individual patient care.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Betacoronavirus/isolation & purification
COVID-19
Carcinoma, Non-Small-Cell Lung/*diagnosis
Consensus
*Coronavirus Infections/diagnosis/epidemiology
Early Detection of Cancer/methods/standards
Humans
*Lung Neoplasms/diagnostic imaging/pathology
Multiple Pulmonary Nodules/*diagnosis
Neoplasm Staging
*Pandemics/prevention & control
*Pneumonia, Viral/diagnosis/epidemiology
Radiography, Thoracic/*methods
Resource Allocation
Risk Assessment/methods
SARS-CoV-2
RevDate: 2024-08-05
CmpDate: 2020-06-25
Neuroanesthesia Practice During the COVID-19 Pandemic: Recommendations From Society for Neuroscience in Anesthesiology and Critical Care (SNACC).
Journal of neurosurgical anesthesiology, 32(3):202-209.
The pandemic of coronavirus disease 2019 (COVID-19) has several implications relevant to neuroanesthesiologists, including neurological manifestations of the disease, impact of anesthesia provision for specific neurosurgical procedures and electroconvulsive therapy, and health care provider wellness. The Society for Neuroscience in Anesthesiology and Critical Care appointed a task force to provide timely, consensus-based expert guidance for neuroanesthesiologists during the COVID-19 pandemic. The aim of this document is to provide a focused overview of COVID-19 disease relevant to neuroanesthesia practice. This consensus statement provides information on the neurological manifestations of COVID-19, advice for neuroanesthesia clinical practice during emergent neurosurgery, interventional radiology (excluding endovascular treatment of acute ischemic stroke), transnasal neurosurgery, awake craniotomy and electroconvulsive therapy, as well as information about health care provider wellness. Institutions and health care providers are encouraged to adapt these recommendations to best suit local needs, considering existing practice standards and resource availability to ensure safety of patients and providers.
Additional Links: PMID-32301764
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32301764,
year = {2020},
author = {Flexman, AM and Abcejo, AS and Avitsian, R and De Sloovere, V and Highton, D and Juul, N and Li, S and Meng, L and Paisansathan, C and Rath, GP and Rozet, I},
title = {Neuroanesthesia Practice During the COVID-19 Pandemic: Recommendations From Society for Neuroscience in Anesthesiology and Critical Care (SNACC).},
journal = {Journal of neurosurgical anesthesiology},
volume = {32},
number = {3},
pages = {202-209},
pmid = {32301764},
issn = {1537-1921},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Anesthesia/*methods ; Betacoronavirus ; Brain Ischemia/complications/*surgery ; COVID-19 ; Coronavirus Infections/*prevention & control ; Critical Care ; Humans ; Neurosurgery/*methods ; Pandemics/*prevention & control ; Pneumonia, Viral/*prevention & control ; SARS-CoV-2 ; Societies, Medical ; Stroke/complications/*surgery ; },
abstract = {The pandemic of coronavirus disease 2019 (COVID-19) has several implications relevant to neuroanesthesiologists, including neurological manifestations of the disease, impact of anesthesia provision for specific neurosurgical procedures and electroconvulsive therapy, and health care provider wellness. The Society for Neuroscience in Anesthesiology and Critical Care appointed a task force to provide timely, consensus-based expert guidance for neuroanesthesiologists during the COVID-19 pandemic. The aim of this document is to provide a focused overview of COVID-19 disease relevant to neuroanesthesia practice. This consensus statement provides information on the neurological manifestations of COVID-19, advice for neuroanesthesia clinical practice during emergent neurosurgery, interventional radiology (excluding endovascular treatment of acute ischemic stroke), transnasal neurosurgery, awake craniotomy and electroconvulsive therapy, as well as information about health care provider wellness. Institutions and health care providers are encouraged to adapt these recommendations to best suit local needs, considering existing practice standards and resource availability to ensure safety of patients and providers.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Anesthesia/*methods
Betacoronavirus
Brain Ischemia/complications/*surgery
COVID-19
Coronavirus Infections/*prevention & control
Critical Care
Humans
Neurosurgery/*methods
Pandemics/*prevention & control
Pneumonia, Viral/*prevention & control
SARS-CoV-2
Societies, Medical
Stroke/complications/*surgery
RevDate: 2022-12-07
CmpDate: 2020-04-28
Guidance for the management of myasthenia gravis (MG) and Lambert-Eaton myasthenic syndrome (LEMS) during the COVID-19 pandemic.
