1
|
Xue M, Liao F, Xu F, Chen Y, Wang S, Zhou Y, Ding H, Lu S, Yao C, Song Z, Shao M. A retrospective study to predict failure of high-flow oxygen therapy for acute hypoxic respiratory failure. Int J Emerg Med 2025; 18:98. [PMID: 40375069 PMCID: PMC12079891 DOI: 10.1186/s12245-025-00891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/26/2025] [Indexed: 05/18/2025] Open
Abstract
OBJECTIVE This study aimed to analyze the characteristics of patients who fail high-flow nasal cannula(HFNC) therapy for acute hypoxemic respiratory failure(AHRF) and to identify predictors of treatment failure. METHODS This single-center, retrospective, observational study analyzed clinical data from 388 patients with AHRF. Patients were divided into two groups: the HFNC success group (HFNC-S, n = 256) and the HFNC failure group (HFNC-F, n = 132). The primary endpoint was the need for escalation of respiratory support to tracheal intubation in the enrolled patients. The demographic data, laboratory tests, blood gas analysis data, CT severity scores, and disease severity scores were analysed to determine the difference between patients who were successful and those who failed HFNC treatment. Univariate and multivariate logistic regression models were used to assess potential predictors of failure of HFNC for patients with acute hypoxaemic respiratory failure. RESULTS The mean age of patients enrolled was 67.97 ± 14.40 years. The HFNC-F group had significantly higher PSI(Pneumonia Severity Index) score, CURB(Confusion, Urea, Respiratory Rate, Blood Pressure, and Age)-65 score, CPIS(Clinical Pulmonary Infection Score) score, CT score and SOFA(Sequential Organ Failure Assessment) scores compared to the HFNC-S group. Within 12 h of the initiation of treatment, the HFNC-F group exhibited significantly lower oxygen saturation index (PaO2/FiO2) and significantly higher respiratory rate. Additionally, the HFNC-F group exhibited significantly higher levels of C-reactive protein (CRP), platelet count (PLT), D-dimer, interleukin-10 (IL-10), total bilirubin (TB) and creatinine (CB), but lower albumin levels. Multivariate analysis identified CT score, SOFA score, interleukin-1β (IL-1β), and albumin as independent predictors of HFNC failure. CONCLUSION HFNC is effective for treating AHRF. CT score, SOFA score, IL-1β, and albumin are independent predictors of HFNC failure.
Collapse
Affiliation(s)
- Mingming Xue
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fengqing Liao
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Feixiang Xu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yumei Chen
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Sheng Wang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yannan Zhou
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hailin Ding
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Su Lu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenling Yao
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhenju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Mian Shao
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Song H, Yang J, Li J, Deng C, Zhang S, Zheng S. C-reactive protein to lymphocyte ratio combined with clinical features to construct a predictive model for upper gastrointestinal bleeding due to peptic ulcer. Clinics (Sao Paulo) 2025; 80:100644. [PMID: 40273489 PMCID: PMC12051625 DOI: 10.1016/j.clinsp.2025.100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE This research aimed to determine the feasibility and accuracy of CLR and clinical features to formulate a prediction model for Peptic Ulcer (PU)-induced Upper Gastrointestinal Bleeding (UGIB). METHODS The clinical data of 146 PU patients were prospectively collected, and patients were divided into the UGIB group (n = 48) and the non-UGIB group (n = 98). The factors affecting UGIB were analyzed using multifactorial logistic regression and collinearity analysis. The prediction model of UGIB was constructed, the predictive value of which was analyzed using the Receiver Operating Characteristic Curve (ROC) and Area Under the Curve (AUC), while the accuracy was analyzed using the calibration curve and Hosmer Lemeshow goodness-of-fit tests, and the application value was assessed using decision curve analysis (DCA). RESULTS Statistical significance was observed between the two groups regarding HP infection, ulcer diameter, ulcer stage, use of nonsteroidal anti-inflammatory drugs, Neutrophil, LYM, NEUT/LYM Ratio (NLR), CRP, and CLR. HP infection, ulcer stage, use of NSAIDs, NLR, and CLR were independent risk factors for UGIB, and PCT was a non-independent risk factor. The AUC for this model was 0.921. The calibration curve of the model matched the actual curve. The model achieved a better fitting effect in predicting UGIB (χ2 = 8.5069, df = 8, p = 0.3856) and had a better clinical application value. CONCLUSION A predictive model for PU-induced UGIB, based on CLR and clinical features, can assist in developing clinical treatment plans to prevent UGIB.
Collapse
Affiliation(s)
- Hong Song
- Department of Gerontology, Taiyuan Central Hospital, Taiyuan City, Shanxi Province, China
| | - Juan Yang
- Department of Gastroenterology, Affiliated Hospital of Yunnan University, Kunming City, Yunnan Province, China
| | - Jiao Li
- Graduate School of Clinical Medicine, Dali University, Dali City, Yunnan Province, China
| | - Cui Deng
- Graduate School of Clinical Medicine, Dali University, Dali City, Yunnan Province, China
| | - SiMin Zhang
- Graduate School of Clinical Medicine, Dali University, Dali City, Yunnan Province, China
| | - Sheng Zheng
- Department of Gastroenterology, The Third People's Hospital of Yunnan Province, Kunming City, Yunnan Province, China.
| |
Collapse
|
3
|
Miroshnichenko MI, Kolpakov FA, Akberdin IR. A Modular Mathematical Model of the Immune Response for Investigating the Pathogenesis of Infectious Diseases. Viruses 2025; 17:589. [PMID: 40431602 DOI: 10.3390/v17050589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
The COVID-19 pandemic highlighted the importance of mathematical modeling for understanding viral infection dynamics and accelerated its application into immunological research. Collaborative efforts among international research groups yielded a wealth of experimental data, which facilitated model development and validation. This study focuses on developing a modular mathematical model of the immune response, capturing the interactions between innate and adaptive immunity, with an application to SARS-CoV-2 infection. The model was validated using experimental data from middle-aged individuals with moderate COVID-19 progression, including measurements of viral load in the upper and lower airways, serum antibodies, CD4+ and CD8+ T cells, and interleukin-6 levels. Parameter optimization and sensitivity analysis were performed to improve the model accuracy. Additionally, identifiability analysis was conducted to assess whether the data were sufficient for reliable parameter estimation. The verified model simulates the dynamics of moderate, severe, and critical COVID-19 progressions using measured data on lung epithelium damage, viral load, and IL-6 levels as key indicators of disease severity. We also performed a series of validation scenarios to assess whether the model correctly reproduces biologically relevant behaviors under various conditions, such as immunity hyperactivation, co-infection with HIV, and interferon administration as a therapeutic strategy. The model was developed as a component of the Digital Twin project and represents a general immune module that integrates both innate and adaptive immunity. It can be utilized for further COVID-19 research or serve as a foundation for studying other infectious diseases, provided sufficient data are available.
Collapse
Affiliation(s)
- Maxim I Miroshnichenko
- Department of Computational Biology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Fedor A Kolpakov
- Department of Computational Biology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ilya R Akberdin
- Department of Computational Biology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
4
|
Astroth C, Shah KS, Agrawal S, Agrawal A. Weathering the Storm: How Age and Biologics Influence the COVID-19 Cytokine Surge. Pathogens 2025; 14:346. [PMID: 40333142 PMCID: PMC12030216 DOI: 10.3390/pathogens14040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025] Open
Abstract
SARS-CoV-2, first identified in December 2019, caused a global pandemic, resulting in over 6.8 million deaths by March 2023. The elderly, or individuals over 65, accounted for the majority of COVID-19 deaths, with 81% of fatalities in the US in 2020 occurring in this group. Beyond mortality, aging populations are also at higher risk of long-term cardiovascular complications and acute respiratory distress syndrome (ARDS). Although these outcomes may be influenced by comorbidities common in the elderly, age has been found to be a standalone risk factor for severe COVID-19 infection. Therefore, investigating age-related factors in COVID-19 outcomes is crucial in protecting this vulnerable group. Of particular interest is the cytokine storm phenomenon, an excessive inflammatory response that contributes to severe COVID-19 symptoms, including ARDS and cardiovascular damage. Elevated levels of multiple cytokines are common in severe cases of COVID-19. We propose that changes that occur to cytokine profiles as we age may contribute to these aberrant inflammatory responses. This review specifically explored the interleukin class cytokines IL-1, IL-6, IL-17, and IL-23 and considered the potential of biologics targeting these cytokines to alleviate severe outcomes in both COVID-19 and aging individuals.
Collapse
Affiliation(s)
| | | | | | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA 92697, USA; (C.A.); (K.S.S.); (S.A.)
| |
Collapse
|
5
|
Hashemian SM, Jafari A, Khoundabi B, Jamaati H, Rahimi P. Hemoperfusion Combined With Continuous Renal Replacement Therapy in the Management of ARDS COVID-19 Patients: A Quasi-Experimental Study. Health Sci Rep 2025; 8:e70571. [PMID: 40177411 PMCID: PMC11961550 DOI: 10.1002/hsr2.70571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/24/2025] [Accepted: 02/22/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aims Critically ill patients in COVID-19 to the intensive care unit (ICU) may develop multiple organ dysfunction syndrome, with some requiring extracorporeal organ support. This study aimed to assess the effects of combined CytoSorb hemoperfusion (HP) and continuous renal replacement therapy (CRRT) on the improvement of the multiorgan failure of patients with COVID-19. Methods Fifty-six patients hospitalized in the ICU with a confirmed diagnosis of COVID-19 were included in this quasi-experimental study. All the patients had acute respiratory distress syndrome (ARDS). They were treated with 1-4 sessions of HP therapy. Results Serum Interleukin-6 (IL6), C-reactive protein (CRP), d-dimer, procalcitonin (PCT), Neutrophil gelatinase-associated lipocalin (NGAL), ferritin, and bilirubin levels were decreased, while the concentration of albumin was significantly increased after HP/CRRT (p < 0.05). No significant differences were observed in O2 saturation (Sao2) and creatinine levels. Conclusion Combined HP and CRRT hold promise as a potential intervention for severe COVID-19 cases with multiple organ dysfunction, leading to improved clinical outcomes.
Collapse
Affiliation(s)
- Seyed MohammadReza Hashemian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD)Shahid Beheshti University of Medical SciencesTehranIran
| | - Ameneh Jafari
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD)Shahid Beheshti University of Medical SciencesTehranIran
| | - Batoul Khoundabi
- Iran Helal Institute of Applied‐Science and TechnologyRed Crescent Society of IranTehranIran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD)Shahid Beheshti University of Medical SciencesTehranIran
| | - Payam Rahimi
- Department of Anesthesiology and Reanimation, Bakırköy Dr. Sadi Konuk Training and Research HospitalUniversity of Health SciencesIstanbulTurkey
| |
Collapse
|
6
|
Delaunay E, Poussard N, Gourjon G, Frouin A, Ducq P, Di Vico L, Moschietto S, Larcher R, Pradel G. Role of InterLeukin-6 monitoring during weaning from volume-controlled ventilation in patients with COVID-19 acute respiratory distress syndrome. Sci Prog 2025; 108:368504251335850. [PMID: 40241622 PMCID: PMC12035380 DOI: 10.1177/00368504251335850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
ObjectiveThe aim of this study was to assess the ability of plasma InterLeukin-6 (IL-6) monitoring to predict the failure to switch from volume-controlled ventilation to spontaneous ventilation (SV) in patients with COVID-19-related acute respiratory distress syndrome (ARDS).MethodsWe conducted an observational, single-center and prospective cohort study in the medico-surgical intensive care unit of Avignon Hospital Center. Participants were adult patients requiring invasive mechanical ventilation for COVID-19-related ARDS between August 2021 and August 2022, who were eligible for switching from volume-controlled ventilation to SV.ResultsAmong the 35 patients included in the study, 13 (37%) successfully switched from controlled ventilation to SV, while 22 failed (63%). In the failure group, mean plasma IL-6 levels were higher than in the successful group from hour 0 (defined as the moment of the switch to SV mode) to 48 h. However, differences between groups became significant from 24 h (362.8 vs. 33.6 pg/mL, P = 0.002). Interestingly, between-group differences in plasma C-reactive protein (CRP) levels were only significant between groups from 48 h (129.3 vs. 52.2 mg/L, P = 0.017). Finally, IL-6 and CRP had a similar ability to predict the failure to switch to SV mode: area under the receiving operative curves 0.763 [95%CI: 0.633-0.893] and 0.753 [95%CI: 0.595-0.911], respectively (P = 0.87).ConclusionsIL-6 and CRP are inflammatory biomarkers predictive of failure to switch to SV mode in COVID-19 ARDS patients. Our results showed that IL-6 can detect failure earlier than CRP. However, larger multicenter studies are needed to confirm our results, particularly in other ARDS models.
Collapse
Affiliation(s)
- Estelle Delaunay
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| | - Nicolas Poussard
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| | - Géraud Gourjon
- Department of Medical Statistics, Henri Duffaut Avignon General Hospital, Avignon, France
- SCientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon (IFO-GA), Avignon, France
| | - Antoine Frouin
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| | - Pierre Ducq
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| | - Lynda Di Vico
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| | | | - Romaric Larcher
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
- PhyMedExp, French National Health and Medical Research Body (INSERM), National Centre for Scientific Research (CNRS), Montpellier University, Montpellier, France
| | - Gaël Pradel
- Intensive Care Unit, Henri Duffaut Avignon General Hospital, Avignon, France
| |
Collapse
|
7
|
Alrasheed AR, Awadalla M, Alnajran H, Alammash MH, Almaqati AM, Qadri I, Alosaimi B. Harnessing immunotherapeutic molecules and diagnostic biomarkers as human-derived adjuvants for MERS-CoV vaccine development. Front Immunol 2025; 16:1538301. [PMID: 40181980 PMCID: PMC11965926 DOI: 10.3389/fimmu.2025.1538301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
The pandemic potential of the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) highlights the critical need for effective vaccines due to its high fatality rate of around 36%. In this review, we identified a variety of immunotherapeutic molecules and diagnostic biomarkers that could be used in MERS vaccine development as human-derived adjuvants. We identified immune molecules that have been incorporated into standard clinical diagnostics such as CXCL10/IP10, CXCL8/IL-8, CCL5/RANTES, IL-6, and the complement proteins Ca3 and Ca5. Utilization of different human monoclonal antibodies in the treatment of MERS-CoV patients demonstrates promising outcomes in combatting MERS-CoV infections in vivo, such as hMS-1, 4C2H, 3B11-N, NBMS10-FC, HR2P-M2, SAB-301, M336, LCA60, REGN3051, REGN3048, MCA1, MERs-4, MERs-27, MERs-gd27, and MERs-gd33. Host-derived adjuvants such as CCL28, CCL27, RANTES, TCA3, and GM-CSF have shown significant improvements in immune responses, underscoring their potential to bolster both systemic and mucosal immunity. In conclusion, we believe that host-derived adjuvants like HBD-2, CD40L, and LL-37 offer significant advantages over synthetic options in vaccine development, underscoring the need for clinical trials to validate their efficacy.