Journal of the neurological sciences, 412:116803.
Additional Links: PMID-32247193
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32247193,
year = {2020},
author = {, and Jacob, S and Muppidi, S and Guidon, A and Guptill, J and Hehir, M and Howard, JF and Illa, I and Mantegazza, R and Murai, H and Utsugisawa, K and Vissing, J and Wiendl, H and Nowak, RJ},
title = {Guidance for the management of myasthenia gravis (MG) and Lambert-Eaton myasthenic syndrome (LEMS) during the COVID-19 pandemic.},
journal = {Journal of the neurological sciences},
volume = {412},
number = {},
pages = {116803},
doi = {10.1016/j.jns.2020.116803},
pmid = {32247193},
issn = {1878-5883},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {*Betacoronavirus ; COVID-19 ; Clinical Trials as Topic ; Coronavirus Infections/*complications/drug therapy/*epidemiology ; Disease Management ; Disease Susceptibility ; Humans ; Immunoglobulins, Intravenous/therapeutic use ; Immunologic Factors/therapeutic use ; Immunosuppressive Agents/therapeutic use ; Immunotherapy ; Lambert-Eaton Myasthenic Syndrome/complications/drug therapy/*therapy ; Myasthenia Gravis/*complications/drug therapy/*therapy ; *Pandemics ; Plasma Exchange ; Pneumonia, Viral/*complications/drug therapy/*epidemiology ; SARS-CoV-2 ; Vaccines ; COVID-19 Drug Treatment ; },
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Betacoronavirus
COVID-19
Clinical Trials as Topic
Coronavirus Infections/*complications/drug therapy/*epidemiology
Disease Management
Disease Susceptibility
Humans
Immunoglobulins, Intravenous/therapeutic use
Immunologic Factors/therapeutic use
Immunosuppressive Agents/therapeutic use
Immunotherapy
Lambert-Eaton Myasthenic Syndrome/complications/drug therapy/*therapy
Myasthenia Gravis/*complications/drug therapy/*therapy
*Pandemics
Plasma Exchange
Pneumonia, Viral/*complications/drug therapy/*epidemiology
SARS-CoV-2
Vaccines
COVID-19 Drug Treatment
RevDate: 2021-09-19
CmpDate: 2020-09-09
Guidance for cardiac electrophysiology during the COVID-19 pandemic from the Heart Rhythm Society COVID-19 Task Force; Electrophysiology Section of the American College of Cardiology; and the Electrocardiography and Arrhythmias Committee of the Council on Clinical Cardiology, American Heart Association.
Heart rhythm, 17(9):e233-e241.
Coronavirus disease 2019 (COVID-19) is a global pandemic that is wreaking havoc on the health and economy of much of human civilization. Electrophysiologists have been impacted personally and professionally by this global catastrophe. In this joint article from representatives of the Heart Rhythm Society, the American College of Cardiology, and the American Heart Association, we identify the potential risks of exposure to patients, allied healthcare staff, industry representatives, and hospital administrators. We also describe the impact of COVID-19 on cardiac arrhythmias and methods of triage based on acuity and patient comorbidities. We provide guidance for managing invasive and noninvasive electrophysiology procedures, clinic visits, and cardiac device interrogations. In addition, we discuss resource conservation and the role of telemedicine in remote patient care along with management strategies for affected patients.