Collapse
Affiliation(s)
- Abdullah R. Alrasheed
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maaweya Awadalla
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| | - Hadeel Alnajran
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | | | - Adil M. Almaqati
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Ishtiaq Qadri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Torres-Poveda K, Bahena-Román M, Contreras-Ochoa CO, Lagunas-Martínez A, Bermúdez-Morales VH, Pando-Robles V, Ortiz-Flores E, Cortés-Pedroza F, Santana-Román ME, Martínez-Campos C, Sánchez-Alemán M, Manzo-Merino J, Morales-Ortega A, Madrid-González DA, Cantú-Cuevas MA, Barón-Olivares H, Madrid-Marina V. High nasopharyngeal and serum IL-6 levels and the - 573G > C polymorphism (rs1800796) are linked with the risk of severe COVID-19 in a Mexican population: a case‒control study. BMC Infect Dis 2025; 25:315. [PMID: 40045221 PMCID: PMC11884130 DOI: 10.1186/s12879-025-10695-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND COVID-19 was the leading cause of death in Mexico between 2020 and 2021. SARS-CoV-2 infection varies widely among individuals and populations. Since variations in genes related to the immune response may play a role in the susceptibility to and outcome of COVID-19, the associations of gene polymorphisms (SNPs) of IL-6 (- 573G > C, rs1800796), TNF-α (- 308G > A, rs1800629), and IFN-γ (- 1615 C > T, rs2069705) with the expression levels of these proteins in the nasopharynx and serum were evaluated in a Mexican population with mild, severe, or critical COVID-19. METHODS A total of 560 COVID-19 patients (309 mild, 163 severe, and 88 critical cases) and 560 age- and sex-matched COVID-19-negative controls were recruited for this case‒control study. The selected SNPs were genotyped via allelic discrimination. Logistic regression analysis was conducted considering four models of inheritance, and ORs were determined for each genotypic variant, adjusting for associated comorbidities in the multivariate model. The nasopharyngeal mRNA expression levels of IL-6, IFN-γ and TNF-α were determined. The levels of IL-6, IFN-γ, IFN-α2, and TNF-α in the serum were quantified. Significant differences were assessed via the Wilcoxon Mann‒Whitney U test. RESULTS The C allele of the IL-6 - 573 SNP was associated with a greater risk of mild and severe COVID-19 (OR: 2.3, CI: 1.897-2.838, p = 0.0001; and OR: 1.5, CI: 1.167-1.949, p = 0.002, respectively), whereas the A allele of the TNF-α - 308 SNP and the T allele of the IFN-γ - 1615 SNP were shown protective roles against severe COVID-19 (OR: 0.3, CI: 0.189-0.537, p = 0.0001; and OR: 0.7, CI: 0.563-1.006, p = 0.05) and against critical COVID-19 (OR: 0.3, CI: 0.158-0.640, p = 0.001; and OR: 0.4, CI: 0.290-0.678, p = 0.0001), adjusting for diabetes and hypertension. Nasopharyngeal IL-6 expression levels were lower in mild COVID-19 patients (p = 0.001) than in critical patients (p = 0.005). Serum IL-6 levels were significantly elevated in the critical cases (p = 0.01). CONCLUSIONS Our results revealed that the IL-6 - 573 G > C SNP and increased IL-6 nasopharyngeal and serum levels are associated with the risk of severe COVID-19 in a Mexican population.
Collapse
Affiliation(s)
- Kirvis Torres-Poveda
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Secretaria de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI)-Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Margarita Bahena-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Alfredo Lagunas-Martínez
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | | | - Victoria Pando-Robles
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Esmeralda Ortiz-Flores
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Fabiola Cortés-Pedroza
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - María E Santana-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Cecilia Martínez-Campos
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Sánchez-Alemán
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Joaquin Manzo-Merino
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ausencio Morales-Ortega
- Laboratorio Estatal de Salud Pública. Health Services of the State of Morelos, Jiutepec, Mexico
| | | | | | - Héctor Barón-Olivares
- Dirección General de Coordinación y Supervisión. Health Services of the State of Morelos, Cuernavaca, Mexico
| | - Vicente Madrid-Marina
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico.
| |
Collapse
|
9
|
Descalsota JRAD, Cana AWR, Chin II, Orcasitas JF. Identifying COVID-19 Confirmed Patients at Elevated Risk for Mortality and Need of Mechanical Ventilation Using a Novel Criteria for Hyperinflammatory Syndrome: A Retrospective Cohort, Single-center, Validation Study. ACTA MEDICA PHILIPPINA 2025; 59:104-115. [PMID: 40151221 PMCID: PMC11936773 DOI: 10.47895/amp.vi0.9370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Background and Objectives A mounting evidence links dysregulated immune response to cases of fatal pneumonia seen in COVID-19 infection. We aimed to validate the COVID-19-associated Hyperinflammatory Syndrome (cHIS) score, a novel clinical tool devised to identify those at risk for adverse outcomes, in a local population and investigate the relationship of cHIS score taken at admission and the risk of mortality and the need of mechanical ventilation. Methods This retrospective cohort study analyzed the sociodemographic, clinical, and laboratory data of 1,881 COVID-19 patients admitted at a tertiary hospital in Davao City, Philippines from January to December 2021. We calculated the cHIS score, composed of six clinical and laboratory criteria from admission, and used multivariate logistic regression to determine the risk of mortality and need of mechanical ventilation. Results The cHIS score taken at admission, regardless of cut-off value, was a significant predictor of mortality (OR 0.979 [99% CI 0.894-1.064]) and need of mechanical ventilation (OR 0.586 [99% CI 0.4975-0.6745]). Using the Youden Index, a cut-off cHIS score of 3 or more was a better predictor of mortality (sensitivity, 88.59%; specificity, 71.72%), and a cut-off score of 2 or more was a better predictor of need of mechanical ventilation (sensitivity, 84.02%; specificity, 70.82%) than other cut-off cHIS scores. Conclusion Among COVID-19 patients, the cHIS score at admission correlated with the risk of mortality and the need of mechanical ventilation. Cutoff scores of 3 and 2 had the optimal sensitivities and specificities to predict the risk of mortality and the need of mechanical ventilation, respectively.
Collapse
Affiliation(s)
| | - Abdul Walli R. Cana
- Department of Internal Medicine, Southern Philippines Medical Center, Davao City
| | - Inofel I. Chin
- Section of Infectious Diseases, Department of Internal Medicine, Southern Philippines Medical Center, Davao City
| | - Jessie F. Orcasitas
- Section of Pulmonology, Department of Internal Medicine, Southern Philippines Medical Center, Davao City
| |
Collapse
|
10
|
Peng Z, Zhou G. Progress on diagnosis and treatment of multisystem inflammatory syndrome in children. Front Immunol 2025; 16:1551122. [PMID: 40046058 PMCID: PMC11879827 DOI: 10.3389/fimmu.2025.1551122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 05/13/2025] Open
Abstract
Since the emergence of COVID-19 in December 2019, the novel SARS-CoV-2 virus has primarily affected adults, with children representing a smaller proportion of cases. However, the escalation of the pandemic has led to a notable increase in pediatric cases of Multisystem Inflammatory Syndrome in Children (MIS-C). The pathogenesis of MIS-C is largely attributed to immune-mediated mechanisms, such as cytokine storms and endothelial damage, following SARS-CoV-2 infection. In this review, we comprehensively describe MIS-C, including its definitions as proposed by the CDC, WHO, and RCPCH, which emphasize persistent fever, excessive inflammatory responses, and multi-organ involvement. Additionally, we summarize current treatment approaches, prioritizing immunotherapy with intravenous immunoglobulin and corticosteroids, along with anticoagulation therapy, and monoclonal antibodies in severe cases.
Collapse
Affiliation(s)
| | - Gang Zhou
- Department of Pediatric Respiratory Diseases, Chongqing University Three Gorges Hospital, Chongqing, China
| |
Collapse
|
11
|
El Azhary K, Ghazi B, Kouhen F, El Bakkouri J, Chamlal H, El Ghanmi A, Badou A. Clinical Impact of Neutrophil Variation on COVID-19 Complications. Diagnostics (Basel) 2025; 15:457. [PMID: 40002608 PMCID: PMC11854688 DOI: 10.3390/diagnostics15040457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Corona virus disease 2019 (COVID-19) poses a threat to global public health. The early identification of critical cases is crucial to providing timely treatment to patients. Here, we investigated whether the neutrophil levels could predict COVID-19 complications. Methods: We performed a retrospective study of patients with COVID-19, admitted to the Cheikh Khalifa International University Hospital, Casablanca, Morocco. Laboratory test results collected upon admission and during hospitalization were analyzed based on clinical information. Results: Our study revealed that a rise in neutrophil "PNN" levels was associated with respiratory deterioration and intubation. They were positively correlated with the procalcitonin and C-reactive protein levels. Interestingly, PNN (polynuclear neutrophil) levels on day 5 proved to be a better predictor of intubation, acute respiratory distress syndrome (ARDS), and mortality than the initial PNN counts, C-reactive protein, or procalcitonin. Moreover, binary logistic regression with stratified PNN-day 5 data revealed that a PNN level on day 5 > 7.7 (109/L) was an independent risk factor for mortality and ARDS. Finally, the PNN levels on day 5 and proinflammatory cytokine IL-6 were positively correlated. Conclusions: Our data showed that neutrophilia proved to be an excellent predictor of complications and mortality during hospitalization and could be used to improve the management of patients with COVID-19.
Collapse
Affiliation(s)
- Khadija El Azhary
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Fadila Kouhen
- Laboratory of Neurosciences and Oncogenetics, Neurooncology and Oncogenetic Team, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco;
- Department of Radiotherapy, International University Hospital Cheikh Khalifa, Casablanca 82403, Morocco
| | - Jalila El Bakkouri
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Cheikh Khalifa International University Hospital, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco
| | - Hasna Chamlal
- Computer Science and Systems Laboratory (LIS), Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| |
Collapse
|
12
|
Kumar A, Yendamuri S, Ahmad F, Mukherjee PB, Kumar R, Manrai M, Muthukrishnan J, Dawra S. Inflammatory biomarkers and adverse outcome in COVID-19: Prelude for future viral pandemics. J Family Med Prim Care 2025; 14:720-728. [PMID: 40115551 PMCID: PMC11922368 DOI: 10.4103/jfmpc.jfmpc_1326_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 03/23/2025] Open
Abstract
Background Dysregulated inflammatory response plays a key role in the pathogenesis of COVID-19. The role of inflammatory markers to predict adverse clinical outcome is still controversial. The aim of this study was to analyze the association of inflammatory markers with disease outcomes independent of the effect of age and co-morbidities. Materials and Methods This is a retrospective analysis of COVID-19 patients admitted at a dedicated COVID center from July 2020 to Mar 2022. Clinical characteristics and inflammatory markers namely serum Ferritin levels, CRP, D-Dimer levels, serum LDH and IL-6 Levels were studied. The following outcome parameters were collected: disease severity at onset and outcome (discharge/death). Results 48.4% of the of 244 COVID-19 cases included had severe disease while 51.6% had moderate disease. Mean age was 61.3 ± 14.17 years and 71.7% were males. Primary Hypertension (48.4%) and Diabetes Mellitus (39.3%) were the most common co-morbidities. Increasing age, smoking, and alcohol consumption were associated with severe disease. CRP, D-dimer, and IL-6 were independent risk factors for disease severity while CRP, D dimer, LDH, Ferritin, and NLR (Neutrophil Lymphocyte ratio) were independent predictors of disease mortality. D-dimer was the most sensitive (95.8%) and specific (92.2%) marker to predict disease severity and serum LDH was the most sensitive (74.7%) to predict disease mortality at baseline. Conclusion Measurement of inflammatory markers might assist clinicians in predicting disease severity and prognosis of COVID-19. This may serve as a benchmark to understand the role of inflammatory markers in other diseases associated with dysregulated inflammatory response.
Collapse
Affiliation(s)
- Ankit Kumar
- Department of Medicine, Military Hospital, Shimla, Himachal Pradesh, India
| | - Sushma Yendamuri
- Department of Internal Medicine, Armed Forces Medical College, Pune, Maharashtra, India
| | - Faiz Ahmad
- Commandant, Military Hospital, Jammu, Jammu and Kashmir, India
| | - Partha B Mukherjee
- Department of Medicine, Command Hospital, Southern Command, Pune, Maharashtra, India
| | - Ravi Kumar
- Department of Internal Medicine, Armed Forces Medical College, Pune, Maharashtra, India
| | - Manish Manrai
- Department of Gastroenterology, Command Hospital, Central Command, Lucknow, Uttar Pradesh, India
| | - J Muthukrishnan
- Department of Internal Medicine, Armed Forces Medical College, Pune, Maharashtra, India
| | - Saurabh Dawra
- Department of Gastroenterology, Command Hospital, Southern Command, Pune, Maharashtra, India
| |
Collapse
|
13
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
14
|
Licata A, Seidita A, Como S, de Carlo G, Cammilleri M, Bonica R, Soresi M, Veronese N, Chianetta R, Citarrella R, Giannitrapani L, Barbagallo M. Herbal and Dietary Supplements as Adjunctive Treatment for Mild SARS-CoV-2 Infection in Italy. Nutrients 2025; 17:230. [PMID: 39861359 PMCID: PMC11767322 DOI: 10.3390/nu17020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
During the COVID-19 pandemic, several observational studies proved a certain efficacy of nutraceuticals, herbal products, and other dietary supplements as adjuvant therapies used alongside antiviral drugs. Although their use has not been widespread in Italy, according to preliminary evidence, many supplements with demonstrated immunomodulatory effects, such as vitamins C and D, herbal medicines and essential oils, might relieve the respiratory symptoms of COVID-19, since SARS-CoV-2 can activate inflammasome-mediated inflammatory signaling pathways. Other observational studies have shown that herbal treatments, such as Echinacea purpurea and ginseng, help alleviate respiratory symptoms and reduce serum levels of inflammatory cytokines, which are typically overexpressed in both adult and pediatric SARS-CoV-2 patients. Further, vitamins C and D can attenuate the immune response thanks to their cytokine suppression ability and to their known antimicrobial activity and potential to modulate T helper cell response. The strong immune response triggered by SARS-CoV-2 infection is responsible for the severity of the disease. Preliminary data have also shown that L-arginine, an endothelial-derived relaxing factor, is able to modulate endothelial damage, which appears to be one of the main targets of this systemic disease. Finally, some essential oils and their isolated compounds, such as eucalyptol, may be helpful in reducing many of the respiratory symptoms of COVID-19, although others, such as menthol, are not recommended, since it can lead to an undervaluation of the clinical status of a patient. In this narrative review, despite the lack of strong evidence in this field, we aimed to give an overview of the current available literature (mainly observational and cross-sectional studies) regarding herbal products and dietary supplements and their use in the treatment of mild disease from SARS-CoV-2 infection. Obviously, dietary supplements and herbal products do not constitute a standardized treatment for COVID-19 disease, but they could represent an adjunctive and useful treatment when used together with antivirals.
Collapse
Affiliation(s)
- Anna Licata
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Aurelio Seidita
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90146 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Silvia Como
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Gabriele de Carlo
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
| | - Marcella Cammilleri
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Bonica
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Maurizio Soresi
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Nicola Veronese
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Chianetta
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberto Citarrella
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Lydia Giannitrapani
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Mario Barbagallo
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| |
Collapse
|
15
|
Onodera Y, Ishiguro T, Uozumi R, Isono T, Nishida T, Kobayashi Y, Takaku Y. Predictive Factors for Invasive Mechanical Ventilation in Community-Acquired Pneumonia. Intern Med 2025:4727-24. [PMID: 39756876 DOI: 10.2169/internalmedicine.4727-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Objective Community-acquired pneumonia is an acute infectious disease with potentialy life-threatening consequences. Because invasive mechanical ventilation (IMV) requires the attention of many medical staff, early risk prediction at the time of admission is expected to lead to a predictable course of patient care and the appropriate allocation of medical resources. There are a limited number of reports on predictive factors for IMV, such as SMART-COP. Therefore, further studies are required. Patients and Methods We retrospectively reviewed cases of patients with community-acquired pneumonia other than COVID-19 admitted to our institution from 2002 to 2019. We performed competing risks analysis with the need for IMV from the day after admission as the outcome and used multivariable analysis to identify predictive factors of IMV from admission characteristics. Results Among 2,227 patients (mean age 67.3 years, 69.0% male), 39 patients required IMV on or after the day following admission. A multivariable analysis showed that predictive factors of IMV were respiratory rate >30 breaths/min [subdistribution hazard ratio (SHR), 5.53; 95% confidence interval (CI), 2.09 to 14.67; p=0.001], PaO2/FiO2 ratio <250 (SHR, 8.02; 95% CI, 2.78 to 23.13; p<0.001), and Legionella pneumonia (SHR, 4.87; 95% CI, 1.56 to 15.13; p=0.006). Conclusion This study revealed that among other factors including mainly vital signs, specific infection by a microorganism itself (Legionella in this study) was a predictive factor for the need of IMV.
Collapse
Affiliation(s)
- Yoko Onodera
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Takashi Ishiguro
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Ryuji Uozumi
- Department of Industrial Engineering and Economics, Tokyo Institute of Technology, Japan
| | - Taisuke Isono
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Takashi Nishida
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Yoichi Kobayashi
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| | - Yotaro Takaku
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Japan
| |
Collapse
|
16
|
Martínez-Martinez AB, Tristancho-Baró A, Garcia-Rodriguez B, Clavel-Millan M, Palacian MP, Milagro A, Rezusta A, Arbones-Mainar JM. Impact of Obesity-Associated SARS-CoV-2 Mutations on COVID-19 Severity and Clinical Outcomes. Viruses 2024; 17:38. [PMID: 39861827 PMCID: PMC11769164 DOI: 10.3390/v17010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
This study explores the relationship between specific SARS-CoV-2 mutations and obesity, focusing on how these mutations may influence COVID-19 severity and outcomes in high-BMI individuals. We analyzed 205 viral mutations from a cohort of 675 patients, examining the association of mutations with BMI, hospitalization, and mortality rates. Logistic regression models and statistical analyses were applied to assess the impact of significant mutations on clinical outcomes, including inflammatory markers and antibody levels. Our findings revealed three key mutations-C14599T, A20268G, and C313T-that were associated with elevated BMI. Notably, C14599T appeared to be protective against hospitalization, suggesting context-dependent effects, while A20268G was linked to a 50% increase in hospitalization risk and elevated antibody levels, potentially indicating an adaptive immune response. C313T showed a 428% increase in mortality risk, marking it as a possible poor-prognosis marker. Interestingly, all three mutations were synonymous, suggesting adaptive roles in obesity-driven environments despite not altering viral protein structures. These results emphasize the importance of studying mutations within the broader context of comorbidities, other mutations, and regional factors to enhance our understanding of SARS-CoV-2 adaptation in high-risk groups. Further validation in larger cohorts is necessary to confirm these associations and to assess their clinical significance.
Collapse
Affiliation(s)
- Ana B. Martínez-Martinez
- Facultad de Ciencias de la Salud, Universidad de Zaragoza, 50009 Zaragoza, Spain;
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
| | - Alexander Tristancho-Baró
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Beatriz Garcia-Rodriguez
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Department of Clinical Biochemistry, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Marina Clavel-Millan
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | - Maria Pilar Palacian
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Ana Milagro
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Antonio Rezusta
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.T.-B.); (M.P.P.); (A.M.)
| | - Jose M. Arbones-Mainar
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain; (B.G.-R.); (M.C.-M.); (A.R.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
17
|
Gammeri L, Sanfilippo S, Alessandrello C, Gangemi S, Minciullo PL. Mast Cells and Basophils in Major Viral Diseases: What Are the Correlations with SARS-CoV-2, Influenza A Viruses, HIV, and Dengue? Cells 2024; 13:2044. [PMID: 39768136 PMCID: PMC11674676 DOI: 10.3390/cells13242044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The SARS-CoV-2 pandemic has significantly impacted global health and has led the population and the scientific community to live in fear of a future pandemic. Based on viral infectious diseases, innate immunity cells such as mast cells and basophils play a fundamental role in the pathogenesis of viral diseases. Understanding these mechanisms could be essential to better study practical therapeutic approaches not only to COVID-19 but also to other viral infections widely spread worldwide, such as influenza A, HIV, and dengue. In this literature review, we want to study these concepts. Mast cells and basophils intervene as a bridge between innate and acquired immunity and seem to have a role in the damage mechanisms during infection and in the stimulation of humoral and cellular immunity. In some cases, these cells can act as reservoirs and favor the replication and spread of the virus in the body. Understanding these mechanisms can be useful not only in therapeutic but also in diagnostic and prognostic perspectives. The prospects of applying artificial intelligence and machine learning algorithms for the creation of very accurate diagnostic/prognostic tools are interesting.
Collapse
Affiliation(s)
| | | | | | | | - Paola Lucia Minciullo
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (L.G.); (S.S.); (C.A.); (S.G.)
| |
Collapse
|
18
|
Woodhouse EW, McClain MT, Woods CW. Harnessing the host response for precision infectious disease diagnosis. Clin Microbiol Rev 2024; 37:e0007824. [PMID: 39404266 PMCID: PMC11629621 DOI: 10.1128/cmr.00078-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024] Open
Abstract
SUMMARYDetection of the presence of infection and its etiology must be accurate and timely to facilitate appropriate antimicrobial use. Diagnostic strategies that rely solely on pathogen detection often are insufficient due to poor test characteristics, inability to differentiate colonization from infection, or protracted delay to result. Understanding the human response across different pathogens on a clinical and molecular level can provide more accurate, timely, and useful answers, especially in critical illness and diagnostic uncertainty. Improvements in understanding the human immune response including genomics, protein analysis, gene expression, and cellular morphology have led to rapid innovation of new host response-based diagnostic tests. This review describes the limitations of pathogen-focused technology and the benefits of examining the breadth of immune response to diagnose infection. It then explores biomarkers that have been studied for this purpose and scrutinizes the performance of host-based multianalyte testing. Currently cleared diagnostics and those in late-stage development are described in depth, with a focus on the purpose of testing and its utility for clinicians. Finally, it concludes by examining opportunities for further host response-derived diagnostic innovation.
Collapse
Affiliation(s)
- E. Wilbur Woodhouse
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Micah T. McClain
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Christopher W. Woods
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| |
Collapse
|
19
|
Allahverdiyeva S, Geyer CE, Veth J, de Vries LM, de Taeye SW, van Gils MJ, den Dunnen J, Chen H. Testosterone and estradiol reduce inflammation of human macrophages induced by anti-SARS-CoV-2 IgG. Eur J Immunol 2024; 54:e2451226. [PMID: 39246165 PMCID: PMC11628899 DOI: 10.1002/eji.202451226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
COVID-19, the disease caused by SARS-CoV-2, particularly causes severe inflammatory disease in elderly, obese, and male patients. Since both aging and obesity are associated with decreased testosterone and estradiol expression, we hypothesized that decreased hormone levels contribute to excessive inflammation in the context of COVID-19. Previously, we and others have shown that hyperinflammation in severe COVID-19 patients is induced by the production of pathogenic anti-spike IgG antibodies that activate alveolar macrophages. Therefore, we developed an in vitro assay in which we stimulated human macrophages with viral stimuli, anti-spike IgG immune complexes, and different sex hormones. Treatment with levels of testosterone reflecting young adults led to a significant reduction in TNF and IFN-γ production by human macrophages. In addition, estradiol significantly attenuated the production of a very broad panel of cytokines, including TNF, IL-1β, IL-6, IL-10, and IFN-γ. Both testosterone and estradiol reduced the expression of Fc gamma receptors IIa and III, the two main receptors responsible for anti-spike IgG-induced inflammation. Combined, these findings indicate that sex hormones reduce the inflammatory response of human alveolar macrophages to specific COVID-19-associated stimuli, thereby providing a potential immunological mechanism for the development of severe COVID-19 in both older male and female patients.
Collapse
Affiliation(s)
- Sona Allahverdiyeva
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Chiara E. Geyer
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jennifer Veth
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Laura M. de Vries
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Steven W. de Taeye
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Marit J. van Gils
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Hung‐Jen Chen
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| |
Collapse
|
20
|
Zhang J, Kim MH, Lee S, Park S. Integration of nanobiosensors into organ-on-chip systems for monitoring viral infections. NANO CONVERGENCE 2024; 11:47. [PMID: 39589620 PMCID: PMC11599699 DOI: 10.1186/s40580-024-00455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
The integration of nanobiosensors into organ-on-chip (OoC) models offers a promising advancement in the study of viral infections and therapeutic development. Conventional research methods for studying viral infection, such as two-dimensional cell cultures and animal models, face challenges in replicating the complex and dynamic nature of human tissues. In contrast, OoC systems provide more accurate, physiologically relevant models for investigating viral infections, disease mechanisms, and host responses. Nanobiosensors, with their miniaturized designs and enhanced sensitivity, enable real-time, continuous, in situ monitoring of key biomarkers, such as cytokines and proteins within these systems. This review highlights the need for integrating nanobiosensors into OoC systems to advance virological research and improve therapeutic outcomes. Although there is extensive literature on biosensors for viral infection detection and OoC models for replicating infections, real integration of biosensors into OoCs for continuous monitoring remains unachieved. We discuss the advantages of nanobiosensor integration for real-time tracking of critical biomarkers within OoC models, key biosensor technologies, and current OoC systems relevant to viral infection studies. Additionally, we address the main technical challenges and propose solutions for successful integration. This review aims to guide the development of biosensor-integrated OoCs, paving the way for precise diagnostics and personalized treatments in virological research.
Collapse
Affiliation(s)
- Jiande Zhang
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| |
Collapse
|
21
|
Mendes LT, Gama-Almeida MC, Reis DL, Silva ACPE, Neris RLS, Galliez RM, Castiñeiras TMPP, on behalf of the UFRJ COVID-19 Working Group, Ludwig C, Valente AP, Costa dos Santos Junior G, El-Bacha T, Assunção-Miranda I. Longitudinal 1H NMR-Based Metabolomics in Saliva Unveils Signatures of Transition from Acute to Post-Acute Phase of SARS-CoV-2 Infection. Viruses 2024; 16:1769. [PMID: 39599883 PMCID: PMC11598993 DOI: 10.3390/v16111769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
COVID-19 can range from a mild to severe acute respiratory syndrome and also could result in multisystemic damage. Additionally, many people develop post-acute symptoms associated with immune and metabolic disturbances in response to viral infection, requiring longitudinal and multisystem studies to understand the complexity of COVID-19 pathophysiology. Here, we conducted a 1H Nuclear Magnetic Resonance metabolomics in saliva of symptomatic subjects presenting mild and moderate respiratory symptoms to investigate prospective changes in the metabolism induced after acute-phase SARS-CoV-2 infection. Saliva from 119 donors presenting non-COVID and COVID-19 respiratory symptoms were evaluated in the acute phase (T1) and the post-acute phase (T2). We found two clusters of metabolite fluctuation in the COVID-19 group. Cluster 1, metabolites such as glucose, (CH3)3 choline-related metabolites, 2-hydroxybutyrate, BCAA, and taurine increased in T2 relative to T1, and in cluster 2, acetate, creatine/creatinine, phenylalanine, histidine, and lysine decreased in T2 relative to T1. Metabolic fluctuations in the COVID-19 group were associated with overweight/obesity, vaccination status, higher viral load, and viral clearance of the respiratory tract. Our data unveil metabolic signatures associated with the transition to the post-acute phase of SARS-CoV-2 infection that may reflect tissue damage, inflammatory process, and activation of tissue repair cascade. Thus, they contribute to describing alterations in host metabolism that may be associated with prolonged symptoms of COVID-19.
Collapse
Affiliation(s)
- Luiza Tomé Mendes
- LaRIV-Laboratory of Cellular Response to Viral Infections, Instituto de Microbiologia Paulo de Góes, Departamento de Virologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.T.M.); (A.C.P.e.S.); (R.L.S.N.)
| | - Marcos C. Gama-Almeida
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.C.G.-A.); (D.L.R.)
| | - Desirée Lopes Reis
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.C.G.-A.); (D.L.R.)
| | - Ana Carolina Pires e Silva
- LaRIV-Laboratory of Cellular Response to Viral Infections, Instituto de Microbiologia Paulo de Góes, Departamento de Virologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.T.M.); (A.C.P.e.S.); (R.L.S.N.)
| | - Rômulo Leão Silva Neris
- LaRIV-Laboratory of Cellular Response to Viral Infections, Instituto de Microbiologia Paulo de Góes, Departamento de Virologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.T.M.); (A.C.P.e.S.); (R.L.S.N.)
| | - Rafael Mello Galliez
- Núcleo de Enfrentamento e Estudos de Doenças Infecciosas Emergentes e Reemergentes (NEEDIER), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-599, Brazil; (R.M.G.); (T.M.P.P.C.)
| | - Terezinha Marta Pereira Pinto Castiñeiras
- Núcleo de Enfrentamento e Estudos de Doenças Infecciosas Emergentes e Reemergentes (NEEDIER), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-599, Brazil; (R.M.G.); (T.M.P.P.C.)
| | | | - Christian Ludwig
- Department of Metabolism and Systems Science, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK;
| | - Ana Paula Valente
- National Center for Nuclear Magnetic Resonance—Jiri Jonas, Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| | - Gilson Costa dos Santos Junior
- LabMet-Laboratory of Metabolomics, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Department of Genetics, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil;
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.C.G.-A.); (D.L.R.)
| | - Iranaia Assunção-Miranda
- LaRIV-Laboratory of Cellular Response to Viral Infections, Instituto de Microbiologia Paulo de Góes, Departamento de Virologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.T.M.); (A.C.P.e.S.); (R.L.S.N.)
| |
Collapse
|
22
|
Gupta P, Dev K, Kaur G. Phytoconstituents as modulator of inflammatory pathways for COVID-19: A comprehensive review and recommendations. Phytother Res 2024; 38:5389-5416. [PMID: 39246209 DOI: 10.1002/ptr.8302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 09/10/2024]
Abstract
SARS-CoV-2 infection causes disruptions in inflammatory pathways, which fundamentally contribute to COVID-19 pathophysiology. The present review critically evaluates the gaps in scientific literature and presents the current status regarding the inflammatory signaling pathways in COVID-19. We propose that phytoconstituents can be used to treat COVID-19 associated inflammation, several already formulated in traditional medications. For this purpose, extensive literature analysis was conducted in the PubMed database to collect relevant in vitro, in vivo, and human patient studies where inflammation pathways were shown to be upregulated in COVID-19. Parallelly, scientific literature was screened for phytoconstituents with known cellular mechanisms implicated for inflammation or COVID-19 associated inflammation. Studies with insufficient evidence on cellular pathways for autophagy and mitophagy were considered out of scope and excluded from the study. The final analysis was visualized in figures and evaluated for accuracy. Our findings demonstrate the frequent participation of NF-κB, a transcription factor, in inflammatory signaling pathways linked to COVID-19. Moreover, the MAPK signaling pathway is also implicated in producing inflammatory molecules. Furthermore, it was also analyzed that the phytoconstituents with flavonoid and phenolic backbones could inhibit either the TLR4 receptor or its consecutive signaling molecules, thereby, decreasing NF-κB activity and suppressing cytokine production. Although, allopathy has treated the early phase of COVID-19, anti-inflammatory phytoconstituents and existing ayurvedic formulations may act on the COVID-19 associated inflammatory pathways and provide an additional treatment strategy. Therefore, we recommend the usage of flavonoids and phenolic phytoconstituents for the treatment of inflammation associated with COVID-19 infection and similar viral ailments.