Additional Links: PMID-32247013
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32247013,
year = {2020},
author = {Lakkireddy, DR and Chung, MK and Gopinathannair, R and Patton, KK and Gluckman, TJ and Turagam, M and Cheung, JW and Patel, P and Sotomonte, J and Lampert, R and Han, JK and Rajagopalan, B and Eckhardt, L and Joglar, J and Sandau, KE and Olshansky, B and Wan, E and Noseworthy, PA and Leal, M and Kaufman, E and Gutierrez, A and Marine, JE and Wang, PJ and Russo, AM},
title = {Guidance for cardiac electrophysiology during the COVID-19 pandemic from the Heart Rhythm Society COVID-19 Task Force; Electrophysiology Section of the American College of Cardiology; and the Electrocardiography and Arrhythmias Committee of the Council on Clinical Cardiology, American Heart Association.},
journal = {Heart rhythm},
volume = {17},
number = {9},
pages = {e233-e241},
pmid = {32247013},
issn = {1556-3871},
support = {UL1 TR001863/TR/NCATS NIH HHS/United States ; },
mesh = {Arrhythmias, Cardiac/*diagnosis/etiology/*therapy ; *Betacoronavirus ; COVID-19 ; Coronavirus Infections/complications/epidemiology/*prevention & control ; *Electrocardiography ; *Electrophysiologic Techniques, Cardiac ; Humans ; Infection Control/organization & administration ; Pandemics/*prevention & control ; Pneumonia, Viral/complications/epidemiology/*prevention & control ; SARS-CoV-2 ; Telemedicine/organization & administration ; Triage/organization & administration ; },
abstract = {Coronavirus disease 2019 (COVID-19) is a global pandemic that is wreaking havoc on the health and economy of much of human civilization. Electrophysiologists have been impacted personally and professionally by this global catastrophe. In this joint article from representatives of the Heart Rhythm Society, the American College of Cardiology, and the American Heart Association, we identify the potential risks of exposure to patients, allied healthcare staff, industry representatives, and hospital administrators. We also describe the impact of COVID-19 on cardiac arrhythmias and methods of triage based on acuity and patient comorbidities. We provide guidance for managing invasive and noninvasive electrophysiology procedures, clinic visits, and cardiac device interrogations. In addition, we discuss resource conservation and the role of telemedicine in remote patient care along with management strategies for affected patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Arrhythmias, Cardiac/*diagnosis/etiology/*therapy
*Betacoronavirus
COVID-19
Coronavirus Infections/complications/epidemiology/*prevention & control
*Electrocardiography
*Electrophysiologic Techniques, Cardiac
Humans
Infection Control/organization & administration
Pandemics/*prevention & control
Pneumonia, Viral/complications/epidemiology/*prevention & control
SARS-CoV-2
Telemedicine/organization & administration
Triage/organization & administration
RevDate: 2024-08-21
CmpDate: 2020-05-14
Surviving Sepsis Campaign: guidelines on the management of critically ill adults with Coronavirus Disease 2019 (COVID-19).
Intensive care medicine, 46(5):854-887.
BACKGROUND: The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of a rapidly spreading illness, Coronavirus Disease 2019 (COVID-19), affecting thousands of people around the world. Urgent guidance for clinicians caring for the sickest of these patients is needed.
METHODS: We formed a panel of 36 experts from 12 countries. All panel members completed the World Health Organization conflict of interest disclosure form. The panel proposed 53 questions that are relevant to the management of COVID-19 in the ICU. We searched the literature for direct and indirect evidence on the management of COVID-19 in critically ill patients in the ICU. We identified relevant and recent systematic reviews on most questions relating to supportive care. We assessed the certainty in the evidence using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach, then generated recommendations based on the balance between benefit and harm, resource and cost implications, equity, and feasibility. Recommendations were either strong or weak, or in the form of best practice recommendations.
RESULTS: The Surviving Sepsis Campaign COVID-19 panel issued 54 statements, of which 4 are best practice statements, 9 are strong recommendations, and 35 are weak recommendations. No recommendation was provided for 6 questions. The topics were: (1) infection control, (2) laboratory diagnosis and specimens, (3) hemodynamic support, (4) ventilatory support, and (5) COVID-19 therapy.
CONCLUSION: The Surviving Sepsis Campaign COVID-19 panel issued several recommendations to help support healthcare workers caring for critically ill ICU patients with COVID-19. When available, we will provide new recommendations in further releases of these guidelines.
Additional Links: PMID-32222812
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid32222812,
year = {2020},
author = {Alhazzani, W and Møller, MH and Arabi, YM and Loeb, M and Gong, MN and Fan, E and Oczkowski, S and Levy, MM and Derde, L and Dzierba, A and Du, B and Aboodi, M and Wunsch, H and Cecconi, M and Koh, Y and Chertow, DS and Maitland, K and Alshamsi, F and Belley-Cote, E and Greco, M and Laundy, M and Morgan, JS and Kesecioglu, J and McGeer, A and Mermel, L and Mammen, MJ and Alexander, PE and Arrington, A and Centofanti, JE and Citerio, G and Baw, B and Memish, ZA and Hammond, N and Hayden, FG and Evans, L and Rhodes, A},
title = {Surviving Sepsis Campaign: guidelines on the management of critically ill adults with Coronavirus Disease 2019 (COVID-19).},
journal = {Intensive care medicine},
volume = {46},
number = {5},
pages = {854-887},
pmid = {32222812},
issn = {1432-1238},
mesh = {COVID-19 ; Coronavirus Infections/prevention & control/*therapy ; Critical Care/*standards ; Humans ; Intensive Care Units/*standards ; Pandemics/prevention & control ; Pneumonia, Viral/prevention & control/*therapy ; Sepsis/diagnosis/etiology/*therapy ; Survivors ; },
abstract = {BACKGROUND: The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of a rapidly spreading illness, Coronavirus Disease 2019 (COVID-19), affecting thousands of people around the world. Urgent guidance for clinicians caring for the sickest of these patients is needed.
METHODS: We formed a panel of 36 experts from 12 countries. All panel members completed the World Health Organization conflict of interest disclosure form. The panel proposed 53 questions that are relevant to the management of COVID-19 in the ICU. We searched the literature for direct and indirect evidence on the management of COVID-19 in critically ill patients in the ICU. We identified relevant and recent systematic reviews on most questions relating to supportive care. We assessed the certainty in the evidence using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach, then generated recommendations based on the balance between benefit and harm, resource and cost implications, equity, and feasibility. Recommendations were either strong or weak, or in the form of best practice recommendations.
RESULTS: The Surviving Sepsis Campaign COVID-19 panel issued 54 statements, of which 4 are best practice statements, 9 are strong recommendations, and 35 are weak recommendations. No recommendation was provided for 6 questions. The topics were: (1) infection control, (2) laboratory diagnosis and specimens, (3) hemodynamic support, (4) ventilatory support, and (5) COVID-19 therapy.
CONCLUSION: The Surviving Sepsis Campaign COVID-19 panel issued several recommendations to help support healthcare workers caring for critically ill ICU patients with COVID-19. When available, we will provide new recommendations in further releases of these guidelines.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
COVID-19
Coronavirus Infections/prevention & control/*therapy
Critical Care/*standards
Humans
Intensive Care Units/*standards
Pandemics/prevention & control
Pneumonia, Viral/prevention & control/*therapy
Sepsis/diagnosis/etiology/*therapy
Survivors
RevDate: 2025-06-18
CmpDate: 2025-06-04
[Meta analysis of the prevalence of depressive symptoms among children and adolescents in China from 2015 to 2024].
Zhongguo dang dai er ke za zhi = Chinese journal of contemporary pediatrics, 27(5):529-539.
OBJECTIVES: To systematically evaluate the prevalence of depressive symptoms among children and adolescents in China.
METHODS: A comprehensive search was conducted in PubMed, Embase, Cochrane Library, Web of Science, Wanfang Data, China Biomedical Literature Database, China National Knowledge Infrastructure, and VIP Database for literature published from January 2015 to May 2024 regarding the prevalence of depressive symptoms among Chinese children and adolescents. Stata 16.0 software was used for Meta analysis, and Egger's test and funnel plot analysis were performed to assess publication bias.
RESULTS: A total of 53 studies were included, encompassing 314 867 children and adolescents, with 65 324 exhibiting depressive symptoms, resulting in a prevalence rate of 23.3% (95%CI: 21.4%-25.1%). Subgroup analysis revealed higher prevalence rates among females, those in rural areas, vocational high school students, individuals from the eastern region, those assessed using the Depression Self-Rating Scale for Children, and during the COVID-19 pandemic, with rates of 24.9%, 29.5%, 28.1%, 25.6%, 27.2%, and 27.7%, respectively. The distribution of studies in the funnel plot was roughly symmetrical, but Egger's test indicated potential publication bias (P<0.001).