Collapse
Affiliation(s)
- Pragati Gupta
- School of Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Kamal Dev
- School of Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
- Department of Pharmacology & Toxicology, Wright State University, Dayton, Ohio, USA
| | - Gurjot Kaur
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
- National Center cum Department of Human Genome Research Center and Studies, Panjab University, Chandigarh, Punjab, India
| |
Collapse
|
23
|
Jalal MM, Algamdi MM, Alkayyal AA, Altayar MA, Mouminah AS, Alamrani AJ, Althaqafi NA, Alamrani RA, Alomrani WS, Alemrani YA, Alhelali M, Elfaki I, Mir R. Association of iron deficiency anaemia with the hospitalization and mortality rate of patients with COVID‑19. MEDICINE INTERNATIONAL 2024; 4:69. [PMID: 39301327 PMCID: PMC11411605 DOI: 10.3892/mi.2024.193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) illness led to the coronavirus disease 2019 (COVID-19) pandemic, which has caused enormous health and financial losses, as well as challenges to global health. Iron deficiency anaemia (IDA) has been linked to adverse outcomes in patients infected with SARS-COV-2. The present study aimed to assess the association between IDA and the severity of COVID-19 in hospitalized patients. For this purpose, a retrospective data analysis of 100 patients with COVID-19 was conducted. Data of patients hospitalized with SARS-COV-2 infection confirmed by RT-PCR were collected between June, 2021 and March, 2022. The collected data included patient demographics, comorbidities, clinical signs, symptoms and IDA medical laboratory findings, including complete blood count and iron profiles. The results revealed that patients with COVID-19 admitted to the isolation unit represented 61.0% of the study sample, whereas 39.0% were admitted to the intensive care unit (ICU). No patients had stage I IDA, whereas 4 patients (4%) had stage II IDA. Furthermore, 19 patients (19.0%) had stage III IDA. A significantly higher proportion of patients with IDA (69.6%) were admitted to the ICU compared with those without IDA (29.9%, P<0.001). Additionally, patients with IDA had a higher proportion of a history of stroke compared with those without IDA (17.4 vs. 2.6%, respectively, P=0.024). The most common comorbidities identified were hypertension (29%), diabetes (23%) and heart problems (17%). On the whole, the present study demonstrates significant associations between IDA and a longer hospitalization period. A greater incidence of complications was observed in the hospitalized patients who were SARS-COV-2-positive. Although further studies with larger sample sizes are required to confirm these findings, the results presented herein may provide insight for physicians as regards the prevention and treatment of patients with IDA who are infected with coronavirus.
Collapse
Affiliation(s)
- Mohammed M Jalal
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Maaidah M Algamdi
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Malik A Altayar
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Amr S Mouminah
- Neuroscience Center, King Abdullah Medical Complex, Jeddah 23816, Kingdom of Saudi Arabia
| | - Ahlam Jumaa Alamrani
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Nouf Abdulaziz Althaqafi
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Reem Ali Alamrani
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Wjdan Salem Alomrani
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Yasmin Attallah Alemrani
- Faculty of Nursing, Community and Mental Health Nursing Department, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Marwan Alhelali
- Department of Statistics, Faculty of Science, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| |
Collapse
|
24
|
Weckman AM, Guagliardo SAJ, Crowley VM, Moro L, Piubelli C, Ursini T, van Ierssel SH, Gobbi FG, Emetulu H, Rizwan A, Angelo KM, Licitra C, Connor BA, Barkati S, Ngai M, Zhong K, Huits R, Hamer DH, Libman M, Kain KC. Host Response Markers of Inflammation and Endothelial Activation Associated with COVID-19 Severity and Mortality: A GeoSentinel Prospective Observational Cohort. Viruses 2024; 16:1615. [PMID: 39459948 PMCID: PMC11512287 DOI: 10.3390/v16101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The effect of the COVID-19 pandemic on healthcare systems emphasized the need for rapid and effective triage tools to identify patients at risk of severe or fatal infection. Measuring host response markers of inflammation and endothelial activation at clinical presentation may help to inform appropriate triage and care practices in patients with SARS-CoV-2 infection. METHODS We enrolled patients with COVID-19 across five GeoSentinel clinical sites (in Italy, Belgium, Canada, and the United States) from September 2020 to December 2021, and analyzed the association of plasma markers, including soluble urokinase-type plasminogen activator receptor (suPAR), soluble tumor necrosis factor receptor-1 (sTREM-1), interleukin-6 (IL-6), interleukin-8 (IL-8), complement component C5a (C5a), von Willebrand factor (VWF-a2), and interleukin-1 receptor antagonist (IL-1Ra), with 28-day (D28) mortality and 7-day (D7) severity (discharged, hospitalized on ward, or died/admitted to the ICU). RESULTS Of 193 patients, 8.9% (16 of 180) died by D28. Higher concentrations of suPAR were associated with increased odds of mortality at D28 and severity at D7 in univariable and multivariable regression models. The biomarkers sTREM-1 and IL-1Ra showed bivariate associations with mortality at D28 and severity at D7. IL-6, VWF, C5a, and IL-8 were not as indicative of progression to severe disease or death. Conclusions: Our findings confirm previous studies' assertions that point-of-care tests for suPAR and sTREM-1 could facilitate the triage of patients with SARS-CoV-2 infection, which may help guide hospital resource allocation.
Collapse
Affiliation(s)
- Andrea M. Weckman
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | | | - Valerie M. Crowley
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Lucia Moro
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Chiara Piubelli
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Tamara Ursini
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Sabrina H. van Ierssel
- Department of Internal Medicine, Antwerp University Hospital (UZA), 2650 Antwerp, Belgium
| | - Federico G. Gobbi
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Hannah Emetulu
- International Society of Travel Medicine, Atlanta, GA 30338, USA
| | - Aisha Rizwan
- International Society of Travel Medicine, Atlanta, GA 30338, USA
| | - Kristina M. Angelo
- Division of Global Migration and Quarantine, Travelers’ Health Branch, Atlanta, GA 30322, USA
| | - Carmelo Licitra
- Orlando Health Travel Medicine and Infectious Disease, Orlando, FL 34761, USA
| | - Bradley A. Connor
- Weill Cornell Medical College and the New York Center for Travel and Tropical Medicine, New York, NY 10022, USA
| | - Sapha Barkati
- J.D. MacLean Centre for Tropical Diseases, McGill University, Montreal, QC H3A 0G4, Canada
| | - Michelle Ngai
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Kathleen Zhong
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Ralph Huits
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
- Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium
| | - Davidson H. Hamer
- Section of Infectious Diseases, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Center on Emerging Infectious Diseases, Boston University, Boston, MA 02118, USA
- Department of Global Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael Libman
- J.D. MacLean Centre for Tropical Diseases, McGill University, Montreal, QC H3A 0G4, Canada
| | - Kevin C. Kain
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
- Division of Infectious Diseases, Department of Medicine, MaRS Centre, TMDT, University of Toronto, 10th Floor 10-351, Toronto, QC M5G 1L7, Canada
| |
Collapse
|
25
|
Saied YM, Abou Warda AE, Allam RM, Syed W, Basil A. Al-Rawi M, Iqbal A, Elgendy MO, M. El-Sabaa R, Hassan A. The Impact of Infliximab on Hyperinflammation State in Hospitalized COVID-19 Patients: A Retrospective Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1670. [PMID: 39459457 PMCID: PMC11509666 DOI: 10.3390/medicina60101670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/21/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Elevated levels of pro-inflammatory cytokines have been linked to increased mortality in COVID-19 patients. Infliximab, a tumor necrosis factor inhibitor, has been reported to improve outcomes in COVID-19 patients by targeting the hyperinflammatory response. Our objective was to evaluate the effectiveness of incorporating Infliximab into standard care guidelines for the management of COVID-19. Materials and Methods: A retrospective analysis was conducted on 111 participants who were moderate to severe COVID-19 patients admitted to the hospital. Among them, 74 individuals received solely standard treatment, while 37 received standard therapy plus Infliximab. The primary outcomes of the study centered around the changes in laboratory test parameters. The secondary clinical findings included clinical recovery defined as improvement in patient oxygenation, time till recovery, and assessing necessity for ICU admission, and mortality rates. Results: There was no statistical difference observed in the inflammatory markers including, LDH, Ferritin, CRP, neutrophil to lymphocyte ratio (NLR), and P/F ratio between both groups and in the clinical outcomes including clinical recovery (p = 1.0), time to improvement (p = 0.436), and mortality rate (p = 0.601). However, there was a significant increase in secondary infection (45.9%, 20.3%; p = 0.005), and in liver enzymes, ALT (79.5, 50.0 IU/L; p = 0.02) and AST (57.5, 38.0 IU/L; p = 0.019) in the Infliximab group and the standard care group, respectively. Conclusions: Infliximab therapy did not demonstrate significant benefits compared to standard of care in moderate to severe hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Yasmine M. Saied
- Microbiology and Immunology Postgraduate Program, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ahmed Essam Abou Warda
- Clinical Pharmacy Department, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt;
| | - Rasha Mahmoud Allam
- Cancer Epidemiology and Biostatistics, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Wajid Syed
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mahmood Basil A. Al-Rawi
- Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Ayesha Iqbal
- Department of Pharmacy Practice and Policy, University Park Campus, University of Nottingham, Nottingham NG7 2QL, UK;
- Office of Lifelong Learning and the Physician Learning Program, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G1C9, Canada
| | - Marwa O. Elgendy
- Department of Clinical Pharmacy, Beni-Suef University Hospitals, Beni-Suef University, Beni-Suef 62521, Egypt;
- Department of Clinical Pharmacy, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef 62764, Egypt
| | - Ramy M. El-Sabaa
- Clinical Pharmacy Department, Faculty of Pharmacy, Menoufia University, Menoufia 32511, Egypt;
| | - Ahmed Hassan
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt;
| |
Collapse
|
26
|
Yuan J, Zhang W, Qie B, Xie Y, Zhu B, Chen C, Qiu W, Sun H, Zhao B, Long Y. Utilizing press needle acupuncture to treat mild-to-moderate COVID-19: A single-blind, randomized controlled trial. Medicine (Baltimore) 2024; 103:e39810. [PMID: 39465704 PMCID: PMC11460845 DOI: 10.1097/md.0000000000039810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND In China, acupuncture has been employed as an adjunctive therapy for coronavirus disease 2019 (COVID-19). Press needle acupuncture is a special type of acupuncture that provides prolonged stimulation to acupuncture points and simultaneously reduces the pain associated with traditional acupuncture. This study assessed the effectiveness of integrating press needles alongside pharmacologic treatment in patients with mild-to-moderate COVID-19. METHODS Patients hospitalized with mild-to-moderate COVID-19 symptoms between December 2022 and January 2023 were included in the study. The enrolled patients were randomly assigned to receive pharmacologic treatment alone (control group) or both pharmacologic treatment and press needle acupuncture (intervention group). Patients were evaluated for clinical outcomes, including symptom scores, deterioration rates, fever durations, and nucleic acid test results. The patients' complete blood count and C-reactive protein levels were also analyzed using venous blood samples both before and after treatment. RESULTS Both groups exhibited a reduction in clinical symptom scores, but symptoms regressed faster in the intervention group. Nucleic acid test negativity was achieved faster in the intervention group than in the control group. The intervention group also had a lower deterioration rate. Furthermore, the increase in the lymphocyte count and decrease in C-reactive protein levels following treatment were more pronounced in the intervention group than in the control group. CONCLUSION This study suggests that utilizing press needle acupuncture as an adjunct to pharmacologic treatment can be effective in patients with mild-to-moderate COVID-19 symptoms.
Collapse
Affiliation(s)
- Jiawei Yuan
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weizhen Zhang
- NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Beibei Qie
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuhua Xie
- Taihe Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Binbin Zhu
- Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong, China
| | - Cheng Chen
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenwei Qiu
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huanwen Sun
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Zhao
- Southern Medical University, Guangzhou, Guangdong, China
| | - Yaqiu Long
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Pejler G, Zhao XO, Fagerström E, Paivandy A. Blockade of endolysosomal acidification suppresses TLR3-mediated proinflammatory signaling in airway epithelial cells. J Allergy Clin Immunol 2024; 154:940-951. [PMID: 38906273 DOI: 10.1016/j.jaci.2024.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Endolysosomal compartments are acidic and contain low pH-dependent proteases, and these conditions are exploited by respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus, for escaping into the cytosol. Moreover, endolysosomes contain various pattern recognition receptors (PRRs), which respond to virus-derived pathogen-associated molecular patterns (PAMPs) by production of proinflammatory cytokines/chemokines. However, excessive proinflammatory responses can lead to a potentially lethal cytokine storm. OBJECTIVES Here we investigated the endosomal PRR expression profile in primary human small airway epithelial cells (HSAECs), and whether blockade of endolysosomal acidification affects their cytokine/chemokine production after challenge with virus-derived stimulants. METHODS HSAECs were exposed to stimulants mimicking virus-derived PAMPs, either in the absence or presence of compounds causing blockade of endolysosomal acidification, followed by measurement of cytokine expression and release. RESULTS We show that Toll-like receptor 3 (TLR3) is the major endosomal PRR expressed by HSAECs, and that TLR3 expression is strongly induced by TLR3 agonists, but not by a range of other PRR agonists. We also demonstrate that TLR3 engagement with its agonists elicits a robust proinflammatory cytokine/chemokine response, which is profoundly suppressed through blockade of endolysosomal acidification, by bafilomycin A1, monensin, or niclosamide. Using TLR3 reporter cells, it was confirmed that TLR3 signaling is strongly induced by Poly(I:C) and that blockade of endolysosomal acidification efficiently blocked TLR3 signaling. Finally, we show that blockade of endolysosomal acidification causes a reduction in the levels of TLR3 mRNA and protein. CONCLUSIONS These findings show that blockade of endolysosomal acidification suppresses TLR3-dependent cytokine and chemokine production in HSAECs.
Collapse
Affiliation(s)
- Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Xinran O Zhao
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ella Fagerström
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Aida Paivandy
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
28
|
Singh K, Rubenstein K, Callier V, Shaw-Saliba K, Rupert A, Dewar R, Laverdure S, Highbarger H, Lallemand P, Huang ML, Jerome KR, Sampoleo R, Mills MG, Greninger AL, Juneja K, Porter D, Benson CA, Dempsey W, El Sahly HM, Focht C, Jilg N, Paules CI, Rapaka RR, Uyeki TM, Clifford Lane H, Beigel J, Dodd LE. SARS-CoV-2 RNA and Nucleocapsid Antigen Are Blood Biomarkers Associated With Severe Disease Outcomes That Improve in Response to Remdesivir. J Infect Dis 2024; 230:624-634. [PMID: 38657001 PMCID: PMC11420797 DOI: 10.1093/infdis/jiae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Although antivirals remain important for the treatment COVID-19, methods to assess treatment efficacy are lacking. Here, we investigated the impact of remdesivir on viral dynamics and their contribution to understanding antiviral efficacy in the multicenter Adaptive COVID-19 Treatment Trial 1, which randomized patients to remdesivir or placebo. METHODS Longitudinal specimens collected during hospitalization from a substudy of 642 patients with COVID-19 were measured for viral RNA (upper respiratory tract and plasma), viral nucleocapsid antigen (serum), and host immunologic markers. Associations with clinical outcomes and response to therapy were assessed. RESULTS Higher baseline plasma viral loads were associated with poorer clinical outcomes, and decreases in viral RNA and antigen in blood but not the upper respiratory tract correlated with enhanced benefit from remdesivir. The treatment effect of remdesivir was most pronounced in patients with elevated baseline nucleocapsid antigen levels: the recovery rate ratio was 1.95 (95% CI, 1.40-2.71) for levels >245 pg/mL vs 1.04 (95% CI, .76-1.42) for levels <245 pg/mL. Remdesivir also accelerated the rate of viral RNA and antigen clearance in blood, and patients whose blood levels decreased were more likely to recover and survive. CONCLUSIONS Reductions in SARS-CoV-2 RNA and antigen levels in blood correlated with clinical benefit from antiviral therapy. CLINICAL TRIAL REGISTRATION NCT04280705 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Kanal Singh
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Kevin Rubenstein
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | - Viviane Callier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | | | - Adam Rupert
- National Laboratory for Cancer Research, Frederick, Maryland
| | - Robin Dewar
- National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Keith R Jerome
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Reigran Sampoleo
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Margaret G Mills
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | | | | | | | - Walla Dempsey
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Nikolaus Jilg
- Massachusetts General Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Catharine I Paules
- Division of Infectious Diseases, Milton S. Hershey Medical Center, Penn State Health, Hershey, Pennsylvania
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, School of Medicine, University of Maryland, Baltimore
| | - Timothy M Uyeki
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - John Beigel
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Lori E Dodd
- National Institute of Allergy and Infectious Diseases, Bethesda
| |
Collapse
|
29
|
Da Silva Filho J, Herder V, Gibbins MP, Dos Reis MF, Melo GC, Haley MJ, Judice CC, Val FFA, Borba M, Tavella TA, de Sousa Sampaio V, Attipa C, McMonagle F, Wright D, de Lacerda MVG, Costa FTM, Couper KN, Marcelo Monteiro W, de Lima Ferreira LC, Moxon CA, Palmarini M, Marti M. A spatially resolved single-cell lung atlas integrated with clinical and blood signatures distinguishes COVID-19 disease trajectories. Sci Transl Med 2024; 16:eadk9149. [PMID: 39259811 DOI: 10.1126/scitranslmed.adk9149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/15/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
COVID-19 is characterized by a broad range of symptoms and disease trajectories. Understanding the correlation between clinical biomarkers and lung pathology during acute COVID-19 is necessary to understand its diverse pathogenesis and inform more effective treatments. Here, we present an integrated analysis of longitudinal clinical parameters, peripheral blood markers, and lung pathology in 142 Brazilian patients hospitalized with COVID-19. We identified core clinical and peripheral blood signatures differentiating disease progression between patients who recovered from severe disease compared with those who succumbed to the disease. Signatures were heterogeneous among fatal cases yet clustered into two patient groups: "early death" (<15 days until death) and "late death" (>15 days). Progression to early death was characterized systemically and in lung histopathological samples by rapid endothelial and myeloid activation and the presence of thrombi associated with SARS-CoV-2+ macrophages. In contrast, progression to late death was associated with fibrosis, apoptosis, and SARS-CoV-2+ epithelial cells in postmortem lung tissue. In late death cases, cytotoxicity, interferon, and T helper 17 (TH17) signatures were only detectable in the peripheral blood after 2 weeks of hospitalization. Progression to recovery was associated with higher lymphocyte counts, TH2 responses, and anti-inflammatory-mediated responses. By integrating antemortem longitudinal blood signatures and spatial single-cell lung signatures from postmortem lung samples, we defined clinical parameters that could be used to help predict COVID-19 outcomes.
Collapse
Affiliation(s)
- João Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Federal University of Amazonas, Manaus, Brazil
- Amazonas Oncology Control Center Foundation, Manaus, Brazil
| | | | - Michael J Haley
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Carla Cristina Judice
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Fernando Fonseca Almeida Val
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Mayla Borba
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Delphina Rinaldi Abdel Aziz Emergency Hospital (HPSDRA), Manaus, Brazil
| | - Tatyana Almeida Tavella
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- INSERM U1016, CNRS UMR8104, University of Paris Cité, Institut Cochin, Paris, France
| | | | - Charalampos Attipa
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona McMonagle
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Glasgow Imaging Facility/School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Derek Wright
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin N Couper
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Christopher Alan Moxon
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- (C.A.M.)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- (M.P.)
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
- (M.M.)
| |
Collapse
|
30
|
Gao J, Zhang C, Wheelock ÅM, Xin S, Cai H, Xu L, Wang XJ. Immunomics in one health: understanding the human, animal, and environmental aspects of COVID-19. Front Immunol 2024; 15:1450380. [PMID: 39295871 PMCID: PMC11408184 DOI: 10.3389/fimmu.2024.1450380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 09/21/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic underscores the critical need to integrate immunomics within the One Health framework to effectively address zoonotic diseases across humans, animals, and environments. Employing advanced high-throughput technologies, this interdisciplinary approach reveals the complex immunological interactions among these systems, enhancing our understanding of immune responses and yielding vital insights into the mechanisms that influence viral spread and host susceptibility. Significant advancements in immunomics have accelerated vaccine development, improved viral mutation tracking, and broadened our comprehension of immune pathways in zoonotic transmissions. This review highlights the role of animals, not merely as carriers or reservoirs, but as essential elements of ecological networks that profoundly influence viral epidemiology. Furthermore, we explore how environmental factors shape immune response patterns across species, influencing viral persistence and spillover risks. Moreover, case studies demonstrating the integration of immunogenomic data within the One Health framework for COVID-19 are discussed, outlining its implications for future research. However, linking humans, animals, and the environment through immunogenomics remains challenging, including the complex management of vast amounts of data and issues of scalability. Despite challenges, integrating immunomics data within the One Health framework significantly enhances our strategies and responses to zoonotic diseases and pandemic threats, marking a crucial direction for future public health breakthroughs.
Collapse
Affiliation(s)
- Jing Gao
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Pulmonary Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Chutian Zhang
- College of Natural Resources and Environment, Northwest Agriculture and Forestry University, Yangling, China
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Siming Xin
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Hui Cai
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Lei Xu
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Xiao-Jun Wang
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
31
|
Karakike E, Metallidis S, Poulakou G, Kosmidou M, Gatselis NK, Petrakis V, Rovina N, Gkeka E, Sympardi S, Papanikolaou I, Koutsodimitropoulos I, Tzavara V, Adamis G, Tsiakos K, Koulouras V, Mouloudi E, Antoniadou E, Vlachogianni G, Anisoglou S, Markou N, Koutsoukou A, Panagopoulos P, Milionis H, Dalekos GN, Kyprianou M, Giamarellos-Bourboulis EJ. Clinical Phenotyping for Prognosis and Immunotherapy Guidance in Bacterial Sepsis and COVID-19. Crit Care Explor 2024; 6:e1153. [PMID: 39263383 PMCID: PMC11390041 DOI: 10.1097/cce.0000000000001153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
OBJECTIVES It is suggested that sepsis may be classified into four clinical phenotypes, using an algorithm employing 29 admission parameters. We applied a simplified phenotyping algorithm among patients with bacterial sepsis and severe COVID-19 and assessed characteristics and outcomes of the derived phenotypes. DESIGN Retrospective analysis of data from prospective clinical studies. SETTING Greek ICUs and Internal Medicine departments. PATIENTS AND INTERVENTIONS We analyzed 1498 patients, 620 with bacterial sepsis and 878 with severe COVID-19. We implemented a six-parameter algorithm (creatinine, lactate, aspartate transaminase, bilirubin, C-reactive protein, and international normalized ratio) to classify patients with bacterial sepsis intro previously defined phenotypes. Patients with severe COVID-19, included in two open-label immunotherapy trials were subsequently classified. Heterogeneity of treatment effect of anakinra was assessed. The primary outcome was 28-day mortality. MEASUREMENTS AND MAIN RESULTS The algorithm validated the presence of the four phenotypes across the cohort of bacterial sepsis and the individual studies included in this cohort. Phenotype α represented younger patients with low risk of death, β was associated with high comorbidity burden, and δ with the highest mortality. Phenotype assignment was independently associated with outcome, even after adjustment for Charlson Comorbidity Index. Phenotype distribution and outcomes in severe COVID-19 followed a similar pattern. CONCLUSIONS A simplified algorithm successfully identified previously derived phenotypes of bacterial sepsis, which were predictive of outcome. This classification may apply to patients with severe COVID-19 with prognostic implications.
Collapse
Affiliation(s)
- Eleni Karakike
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Simeon Metallidis
- 1st Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Garyfallia Poulakou
- 3rd Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Maria Kosmidou
- 1st Department of Internal Medicine, University General Hospital of Ioannina, Ioannina, Greece
| | - Nikolaos K. Gatselis
- Department of Internal Medicine, Larissa University General Hospital, University of Thessaly, Larissa, Greece
| | - Vasileios Petrakis
- 2nd Department of Internal Medicine, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Nikoletta Rovina
- 1st Department of Pulmonary Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Eleni Gkeka
- Intensive Care Unit, AHEPA University General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Styliani Sympardi
- 1st Department of Internal Medicine, Elefsis General Hospital Thriassio, Elefsis, Greece
| | - Ilias Papanikolaou
- Department of Pulmonary Medicine, Kerkyra General Hospital, Corfu, Greece
| | | | - Vasiliki Tzavara
- 1st Department of Internal Medicine, “Korgialeneio-Benakeio” Athens General Hospital, Athens, Greece
| | - Georgios Adamis
- 1st Department of Internal Medicine, “G.Gennimatas” Athens General Hospital, Athens, Greece
| | - Konstantinos Tsiakos
- 3rd Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Vasilios Koulouras
- Department of Critical Care Medicine, University General Hospital of Ioannina, Ioannina, Greece
| | - Eleni Mouloudi
- Intensive Care Unit, “Ippokrateion” General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Eleni Antoniadou
- Intensive Care Unit, “G.Gennimatas” General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Gykeria Vlachogianni
- Intensive Care Unit, “Aghios Dimitrios” General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Souzana Anisoglou
- Intensive Care Unit, “Theageneion” General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Markou
- Intensive Care Unit of Latseion Burn Center, Elefsis General Hospital Thriassio, Elefsis, Greece
| | - Antonia Koutsoukou
- 1st Department of Pulmonary Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Periklis Panagopoulos
- 2nd Department of Internal Medicine, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Haralampos Milionis
- 1st Department of Internal Medicine, University General Hospital of Ioannina, Ioannina, Greece
| | - George N. Dalekos
- Department of Internal Medicine, Larissa University General Hospital, University of Thessaly, Larissa, Greece
| | | | - Evangelos J. Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| |
Collapse
|
32
|
Beurton A, Kooistra EJ, De Jong A, Schiffl H, Jourdain M, Garcia B, Vimpère D, Jaber S, Pickkers P, Papazian L. Specific and Non-specific Aspects and Future Challenges of ICU Care Among COVID-19 Patients with Obesity: A Narrative Review. Curr Obes Rep 2024; 13:545-563. [PMID: 38573465 DOI: 10.1007/s13679-024-00562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW Since the end of 2019, the coronavirus disease 2019 (COVID-19) pandemic has infected nearly 800 million people and caused almost seven million deaths. Obesity was quickly identified as a risk factor for severe COVID-19, ICU admission, acute respiratory distress syndrome, organ support including mechanical ventilation and prolonged length of stay. The relationship among obesity; COVID-19; and respiratory, thrombotic, and renal complications upon admission to the ICU is unclear. RECENT FINDINGS The predominant effect of a hyperinflammatory status or a cytokine storm has been suggested in patients with obesity, but more recent studies have challenged this hypothesis. Numerous studies have also shown increased mortality among critically ill patients with obesity and COVID-19, casting doubt on the obesity paradox, with survival advantages with overweight and mild obesity being reported in other ICU syndromes. Finally, it is now clear that the increase in the global prevalence of overweight and obesity is a major public health issue that must be accompanied by a transformation of our ICUs, both in terms of equipment and human resources. Research must also focus more on these patients to improve their care. In this review, we focused on the central role of obesity in critically ill patients during this pandemic, highlighting its specificities during their stay in the ICU, identifying the lessons we have learned, and identifying areas for future research as well as the future challenges for ICU activity.
Collapse
Affiliation(s)
- Alexandra Beurton
- Department of Intensive Care, Hôpital Tenon, APHP, Paris, France.
- UMR_S 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM, Sorbonne Université, Paris, France.
| | - Emma J Kooistra
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Audrey De Jong
- Anesthesia and Critical Care Department, Saint Eloi Teaching Hospital, University Montpellier 1, Montpellier, France
- Phymed Exp INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Helmut Schiffl
- Division of Nephrology, Department of Internal Medicine IV, University Hospital LMU Munich, Munich, Germany
| | - Mercedes Jourdain
- CHU Lille, Univ-Lille, INSERM UMR 1190, ICU Department, F-59037, Lille, France
| | - Bruno Garcia
- CHU Lille, Univ-Lille, INSERM UMR 1190, ICU Department, F-59037, Lille, France
| | - Damien Vimpère
- Anesthesia and Critical Care Department, Hôpital Necker, APHP, Paris, France
| | - Samir Jaber
- Anesthesia and Critical Care Department, Saint Eloi Teaching Hospital, University Montpellier 1, Montpellier, France
- Phymed Exp INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Laurent Papazian
- Intensive Care Unit, Centre Hospitalier de Bastia, Bastia, Corsica, France
- Aix-Marseille University, Marseille, France
| |
Collapse
|
33
|
Griffin DO. Postacute Sequelae of COVID (PASC or Long COVID): An Evidenced-Based Approach. Open Forum Infect Dis 2024; 11:ofae462. [PMID: 39220656 PMCID: PMC11363684 DOI: 10.1093/ofid/ofae462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
While the acute manifestations of infectious diseases are well known, in some individuals, symptoms can either persist or appear after the acute period. Postviral fatigue syndromes are recognized with other viral infections and are described after coronavirus disease 2019 (COVID-19). We have a growing number of individuals with symptoms that persist for weeks, months, and years. Here, we share the evidence regarding the abnormalities associated with postacute sequelae of COVID-19 (PASC) and therapeutics. We describe physiological and biochemical abnormalities seen in individuals reporting PASC. We describe the several evidence-based interventions to offer patients. It is expected that this growing understanding of the mechanisms driving PASC and the benefits seen with certain therapeutics may not only lead to better outcomes for those with PASC but may also have the potential for understanding and treating other postinfectious sequelae.
Collapse
Affiliation(s)
- Daniel O Griffin
- Division of Infectious Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
34
|
MacGregor F, Oprey A, Caulfield C, MacTavish P, Lowrie R, Henderson P. Does timing of tocilizumab administration affect mortality in COVID-19? A Scottish multicentre retrospective cohort study. BMJ Open Respir Res 2024; 11:e002264. [PMID: 39214629 PMCID: PMC11367351 DOI: 10.1136/bmjresp-2023-002264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The optimal timing of tocilizumab treatment during the disease course of COVID-19 has yet to be adequately defined in the context of randomised controlled trials and the effect of tocilizumab on real-world populations remains unclear. We examined the effect of different timing of tocilizumab, on mortality, in a cohort of adults with COVID-19. METHODS All adults (≥18 years old) with confirmed COVID-19 admitted to four hospitals in the West of Scotland between 8 January 2021 and 31 March 2021 and who received tocilizumab were included in a retrospective observational cohort study. Patients were assigned to either an early (day of admission or first day after admission) or late (days 2-7 of admission) cohort based on tocilizumab initiation. The primary outcome was 90-day all-cause mortality in early versus late cohorts. Secondary outcomes were 28 and 180-day all-cause mortality. RESULTS 203 patients were included in the analysis (138 in the early cohort, 65 in the late cohort). Mortality in 90 days in the early cohort was 22% (n=30) compared with 45% (n=29) in the late cohort (p<0.001). The adjusted mortality was significantly higher in the late cohort compared with the early cohort (adjusted OR: 3.33; 95% CI: 1.29 to 8.54; p=0.012). The secondary outcomes demonstrated the same effect with higher rates of death in 28 days (late cohort adjusted OR: 3.28; 95% CI: 1.23 to 8.75; p=0.018) and 180 days (late cohort adjusted OR: 3.70; 95% CI: 1.45 to 9.45; p=0.006). The effect was seen whether the outcome was adjusted or unadjusted. CONCLUSION Early administration of tocilizumab within the first 2 days of hospitalisation was associated with a significant survival benefit compared with late exposure. Late administration was associated with particularly high mortality. The observed association may be a result of residual confounders and further research is needed.