CONCLUSIONS: The prevalence of depressive symptoms among children and adolescents in China is relatively high, particularly among females, those in rural areas, vocational high school students, and those in the eastern regions. Additionally, the COVID-19 pandemic may have impacted the incidence of depressive symptoms in this population.
Additional Links: PMID-40462425
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40462425,
year = {2025},
author = {Feng, Y and Wang, F},
title = {[Meta analysis of the prevalence of depressive symptoms among children and adolescents in China from 2015 to 2024].},
journal = {Zhongguo dang dai er ke za zhi = Chinese journal of contemporary pediatrics},
volume = {27},
number = {5},
pages = {529-539},
pmid = {40462425},
issn = {1008-8830},
mesh = {Humans ; Adolescent ; Child ; China/epidemiology ; Prevalence ; *Depression/epidemiology ; Female ; Male ; COVID-19 ; },
abstract = {OBJECTIVES: To systematically evaluate the prevalence of depressive symptoms among children and adolescents in China.
METHODS: A comprehensive search was conducted in PubMed, Embase, Cochrane Library, Web of Science, Wanfang Data, China Biomedical Literature Database, China National Knowledge Infrastructure, and VIP Database for literature published from January 2015 to May 2024 regarding the prevalence of depressive symptoms among Chinese children and adolescents. Stata 16.0 software was used for Meta analysis, and Egger's test and funnel plot analysis were performed to assess publication bias.
RESULTS: A total of 53 studies were included, encompassing 314 867 children and adolescents, with 65 324 exhibiting depressive symptoms, resulting in a prevalence rate of 23.3% (95%CI: 21.4%-25.1%). Subgroup analysis revealed higher prevalence rates among females, those in rural areas, vocational high school students, individuals from the eastern region, those assessed using the Depression Self-Rating Scale for Children, and during the COVID-19 pandemic, with rates of 24.9%, 29.5%, 28.1%, 25.6%, 27.2%, and 27.7%, respectively. The distribution of studies in the funnel plot was roughly symmetrical, but Egger's test indicated potential publication bias (P<0.001).
CONCLUSIONS: The prevalence of depressive symptoms among children and adolescents in China is relatively high, particularly among females, those in rural areas, vocational high school students, and those in the eastern regions. Additionally, the COVID-19 pandemic may have impacted the incidence of depressive symptoms in this population.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Adolescent
Child
China/epidemiology
Prevalence
*Depression/epidemiology
Female
Male
COVID-19
RevDate: 2026-01-11
CmpDate: 2026-01-11
mRNA-Encoded antibodies as a next-generation therapeutic paradigm: a rapid and adaptive platform for the prevention and treatment of emerging and re-emerging infectious diseases - A critical review.
Immunologic research, 74(1):7.
Messenger RNA (mRNA)-encoded antibodies represent a transformative therapeutic platform with the potential to rapidly combat emerging infectious diseases by enabling in situ expression of potent neutralizing antibodies directly in the patient's body. Unlike conventional monoclonal antibody (mAb) therapies, which rely on labor-intensive and time-consuming cell culture production, mRNA-encoded antibodies offer a faster, scalable, and cell-free approach that bypasses protein purification and cold-chain constraints. This strategy has demonstrated considerable promise during the COVID-19 pandemic, where Moderna's mRNA-1940, an mRNA-based neutralizing antibody targeting the SARS-CoV-2 spike protein, entered preclinical and early-phase trials within months of viral emergence, underscoring the potential for rapid response in outbreak settings. The platform leverages advances in nucleoside-modified mRNA, codon optimization, and lipid nanoparticle (LNP) delivery systems to achieve transient, high-level expression of functional antibodies with reduced innate immune activation. Beyond COVID-19, mRNA-encoded antibody approaches have been explored in preclinical models of Zika virus, Ebola virus, and rabies, where a single intramuscular dose provided prophylactic and therapeutic benefits in animal models. As the world faces recurrent viral threats, the development of mRNA-encoded antibodies as a plug-and-play system offers a compelling, adaptable, and clinically feasible strategy for infectious disease preparedness. This review explores the mechanistic foundation, delivery technologies, translational progress, case studies, safety considerations, and future clinical potential of mRNA-encoded antibodies in combating both pandemic and endemic infections.
Additional Links: PMID-41521363
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41521363,
year = {2026},
author = {Singh, D},
title = {mRNA-Encoded antibodies as a next-generation therapeutic paradigm: a rapid and adaptive platform for the prevention and treatment of emerging and re-emerging infectious diseases - A critical review.},
journal = {Immunologic research},
volume = {74},
number = {1},
pages = {7},
pmid = {41521363},
issn = {1559-0755},
mesh = {Humans ; *COVID-19/immunology/prevention & control/therapy ; *SARS-CoV-2/immunology ; *Antibodies, Neutralizing/genetics/therapeutic use/immunology ; Animals ; *RNA, Messenger/genetics ; Spike Glycoprotein, Coronavirus/immunology ; *Antibodies, Viral/genetics/therapeutic use/immunology ; *Communicable Diseases, Emerging/therapy/immunology/prevention & control ; },
abstract = {Messenger RNA (mRNA)-encoded antibodies represent a transformative therapeutic platform with the potential to rapidly combat emerging infectious diseases by enabling in situ expression of potent neutralizing antibodies directly in the patient's body. Unlike conventional monoclonal antibody (mAb) therapies, which rely on labor-intensive and time-consuming cell culture production, mRNA-encoded antibodies offer a faster, scalable, and cell-free approach that bypasses protein purification and cold-chain constraints. This strategy has demonstrated considerable promise during the COVID-19 pandemic, where Moderna's mRNA-1940, an mRNA-based neutralizing antibody targeting the SARS-CoV-2 spike protein, entered preclinical and early-phase trials within months of viral emergence, underscoring the potential for rapid response in outbreak settings. The platform leverages advances in nucleoside-modified mRNA, codon optimization, and lipid nanoparticle (LNP) delivery systems to achieve transient, high-level expression of functional antibodies with reduced innate immune activation. Beyond COVID-19, mRNA-encoded antibody approaches have been explored in preclinical models of Zika virus, Ebola virus, and rabies, where a single intramuscular dose provided prophylactic and therapeutic benefits in animal models. As the world faces recurrent viral threats, the development of mRNA-encoded antibodies as a plug-and-play system offers a compelling, adaptable, and clinically feasible strategy for infectious disease preparedness. This review explores the mechanistic foundation, delivery technologies, translational progress, case studies, safety considerations, and future clinical potential of mRNA-encoded antibodies in combating both pandemic and endemic infections.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/immunology/prevention & control/therapy
*SARS-CoV-2/immunology
*Antibodies, Neutralizing/genetics/therapeutic use/immunology
Animals
*RNA, Messenger/genetics
Spike Glycoprotein, Coronavirus/immunology
*Antibodies, Viral/genetics/therapeutic use/immunology
*Communicable Diseases, Emerging/therapy/immunology/prevention & control
RevDate: 2026-01-11
Advancements in nanotrap technology for the prevention, diagnosis and treatment of infectious diseases.
Nanomedicine (London, England) [Epub ahead of print].
Nanotraps are particles designed to capture and concentrate target molecules and have numerous applications in infectious diseases. This review outlines how nanotrap technologies may improve the detection and treatment of bacterial and viral pathogens, including Mycobacterium tuberculosis, Borrelia burgdorferi, Yersinia pestis, HIV, SARS-CoV-2, and others. Nanotraps can enhance the sensitivity of diagnostic tools and support treatment by neutralizing bacterial toxins, capturing inflammatory mediators, and preserving viral proteins for detection. Nanotraps have also been investigated for vaccine development. While results from in vitro and in vivo models are encouraging, there is significant room for further research regarding safety and other unexplored applications of these technologies. Nanotraps offer a flexible platform with the potential to improve how we diagnose and manage a multitude of infectious diseases.