Collapse
Affiliation(s)
- Fiona MacGregor
- Royal Alexandra Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Alison Oprey
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Carolyn Caulfield
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Pamela MacTavish
- Glasgow Royal Infirmary, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Richard Lowrie
- Pharmacy Services, NHS Greater Glasgow and Clyde, Glasgow, Glasgow, UK
| | - Philip Henderson
- Royal Alexandra Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| |
Collapse
|
35
|
Hermann M, König S, Laxar D, Krall C, Kraft F, Krenn K, Baumgartner C, Tretter V, Maleczek M, Hermann A, Fraunschiel M, Ullrich R. Low-Frequency Ventilation May Facilitate Weaning in Acute Respiratory Distress Syndrome Treated with Extracorporeal Membrane Oxygenation: A Randomized Controlled Trial. J Clin Med 2024; 13:5094. [PMID: 39274307 PMCID: PMC11396271 DOI: 10.3390/jcm13175094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
Although extracorporeal membrane ventilation offers the possibility for low-frequency ventilation, protocols commonly used in patients with acute respiratory distress syndrome (ARDS) and treated with extracorporeal membrane oxygenation (ECMO) vary largely. Whether strict adherence to low-frequency ventilation offers benefit on important outcome measures is poorly understood. Background/Objectives: This pilot clinical study investigated the efficacy of low-frequency ventilation on ventilator-free days (VFDs) in patients suffering from ARDS who were treated with ECMO therapy. Methods: In this single-center randomized controlled trial, 44 (70% male) successive ARDS patients treated with ECMO (aged 56 ± 12 years, SAPS III 64 (SD ± 14)) were randomly assigned 1:1 to the control group (conventional ventilation) or the treatment group (low-frequency ventilation during first 72 h on ECMO: respiratory rate 4-5/min; PEEP 14-16 cm H2O; plateau pressure 23-25 cm H2O, tidal volume: <4 mL/kg). The primary endpoint was VFDs at day 28 after starting ECMO treatment. The major secondary endpoint was ICU mortality, 28-day mortality and 90-day mortality. Results: Twenty-three (52%) patients were successfully weaned from ECMO and were discharged from the intensive care unit (ICU). Twelve patients in the treatment group and five patients in the control group showed more than one VFD at day 28 of ECMO treatment. VFDs were 3.0 (SD ± 5.5) days in the control group and 5.4 (SD ± 6) days in the treatment group (p = 0.117). Until day 28 of ECMO initiation, patients in the treatment group could be successfully weaned off of the ventilator more often (OR of 0.164 of 0 VFDs at day 28 after ECMO start; 95% CI 0.036-0.758; p = 0.021). ICU mortality did not differ significantly (36% in treatment group and 59% in control group; p = 0.227). Conclusions: Low-frequency ventilation is comparable to conventional protective ventilation in patients with ARDS who have been treated with ECMO. However, low-frequency ventilation may support weaning from invasive mechanical ventilation in patients suffering from ARDS and treated with ECMO therapy.
Collapse
Affiliation(s)
- Martina Hermann
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Währingerstraße 104/10, 1180 Vienna, Austria
| | - Sebastian König
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Daniel Laxar
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Währingerstraße 104/10, 1180 Vienna, Austria
| | - Christoph Krall
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria
| | - Felix Kraft
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Katharina Krenn
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Clemens Baumgartner
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Verena Tretter
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Mathias Maleczek
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Alexander Hermann
- Department of Medicine I, Intensive Care Unit 13i2, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Melanie Fraunschiel
- IT4Science, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Roman Ullrich
- Department of Anesthesiology and Intensive Care Medicine, AUVA Trauma Center Vienna, Kundratstraße 37, 1120 Vienna, Austria
| |
Collapse
|
36
|
Seguí E, Torres JM, Auclin E, Casadevall D, Peiro Carmona S, Aguilar-Company J, García de Herreros M, Gorría T, Laguna JC, Rodríguez M, González A, Epaillard N, Gavira J, Bolaño V, Tapia JC, Tagliamento M, Teixidó C, Arasanz H, Pilotto S, Lopez-Castro R, Mielgo-Rubio X, Urbano C, Recondo G, Diaz Pavon M, Bluthgen MV, Minatta JN, Lupinacci L, Brasó-Maristany F, Prat A, Vlagea A, Mezquita L. The FLARE Score and Circulating Neutrophils in Patients with Cancer and COVID-19 Disease. Cancers (Basel) 2024; 16:2974. [PMID: 39272832 PMCID: PMC11393969 DOI: 10.3390/cancers16172974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
PURPOSE Inflammation and neutrophils play a central role in both COVID-19 disease and cancer. We aimed to assess the impact of pre-existing tumor-related inflammation on COVID-19 outcomes in patients with cancer and to elucidate the role of circulating neutrophil subpopulations. METHODS We conducted a multicenter retrospective analysis of 524 patients with cancer and SARS-CoV-2 infection, assessing the relationship between clinical outcomes and circulating inflammatory biomarkers collected before and during COVID-19 infection. Additionally, a single-center prospective cohort study provided data for an exploratory analysis, assessing the immunophenotype of circulating neutrophils and inflammatory cytokines. The primary endpoints were 30-day mortality and the severity of COVID-19 disease. RESULTS Prior to COVID-19, 25% of patients with cancer exhibited elevated dNLR, which increased to 55% at the time of COVID-19 diagnosis. We developed the FLARE score, incorporating both tumor- and infection-induced inflammation, which categorized patients into four prognostic groups. The poor prognostic group had a 30-day mortality rate of 68%, significantly higher than the 23% in the favorable group (p < 0.0001). This score proved to be an independent predictor of early mortality. This prospective analysis revealed a shift towards immature forms of neutrophils and higher IL-6 levels in patients with cancer and severe COVID-19 infection. CONCLUSIONS A pre-existing tumor-induced pro-inflammatory state significantly impacts COVID-19 outcomes in patients with cancer. The FLARE score, derived from circulating inflammatory markers, emerges as an easy-to-use, globally accessible, effective tool for clinicians to identify patients with cancer at heightened risk of severe COVID-19 complications and early mortality who might benefit most from immediate and intensive treatment strategies. Furthermore, our findings underscore the significance of immature neutrophils in the progression of COVID-19 in patients with cancer, advocating for further investigation into how these cells contribute to both cancer and COVID-19 disease.
Collapse
Affiliation(s)
- Elia Seguí
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Juan Manuel Torres
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Edouard Auclin
- Department of Medical Oncology, Hopital Europeen George Pompidou, AP-HP, Université Paris Cité, 75015 Paris, France
| | - David Casadevall
- Department of Medical Oncology, Hospital del Mar, 08036 Barcelona, Spain
| | - Sara Peiro Carmona
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Juan Aguilar-Company
- Department of Medical Oncology, Vall d'Hebron University Hospital, 08036 Barcelona, Spain
| | - Marta García de Herreros
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Teresa Gorría
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Juan Carlos Laguna
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Marta Rodríguez
- Department of Medical Oncology, Vall d'Hebron University Hospital, 08036 Barcelona, Spain
- Department of Medical Oncology, Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
| | - Azucena González
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Nicolas Epaillard
- Department of Medical Oncology, Hopital Europeen George Pompidou, AP-HP, Université Paris Cité, 75015 Paris, France
| | - Javier Gavira
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08036 Barcelona, Spain
| | - Victor Bolaño
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Jose C Tapia
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08036 Barcelona, Spain
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties, University of Genova, 16126 Genova, Italy
- Academic Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Cristina Teixidó
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Pathology and CORE Molecular Biology laboratory, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Hugo Arasanz
- Department of Medical Oncology, Hospital Universitario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Sara Pilotto
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37126 Verona, Italy
| | - Rafael Lopez-Castro
- Department of Medical Oncology, Hospital Clinico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Xabier Mielgo-Rubio
- Department of Medical Oncology, Hospital Universitario Fundación Alcorcon, 28922 Alcorcon, Spain
| | - Cristina Urbano
- Department of Medical Oncology, Hospital General de Granollers, 08402 Granollers, Spain
| | - Gonzalo Recondo
- Department of Medical Oncology, Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Buenos Aires C1000, Argentina
| | - Mar Diaz Pavon
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | | | - José Nicolas Minatta
- Department of Medical Oncology, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Lorena Lupinacci
- Department of Medical Oncology, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Institute of Oncology (IOB)-Hospital Quirón Salud, 08023 Barcelona, Spain
- Reveal Genomics, 08036 Barcelona, Spain
| | - Alexandru Vlagea
- Department of Immunology, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Laura Mezquita
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
37
|
Binabaji S, Rahimi M, Rajabi H, Keshavarz M, Rahimi R, Ahmadi A, Gahreman D. Effects of physical training on coagulation parameters, interleukin-6, and angiotensin-converting enzyme-2 in COVID-19 survivors. Sci Rep 2024; 14:18968. [PMID: 39152162 PMCID: PMC11329640 DOI: 10.1038/s41598-024-67522-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/11/2024] [Indexed: 08/19/2024] Open
Abstract
COVID-19 is a highly contagious virus that uses Angiotensin-converting enzyme 2 (ACE2) as a receptor to enter human cells. The virus leads to an increase in inflammatory cytokines (i.e. IL-6) and an impaired coagulation system, which can cause serious complications during and after the disease. Physical exercise has been shown to improve COVID-19 complications through various mechanisms, such as modulation of the immune and coagulation systems. Therefore, this study investigated the effects of 8 weeks of training on inflammatory, coagulation, and physical factors in patients with COVID-19 during the recovery phase. Twenty-seven male and female volunteers (age 20-45 years) who recently recovered from COVID-19 were assigned to the control (n = 13) or the training group (n = 14). Blood samples, aerobic capacity and muscle endurance were collected 24 h before the start of the interventions and 24 h after the final training session in week 4 and 48 h after the final training session in week 8. IL-6, ACE2, fibrinogen, and D-dimer were measured using ELISA. The training group showed a significant increase in muscle endurance (p = 0.004) and aerobic capacity (p = 0.009) compared to the control group. Serum levels of IL-6 and fibrinogen decreased in the training group but this decrease was not statistically significant (p > 0.05). Despite a slight increase in the quality of life and sleep in the training group, no statistically significant difference was observed between the training and the control group. It appears that physical training has beneficial effects on the coagulation system, inflammatory factors, and sleep quality and can facilitate the recovery of COVID-19 patients.
Collapse
Affiliation(s)
- Soheila Binabaji
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Rahimi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Kharazmi University, Tehran, Iran
| | - Hamid Rajabi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Kharazmi University, Tehran, Iran.
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Rahimeh Rahimi
- Department of Biochemistry, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Azam Ahmadi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Kharazmi University, Tehran, Iran
| | - Daniel Gahreman
- Department of Sport, Exercise, Recreation, and Kinesiology, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
38
|
Lee KG, Roca O, Casey JD, Semler MW, Roman-Sarita G, Yarnell CJ, Goligher EC. When to intubate in acute hypoxaemic respiratory failure? Options and opportunities for evidence-informed decision making in the intensive care unit. THE LANCET. RESPIRATORY MEDICINE 2024; 12:642-654. [PMID: 38801827 DOI: 10.1016/s2213-2600(24)00118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 05/29/2024]
Abstract
The optimal timing of intubation in acute hypoxaemic respiratory failure is uncertain and became a point of controversy during the COVID-19 pandemic. Invasive mechanical ventilation is a potentially life-saving intervention but carries substantial risks, including injury to the lungs and diaphragm, pneumonia, intensive care unit-acquired muscle weakness, and haemodynamic impairment. In deciding when to intubate, clinicians must balance premature exposure to the risks of ventilation with the potential harms of unassisted breathing, including disease progression and worsening multiorgan failure. Currently, the optimal timing of intubation is unclear. In this Personal View, we examine a range of parameters that could serve as triggers to initiate invasive mechanical ventilation. The utility of a parameter (eg, the ratio of arterial oxygen tension to fraction of inspired oxygen) to predict the likelihood of a patient undergoing intubation does not necessarily mean that basing the timing of intubation on that parameter will improve therapeutic outcomes. We examine options for clinical investigation to make progress on establishing the optimal timing of intubation.
Collapse
Affiliation(s)
- Kevin G Lee
- Department of Physiology, Toronto, ON, Canada; Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Oriol Roca
- Servei de Medicina Intensiva, Parc Taulí Hospital Universitari, Institut de Recerca Parc Taulí-I3PT, Sabadell, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain; Ciber Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Jonathan D Casey
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew W Semler
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Christopher J Yarnell
- Interdepartmental Division of Critical Care Medicine University of Toronto, Toronto, ON, Canada; Department of Medicine, Division of Respirology, University Health Network, Toronto, ON, Canada; Institute of Health Policy, Management, and Evaluation at the University of Toronto, Toronto, ON, Canada; Scarborough Health Network, Department of Critical Care Medicine, Toronto, ON, Canada; Scarborough Health Network Research Institute, Toronto, ON, Canada.
| | - Ewan C Goligher
- Department of Physiology, Toronto, ON, Canada; Interdepartmental Division of Critical Care Medicine University of Toronto, Toronto, ON, Canada; Department of Medicine, Division of Respirology, University Health Network, Toronto, ON, Canada; Toronto General Hospital Research Institute, Toronto, ON, Canada
| |
Collapse
|
39
|
Méndez Rodríguez ML, Ponciano-Gómez A, Campos-Aguilar M, Tapia-Sánchez WD, Duarte-Martínez CL, Romero-Herrera JS, Olivas-Quintero S, Saucedo-Campos AD, Méndez-Cruz AR, Jimenez-Flores R, Ortiz-Navarrete V, Romero-Ramírez H, Santos-Argumedo L, Rosales-García VH. Neutrophil-to-Lymphocyte Ratio and Cytokine Profiling as Predictors of Disease Severity and Survival in Unvaccinated COVID-19 Patients. Vaccines (Basel) 2024; 12:861. [PMID: 39203987 PMCID: PMC11360520 DOI: 10.3390/vaccines12080861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND During the COVID-19 pandemic, identifying reliable biomarkers for predicting disease severity and patient outcomes in unvaccinated individuals is essential. This study evaluates the efficacy of key hematological markers, including leukocyte and neutrophil counts, Neutrophil-to-Lymphocyte Ratio (NLR), and cytokine profiles (IL-6, INF-γ, TNF-α, IL-17A, CCL2, and CXCL10) for predicting the necessity for mechanical ventilation and assessing survival probabilities. METHODS We conducted an in-depth analysis on a cohort of COVID-19 patients, emphasizing the relationship between NLR, cytokine profiles, and clinical outcomes, utilizing routine leukocyte counting and cytokine quantification by flow cytometry. RESULTS Elevated leukocyte and neutrophil counts, increased NLR, and significant cytokine elevations such as IL-6 and IL-10 were strongly associated with the need for mechanical ventilation, reflecting a pronounced systemic inflammatory response indicative of severe disease outcomes. CONCLUSION Integrating hematological markers, particularly NLR and cytokine profiles, is crucial in predicting mechanical ventilation needs and survival in non-vaccinated COVID-19 patients. Our findings provide critical insights into the pathophysiology of COVID-19, supporting the development of more targeted clinical interventions and potentially informing future strategies for managing infectious disease outbreaks.