Additional Links: PMID-41521058
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41521058,
year = {2026},
author = {Keefer, S and Lorenzo-Leal, AC and Bach, H},
title = {Advancements in nanotrap technology for the prevention, diagnosis and treatment of infectious diseases.},
journal = {Nanomedicine (London, England)},
volume = {},
number = {},
pages = {1-11},
doi = {10.1080/17435889.2026.2614545},
pmid = {41521058},
issn = {1748-6963},
abstract = {Nanotraps are particles designed to capture and concentrate target molecules and have numerous applications in infectious diseases. This review outlines how nanotrap technologies may improve the detection and treatment of bacterial and viral pathogens, including Mycobacterium tuberculosis, Borrelia burgdorferi, Yersinia pestis, HIV, SARS-CoV-2, and others. Nanotraps can enhance the sensitivity of diagnostic tools and support treatment by neutralizing bacterial toxins, capturing inflammatory mediators, and preserving viral proteins for detection. Nanotraps have also been investigated for vaccine development. While results from in vitro and in vivo models are encouraging, there is significant room for further research regarding safety and other unexplored applications of these technologies. Nanotraps offer a flexible platform with the potential to improve how we diagnose and manage a multitude of infectious diseases.},
}
▼ ▼ LOAD NEXT 100 CITATIONS
RJR Experience and Expertise
Researcher
Robbins holds BS, MS, and PhD degrees in the life sciences. He served as a tenured faculty member in the Zoology and Biological Science departments at Michigan State University. He is currently exploring the intersection between genomics, microbial ecology, and biodiversity — an area that promises to transform our understanding of the biosphere.
Educator
Robbins has extensive experience in college-level education: At MSU he taught introductory biology, genetics, and population genetics. At JHU, he was an instructor for a special course on biological database design. At FHCRC, he team-taught a graduate-level course on the history of genetics. At Bellevue College he taught medical informatics.
Administrator
Robbins has been involved in science administration at both the federal and the institutional levels. At NSF he was a program officer for database activities in the life sciences, at DOE he was a program officer for information infrastructure in the human genome project. At the Fred Hutchinson Cancer Research Center, he served as a vice president for fifteen years.
Technologist
Robbins has been involved with information technology since writing his first Fortran program as a college student. At NSF he was the first program officer for database activities in the life sciences. At JHU he held an appointment in the CS department and served as director of the informatics core for the Genome Data Base. At the FHCRC he was VP for Information Technology.
Publisher
While still at Michigan State, Robbins started his first publishing venture, founding a small company that addressed the short-run publishing needs of instructors in very large undergraduate classes. For more than 20 years, Robbins has been operating The Electronic Scholarly Publishing Project, a web site dedicated to the digital publishing of critical works in science, especially classical genetics.
Speaker
Robbins is well-known for his speaking abilities and is often called upon to provide keynote or plenary addresses at international meetings. For example, in July, 2012, he gave a well-received keynote address at the Global Biodiversity Informatics Congress, sponsored by GBIF and held in Copenhagen. The slides from that talk can be seen HERE.
Facilitator
Robbins is a skilled meeting facilitator. He prefers a participatory approach, with part of the meeting involving dynamic breakout groups, created by the participants in real time: (1) individuals propose breakout groups; (2) everyone signs up for one (or more) groups; (3) the groups with the most interested parties then meet, with reports from each group presented and discussed in a subsequent plenary session.
Designer
Robbins has been engaged with photography and design since the 1960s, when he worked for a professional photography laboratory. He now prefers digital photography and tools for their precision and reproducibility. He designed his first web site more than 20 years ago and he personally designed and implemented this web site. He engages in graphic design as a hobby.
RJR Picks from Around the Web (updated 11 MAY 2018 )
Old Science
Weird Science
Treating Disease with Fecal Transplantation
Fossils of miniature humans (hobbits) discovered in Indonesia
Paleontology
Dinosaur tail, complete with feathers, found preserved in amber.
Astronomy
Mysterious fast radio burst (FRB) detected in the distant universe.
Big Data & Informatics
Big Data: Buzzword or Big Deal?
Hacking the genome: Identifying anonymized human subjects using publicly available data.