Collapse
Affiliation(s)
- Miguel Leonardo Méndez Rodríguez
- Servicio de Inmunología y Alergia, Centro Médico Naval (CEMENAV), Secretaria de Marina (SEMAR), Avenida Heroica Escuela Naval Militar 745, Coapa, Presidentes Ejidales 1ra Sección, Coyoacán, Mexico City 04470, Mexico; (M.L.M.R.); (J.S.R.-H.)
| | - Alberto Ponciano-Gómez
- Laboratorio de Inmunología (UMF), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Barrios N° 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico; (A.P.-G.); (M.C.-A.); (A.D.S.-C.); (A.R.M.-C.); (R.J.-F.)
| | - Myriam Campos-Aguilar
- Laboratorio de Inmunología (UMF), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Barrios N° 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico; (A.P.-G.); (M.C.-A.); (A.D.S.-C.); (A.R.M.-C.); (R.J.-F.)
| | - Wilfrido David Tapia-Sánchez
- Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Basiliso Romo Anguiano 124, Industrial, Gustavo A. Madero, Mexico City 07800, Mexico; (W.D.T.-S.); (C.L.D.-M.)
| | - Carlos Leonardo Duarte-Martínez
- Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Basiliso Romo Anguiano 124, Industrial, Gustavo A. Madero, Mexico City 07800, Mexico; (W.D.T.-S.); (C.L.D.-M.)
| | - Jesús Salvador Romero-Herrera
- Servicio de Inmunología y Alergia, Centro Médico Naval (CEMENAV), Secretaria de Marina (SEMAR), Avenida Heroica Escuela Naval Militar 745, Coapa, Presidentes Ejidales 1ra Sección, Coyoacán, Mexico City 04470, Mexico; (M.L.M.R.); (J.S.R.-H.)
| | - Sandra Olivas-Quintero
- Departamento de Ciencias de la Salud Culiacán, Universidad Autónoma de Occidente, Culiacan 80020, Sinaloa, Mexico;
| | - Alberto Daniel Saucedo-Campos
- Laboratorio de Inmunología (UMF), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Barrios N° 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico; (A.P.-G.); (M.C.-A.); (A.D.S.-C.); (A.R.M.-C.); (R.J.-F.)
| | - Adolfo Rene Méndez-Cruz
- Laboratorio de Inmunología (UMF), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Barrios N° 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico; (A.P.-G.); (M.C.-A.); (A.D.S.-C.); (A.R.M.-C.); (R.J.-F.)
| | - Rafael Jimenez-Flores
- Laboratorio de Inmunología (UMF), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Barrios N° 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico; (A.P.-G.); (M.C.-A.); (A.D.S.-C.); (A.R.M.-C.); (R.J.-F.)
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico; (V.O.-N.); (H.R.-R.); (L.S.-A.)
| | - Hector Romero-Ramírez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico; (V.O.-N.); (H.R.-R.); (L.S.-A.)
- Centro de Investigación Sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico; (V.O.-N.); (H.R.-R.); (L.S.-A.)
- Centro de Investigación Sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Victor Hugo Rosales-García
- Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Basiliso Romo Anguiano 124, Industrial, Gustavo A. Madero, Mexico City 07800, Mexico; (W.D.T.-S.); (C.L.D.-M.)
- Laboratorios Nacionales de Servicios Experimentales, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 14330, Mexico
| |
Collapse
|
40
|
Feng L, Liu S, Wang J, Gao Y, Xie F, Gong J, Bi S, Yao Z, Li Y, Liu W, Guan C, Zhang M, Wang H, Zheng J. The performance of a combination of heparin-binding protein with other biomarkers for sepsis diagnosis: an observational cohort study. BMC Infect Dis 2024; 24:755. [PMID: 39080540 PMCID: PMC11290073 DOI: 10.1186/s12879-024-09666-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND HBP, a novel biomarker released from neutrophils, may induce inflammatory responses and exacerbate vascular permeability, representing the pathophysiological characteristics of sepsis and septic shock. However, it remains uncertain whether the combination of HBP with other biomarkers yields enhanced diagnostic capacity for sepsis. We hypothesized that measurements included IL-6·IL-8·HBP, IL-6·IL-8·HBP/ALB and HBP/ALB which based on HBP will improve its diagnostic efficacy and even better than the traditional infection biomarkers. METHODS Between July 2021 and June 2022, we carried out a comprehensive, multi-center, observational cohort study spanning six leading tertiary hospitals located in Heilongjiang Province, China. Patients were stratified into three categories based on the severity of infection: non-sepsis, sepsis, and septic shock. We collected clinical and laboratory data, along with infection and inflammation biomarkers, for analysis. RESULTS A total of 195 patients were enrolled. Among the three groups, patients with septic shock (n = 75, 38.5%) had significantly higher baseline levels of HBP, WBC, Lac, CRP, PCT, IL-6, IL-8, and IL-10 compared to non-sepsis patients (n = 43, 22.0%) and sepsis patients (n = 77, 39.5%), with statistically significant differences (p < 0.05) observed for all parameters. When compared to SOFA score and traditional markers of CRP, PCT, IL-6 and IL-8, the combined indexes of IL-6·IL-8·HBP and IL-6·IL-8·HBP/ALB demonstrated significantly improved diagnostic performance for sepsis and septic shock (AUC 0.911 and 0.902 respectively, p < 0.001). CONCLUSIONS The combined measurements of IL-6·IL-8·HBP and IL-6·IL-8·HBP/ALB can augment the diagnostic capacity of HBP for sepsis, and offer reliable early supplementary indicators to traditional biomarkers for assessing disease severity in patients with infection.
Collapse
Affiliation(s)
- Liwei Feng
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Department of Intensive Care Medicine, The Arong Banner People's Hospital, Hulunbuir, Inner Mongolia Autonomous Region, 162750, China
| | - Shujie Liu
- Department of Intensive Care Medicine, The Second People's Hospital of Mudanjiang, Mudanjiang, Heilongjiang, 157000, China
| | - Jieying Wang
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yan Gao
- Department of Intensive Care Medicine, The Fouth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Fengjie Xie
- Department of Intensive Care Medicine, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang, 157011, China
| | - Jianguo Gong
- Department of Intensive Care Medicine, The People's Hospital of Daqing, Daqing, Heilongjiang, 163711, China
| | - Sheng Bi
- Department of Intensive Care Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, 161005, China
| | - Zhipeng Yao
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yue Li
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wenhua Liu
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chunming Guan
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Ming Zhang
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Hongliang Wang
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Junbo Zheng
- Department of Intensive Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
41
|
Chhabra M, Shanthamurthy CD, Kumar NV, Mardhekar S, Vishweshwara SS, Wimmer N, Modhiran N, Watterson D, Amarilla AA, Cha JS, Beckett JR, De Voss JJ, Kayal Y, Vlodavsky I, Dorsett LR, Smith RAA, Gandhi NS, Kikkeri R, Ferro V. Amphiphilic Heparinoids as Potent Antiviral Agents against SARS-CoV-2. J Med Chem 2024; 67:11885-11916. [PMID: 38995734 DOI: 10.1021/acs.jmedchem.4c00487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Herein, we report the synthesis and biological evaluation of a novel series of heparinoid amphiphiles as inhibitors of heparanase and SARS-CoV-2. By employing a tailor-made synthetic strategy, a library of highly sulfated homo-oligosaccharides bearing d-glucose or a C5-epimer (i.e., l-idose or l-iduronic acid) conjugated with various lipophilic groups was synthesized and investigated for antiviral activity. Sulfated higher oligosaccharides of d-glucose or l-idose with lipophilic aglycones displayed potent anti-SARS-CoV-2 and antiheparanse activity, similar to or better than pixatimod (PG545), and were more potent than their isosteric l-iduronic acid congeners. Lipophilic groups such as cholestanol and C18-aliphatic substitution are more advantageous than functional group appended lipophilic moieties. These findings confirm that fine-tuning of higher oligosaccharides, degree of sulfation, and lipophilic groups can yield compounds with potent anti-SARS-CoV-2 activity.
Collapse
Affiliation(s)
- Mohit Chhabra
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chethan D Shanthamurthy
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune 411008, India
| | | | - Sandhya Mardhekar
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune 411008, India
| | - Sharath S Vishweshwara
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune 411008, India
| | - Norbert Wimmer
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jonathan S Cha
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - James R Beckett
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - James J De Voss
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yasmin Kayal
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion─Israel Institute of Technology, Haifa 31096, Israel
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion─Israel Institute of Technology, Haifa 31096, Israel
| | - Lauren R Dorsett
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia
| | - Raymond A A Smith
- School of Chemical Engineering, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Neha S Gandhi
- Centre for Genomics and Personalised Health, School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia
- Department of Computer Science and Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Raghavendra Kikkeri
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune 411008, India
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
42
|
Karahmet Sher E, Alebić M, Marković Boras M, Boškailo E, Karahmet Farhat E, Karahmet A, Pavlović B, Sher F, Lekić L. Nanotechnology in medicine revolutionizing drug delivery for cancer and viral infection treatments. Int J Pharm 2024; 660:124345. [PMID: 38885775 DOI: 10.1016/j.ijpharm.2024.124345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Advancements in nanotechnology were vastly applied in medicine and pharmacy, especially in the field of nano-delivery systems. It took a long time for these systems to ensure precise delivery of very delicate molecules, such as RNA, to cells at concentrations that yield remarkable efficiency, with success rates reaching 95.0% and 94.5%. These days, there are several advantages of using nanotechnological solutions in the prevention and treatment of cancer and viral infections. Its interventions improve treatment outcomes both due to increased effectiveness of the drug at target location and by reducing adverse reactions, thereby increasing patient adherence to the therapy. Based on the current knowledge an updated review was made, and perspective, opportunities and challenges in nanomedicine were discussed. The methods employed include comprehensive examination of existing literature and studies on nanoparticles and nano-delivery systems including both in vitro tests performed on cell cultures and in vivo assessments carried out on appropriate animal models, with a specific emphasis on their applications in oncology and virology. This brings together various aspects including both structure and formation as well as its association with characteristic behaviour in organisms, providing a novel perspective. Furthermore, the practical application of these systems in medicine and pharmacy with a focus on viral diseases and malignancies was explored. This review can serve as a valuable guide for fellow researchers, helping them navigate the abundance of findings in this field. The results indicate that applications of nanotechnological solutions for the delivery of medicinal products improving therapeutic outcomes will continue to expand.
Collapse
Affiliation(s)
- Emina Karahmet Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Mirna Alebić
- Department of Pharmacy, University Hospital Centre Zagreb, Zagreb 10000, Croatia
| | - Marijana Marković Boras
- Department of Laboratory Diagnostic, University Clinical Hospital Mostar, Mostar 88000, Bosnia and Herzegovina; International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Emina Boškailo
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Esma Karahmet Farhat
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Department of Food and Nutrition, Faculty of Food Technology, Juraj Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Alma Karahmet
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Bojan Pavlović
- Faculty of Physical Education and Sports, University of East Sarajevo, Lukavica, Republika Srpska 75327, Bosnia and Herzegovina
| | - Farooq Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Lana Lekić
- Faculty of Health Studies, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina
| |
Collapse
|
43
|
Porter JC, Inshaw J, Solis VJ, Denneny E, Evans R, Temkin MI, De Vasconcelos N, Aramburu IV, Hoving D, Basire D, Crissell T, Guinto J, Webb A, Esmail H, Johnston V, Last A, Rampling T, Lippert L, Helbig ET, Kurth F, Williams B, Flynn A, Lukey PT, Birault V, Papayannopoulos V. Anti-inflammatory therapy with nebulized dornase alfa for severe COVID-19 pneumonia: a randomized unblinded trial. eLife 2024; 12:RP87030. [PMID: 39009040 PMCID: PMC11251720 DOI: 10.7554/elife.87030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
Background Prinflammatory extracellular chromatin from neutrophil extracellular traps (NETs) and other cellular sources is found in COVID-19 patients and may promote pathology. We determined whether pulmonary administration of the endonuclease dornase alfa reduced systemic inflammation by clearing extracellular chromatin. Methods Eligible patients were randomized (3:1) to the best available care including dexamethasone (R-BAC) or to BAC with twice-daily nebulized dornase alfa (R-BAC + DA) for seven days or until discharge. A 2:1 ratio of matched contemporary controls (CC-BAC) provided additional comparators. The primary endpoint was the improvement in C-reactive protein (CRP) over time, analyzed using a repeated-measures mixed model, adjusted for baseline factors. Results We recruited 39 evaluable participants: 30 randomized to dornase alfa (R-BAC +DA), 9 randomized to BAC (R-BAC), and included 60 CC-BAC participants. Dornase alfa was well tolerated and reduced CRP by 33% compared to the combined BAC groups (T-BAC). Least squares (LS) mean post-dexamethasone CRP fell from 101.9 mg/L to 23.23 mg/L in R-BAC +DA participants versus a 99.5 mg/L to 34.82 mg/L reduction in the T-BAC group at 7 days; p=0.01. The anti-inflammatory effect of dornase alfa was further confirmed with subgroup and sensitivity analyses on randomised participants only, mitigating potential biases associated with the use of CC-BAC participants. Dornase alfa increased live discharge rates by 63% (HR 1.63, 95% CI 1.01-2.61, p=0.03), increased lymphocyte counts (LS mean: 1.08 vs 0.87, p=0.02) and reduced circulating cf-DNA and the coagulopathy marker D-dimer (LS mean: 570.78 vs 1656.96 μg/mL, p=0.004). Conclusions Dornase alfa reduces pathogenic inflammation in COVID-19 pneumonia, demonstrating the benefit of cost-effective therapies that target extracellular chromatin. Funding LifeArc, Breathing Matters, The Francis Crick Institute (CRUK, Medical Research Council, Wellcome Trust). Clinical trial number NCT04359654.
Collapse
Affiliation(s)
- Joanna C Porter
- UCL Respiratory, University College LondonLondonUnited Kingdom
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | | | | | - Emma Denneny
- UCL Respiratory, University College LondonLondonUnited Kingdom
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Rebecca Evans
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Mia I Temkin
- Antimicrobial Defence Lab, The Francis Crick InstituteLondonUnited Kingdom
| | | | | | - Dennis Hoving
- Antimicrobial Defence Lab, The Francis Crick InstituteLondonUnited Kingdom
| | - Donna Basire
- UCL Respiratory, University College LondonLondonUnited Kingdom
| | - Tracey Crissell
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Jesusa Guinto
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Alison Webb
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Hanif Esmail
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Victoria Johnston
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Anna Last
- University College London Hospitals NHS TrustLondonUnited Kingdom
- Clinical Research Department, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Thomas Rampling
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Lena Lippert
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Elisa Theresa Helbig
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Florian Kurth
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Bryan Williams
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | | | | | | | | |
Collapse
|
44
|
Rabøl Andersen L, Hindsberger B, Bastrup Israelsen S, Pedersen L, Bela Szecsi P, Benfield T. Higher levels of IL-1ra, IL-6, IL-8, MCP-1, MIP-3α, MIP-3β, and fractalkine are associated with 90-day mortality in 132 non-immunomodulated hospitalized patients with COVID-19. PLoS One 2024; 19:e0306854. [PMID: 38985797 PMCID: PMC11236197 DOI: 10.1371/journal.pone.0306854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION Immune dysregulation with an excessive release of cytokines has been identified as a key driver in the development of severe COVID-19. The aim of this study was to evaluate the initial cytokine profile associated with 90-day mortality and respiratory failure in a cohort of patients hospitalized with COVID 19 that did not receive immunomodulatory therapy. METHODS Levels of 45 cytokines were measured in blood samples obtained at admission from patients with confirmed COVID-19. Logistic regression analysis was utilized to determine the association between cytokine levels and outcomes. The primary outcome was death within 90 days from admission and the secondary outcome was need for mechanical ventilation. RESULTS A total of 132 patients were included during the spring of 2020. We found that one anti-inflammatory cytokine, one pro-inflammatory cytokine, and five chemokines were associated with the odds of 90-day mortality, specifically: interleukin-1 receptor antagonist, interleukin-6, interleukin-8, monocyte chemoattractant protein-1, macrophage inflammatory protein-3α, macrophage inflammatory protein-3β, and fractalkine. All but fractalkine were also associated with the odds of respiratory failure during admission. Monocyte chemoattractant protein-1 showed the strongest estimate of association with both outcomes. CONCLUSION We showed that one anti-inflammatory cytokine, one pro-inflammatory cytokine, and five chemokines were associated with 90-day mortality in patients hospitalized with COVID-19 that did not receive immunomodulatory therapy.
Collapse
Affiliation(s)
- Liv Rabøl Andersen
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Bettina Hindsberger
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Simone Bastrup Israelsen
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Lise Pedersen
- Department of Clinical Biochemistry, Holbaek Hospital, Holbaek, Denmark
| | - Pal Bela Szecsi
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Biochemistry, Holbaek Hospital, Holbaek, Denmark
| | - Thomas Benfield
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Bhalerao KS, De Silva PIT, Hiniduma K, Grunbaum A, Rozza N, Kremer R, Rusling JF. Microfluidic Immunoarray for Point-of-Care Detection of Cytokines in COVID-19 Patients. ACS OMEGA 2024; 9:29320-29330. [PMID: 39005811 PMCID: PMC11238202 DOI: 10.1021/acsomega.4c00735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
The "cytokine storm" often induced in COVID-19 patients contributes to the onset of "acute respiratory distress syndrome" (ARDS) accompanied by lung infection and damage, multiorgan failure, and even death. This large increase in pro-inflammatory cytokines in blood may be related to severity. Rapid, on-demand cytokine analyses can thus be critical to inform treatment plans and improve survival rates. Here, we report a sensitive, low-cost, semiautomated 3D-printed microfluidic immunoarray to detect 2 cytokines and CRP simultaneously in a single 10 μL serum sample in 25 min. Accuracy was validated by analyzing 80 COVID-19 patient serum samples, with results well correlated to a commercial Meso Scale protein immunoassay. Capture antibodies immobilized in detection microwells in a flat well plate-type flow chamber facilitate the immunoassay, with a programmable syringe pump automatically delivering reagents. Chemiluminescence signals were captured in a dark box with a CCD camera integrated for 30 s. This system was optimized to detect inflammation biomarkers IL-6, IFN-γ, and CRP simultaneously in blood serum. Ultralow limits of detection (LODs) of 0.79 fg/mL for IL-6, 4.2 fg/mL for CRP, and 2.7 fg/mL for IFN-γ with dynamic ranges of up to 100 pg/mL were achieved. ROC statistical analyses showed a relatively good diagnostic value related to the samples assigned WHO COVID-19 scores for disease severity, with the best results for IL-6 and CRP. Monitoring these biomarkers for coronavirus severity may allow prediction of disease severity as a basis for critical treatment decisions and better survival rates.
Collapse
Affiliation(s)
- Ketki S Bhalerao
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - P I Thilini De Silva
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Keshani Hiniduma
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ami Grunbaum
- Department of Medicine, McGill University Health Centre, 1001 Decarie Blvd., Montreal, QC H3A 1A1, Canada
| | - Nicholas Rozza
- Department of Medicine, McGill University Health Centre, 1001 Decarie Blvd., Montreal, QC H3A 1A1, Canada
| | - Richard Kremer
- Department of Medicine, McGill University Health Centre, 1001 Decarie Blvd., Montreal, QC H3A 1A1, Canada
| | - James F Rusling
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
- Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- School of Chemistry, National University of Ireland at Galway, Galway H91 TK33, Ireland
| |
Collapse
|
46
|
Nauriyal D, Dubey R, Agrawal P, Kumar D, Punj A, Nasser K. A cross-sectional study on clinical characteristics and severity of children with COVID-19 admitted to a teaching institute in North India. J Family Med Prim Care 2024; 13:2653-2662. [PMID: 39071009 PMCID: PMC11272026 DOI: 10.4103/jfmpc.jfmpc_1734_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 07/30/2024] Open
Abstract
Background SARS-CoV-2 infection presentation in children is usually milder than in adults but can be severe and fatal as well. Data on the pediatric population regarding severity and clinical presentation are still limited, and there is a need to have a better understanding of clinical features, severity, and laboratory parameters. Aims and Objective To document clinical and laboratory characteristics and outcomes of children with SARS-CoV-2 in a low-middle-income country and to evaluate clinicodemographic factors and biochemical markers associated with severity and mortality. Materials and Methods A hospital-based cross-sectional study was conducted among 112 COVID-19-positive children at a designated Level-3 center in North India. Clinical characteristics, laboratory parameters, and severity of COVID-19 cases as well as factors associated with the severity of the disease, were analyzed by descriptive statistics and a Chi-square test. Results The adolescent age group (age 12-18 years) was affected most (64.3%). Male patients accounted for 56.3% of total cases. Fever was the most common symptom (41.1%) followed by cough. Presenting complaints were highest from the respiratory system (32.1%) followed by the gastrointestinal (8.9%) and the neurological system (7.1%). Majority of patients had mild disease (87%) while 13% had the moderate-severe disease. Spo2 < 95% (P = 0.00001), neutrophilia (P < 0.000001), lymphopenia (P < 0.000001), elevated values of C-reactive protein (P < 0.00001), Interleukin-6 (P = 0.002), D- dimer (P = 0.00014) and respiratory symptoms as presenting complaints (P < 0.000001) were found to be significantly associated with severity of disease. Conclusion The male and adolescent age group was affected most. Presenting complaints were highest from the respiratory system. Unusual presentation may have gastrointestinal or neurological presentation. Most children with COVID-19 had mild disease. Moderate to severe disease was not uncommon. Factors including neutrophilia, lymphopenia, elevated lab values of C-reactive protein, D-dimer, and interleukin-6 had a significant association with the severity of the disease. These biomarkers can help predict the severity of the disease.
Collapse
Affiliation(s)
- Deepty Nauriyal
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Rishabh Dubey
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Pulak Agrawal
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Deepak Kumar
- Department of Community Medicine, Maharishi Markandeshwar University and Hospital, Solan, Himachal Pradesh, India
| | - Ajay Punj
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Kaynat Nasser
- Department of Community Medicine, Subharti Medical College, Meerut, Uttar Pradesh, India
| |
Collapse
|
47
|
Aggarwal NR, Nordwall J, Braun DL, Chung L, Coslet J, Der T, Eriobu N, Ginde AA, Hayanga AJ, Highbarger H, Holodniy M, Horcajada JP, Jain MK, Kim K, Laverdure S, Lundgren J, Natarajan V, Nguyen HH, Pett SL, Phillips A, Poulakou G, Price DA, Robinson P, Rogers AJ, Sandkovsky U, Shaw-Saliba K, Sturek JM, Trautner BW, Waters M, Reilly C. Viral and Host Factors Are Associated With Mortality in Hospitalized Patients With COVID-19. Clin Infect Dis 2024; 78:1490-1503. [PMID: 38376212 PMCID: PMC11175705 DOI: 10.1093/cid/ciad780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Persistent mortality in adults hospitalized due to acute COVID-19 justifies pursuit of disease mechanisms and potential therapies. The aim was to evaluate which virus and host response factors were associated with mortality risk among participants in Therapeutics for Inpatients with COVID-19 (TICO/ACTIV-3) trials. METHODS A secondary analysis of 2625 adults hospitalized for acute SARS-CoV-2 infection randomized to 1 of 5 antiviral products or matched placebo in 114 centers on 4 continents. Uniform, site-level collection of participant baseline clinical variables was performed. Research laboratories assayed baseline upper respiratory swabs for SARS-CoV-2 viral RNA and plasma for anti-SARS-CoV-2 antibodies, SARS-CoV-2 nucleocapsid antigen (viral Ag), and interleukin-6 (IL-6). Associations between factors and time to mortality by 90 days were assessed using univariate and multivariable Cox proportional hazards models. RESULTS Viral Ag ≥4500 ng/L (vs <200 ng/L; adjusted hazard ratio [aHR], 2.07; 1.29-3.34), viral RNA (<35 000 copies/mL [aHR, 2.42; 1.09-5.34], ≥35 000 copies/mL [aHR, 2.84; 1.29-6.28], vs below detection), respiratory support (<4 L O2 [aHR, 1.84; 1.06-3.22]; ≥4 L O2 [aHR, 4.41; 2.63-7.39], or noninvasive ventilation/high-flow nasal cannula [aHR, 11.30; 6.46-19.75] vs no oxygen), renal impairment (aHR, 1.77; 1.29-2.42), and IL-6 >5.8 ng/L (aHR, 2.54 [1.74-3.70] vs ≤5.8 ng/L) were significantly associated with mortality risk in final adjusted analyses. Viral Ag, viral RNA, and IL-6 were not measured in real-time. CONCLUSIONS Baseline virus-specific, clinical, and biological variables are strongly associated with mortality risk within 90 days, revealing potential pathogen and host-response therapeutic targets for acute COVID-19 disease.
Collapse
Affiliation(s)
- Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jacquie Nordwall
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dominique L Braun
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lucy Chung
- CAMRIS International (under contract no. 75N93019D00025 with National Institute of Allergy and Infectious Diseases, Department of Health and Human Services), National Institute of Health, Bethesda, Maryland, USA
| | - Jordan Coslet
- Velocity Clinical Research, Chula Vista, California, USA
| | - Tatyana Der
- Department of General Internal Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Adit A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Awori J Hayanga
- Department of Cardiovascular Thoracic Surgery, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Helene Highbarger
- Virus Isolation and Serology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Mark Holodniy
- Veterans Affairs Palo Alto Health Care System, Division of Infectious Diseases and Geographic Medicine, Stanford University, Palo Alto, California, USA
| | - Juan P Horcajada
- Department of Infectious Diseases, Hospital del Mar Research Insititute, UPF, Barcelona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Mamta K Jain
- Division of Infectious Diseases and Geotropical Medicine, UT Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Kami Kim
- Division of Infectious Disease and International Medicine, Morsani College of Medicine, University of South Florida and Global Emerging Diseases Institute, Tampa General Hospital, Tampa, Florida, USA
| | - Sylvain Laverdure
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Jens Lundgren
- CHIP Center of Excellence for Health, Immunity, and Infections and Department of Infectious Diseases, Righospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Hien H Nguyen
- Division of Infectious Diseases, Veterans Affairs Northern California, University of California, Davis, Sacramento, California, USA
| | - Sarah L Pett
- The Medical Research Council Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, United Kingdom
- Institute for Global Health, University College London, London, United Kingdom
| | - Andrew Phillips
- Institute for Global Health, University College London, London, United Kingdom
| | - Garyphallia Poulakou
- Third Department of Medicine and Laboratory National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - David A Price
- Newcastle Upon Tyne NHUS Hospitals Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Philip Robinson
- Infection Prevention and Hospital Epidemiology, Hoag Memorial Hospital Presbyterian, Newport Beach, California, USA
| | - Angela J Rogers
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Palo Alto, California, USA
| | - Uriel Sandkovsky
- Division of Infectious Diseases, Baylor University Medical Center, Dallas, Texas, USA
| | - Katy Shaw-Saliba
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey M Sturek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UVA Health, Charlottesville, Virginia, USA
| | - Barbara W Trautner
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Michael Waters
- Velocity Clinical Research, Chula Vista, California, USA
| | - Cavan Reilly
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
48
|
Wolf MA, O'Hara JM, Bitzer GJ, Narayanan E, Boehm DT, Bevere JR, DeJong MA, Hall JM, Wong TY, Falcone S, Deal CE, Richards A, Green S, Nguyen B, King E, Ogega C, Russo L, Sen-Kilic E, Plante O, Himansu S, Barbier M, Carfi A, Damron FH. Multivalent mRNA-DTP vaccines are immunogenic and provide protection from Bordetella pertussis challenge in mice. NPJ Vaccines 2024; 9:103. [PMID: 38858423 PMCID: PMC11164898 DOI: 10.1038/s41541-024-00890-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Acellular multivalent vaccines for pertussis (DTaP and Tdap) prevent symptomatic disease and infant mortality, but immunity to Bordetella pertussis infection wanes significantly over time resulting in cyclic epidemics of pertussis. The messenger RNA (mRNA) vaccine platform provides an opportunity to address complex bacterial infections with an adaptable approach providing Th1-biased responses. In this study, immunogenicity and challenge models were used to evaluate the mRNA platform with multivalent vaccine formulations targeting both B. pertussis antigens and diphtheria and tetanus toxoids. Immunization with mRNA formulations were immunogenetic, induced antigen specific antibodies, as well as Th1 T cell responses. Upon challenge with either historical or contemporary B. pertussis strains, 6 and 10 valent mRNA DTP vaccine provided protection equal to that of 1/20th human doses of either DTaP or whole cell pertussis vaccines. mRNA DTP immunized mice were also protected from pertussis toxin challenge as measured by prevention of lymphocytosis and leukocytosis. Collectively these pre-clinical mouse studies illustrate the potential of the mRNA platform for multivalent bacterial pathogen vaccines.
Collapse
Affiliation(s)
- M Allison Wolf
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - Graham J Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Megan A DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | | | | | | | | | | | | | | | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | | | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA.
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA.
| |
Collapse
|
49
|
Uno K, Hasan A, Nakayama EE, Rahim R, Harada H, Kaneko M, Hashimoto S, Tanaka T, Matsumoto H, Fujimiya H, Shioda T, Rahman M, Yoshizaki K. Predictive biomarkers of COVID-19 prognosis identified in Bangladesh patients and validated in Japanese cohorts. Sci Rep 2024; 14:12713. [PMID: 38830928 PMCID: PMC11148188 DOI: 10.1038/s41598-024-63184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Despite high vaccination rates globally, countries are still grappling with new COVID infections, and patients diagnosed as mild dying at home during outpatient treatment. Hence, this study aim to identify, then validate, biomarkers that could predict if newly infected COVID-19 patients would subsequently require hospitalization or could recover safely with medication as outpatients. Serum cytokine/chemokine data from 129 COVID-19 patients within 7 days after the onset of symptoms in Bangladesh were used as training data. The majority of patients were infected with the Omicron variant and over 88% were vaccinated. Patients were divided into those with mild symptoms who recovered, and those who deteriorated to moderate or severe illness. Using the Lasso method, 15 predictive markers were identified and used to classify patients into these two groups. The biomarkers were then validated in a cohort of 194 Covid patients in Japan with a predictive accuracy that exceeded 80% for patients infected with Delta and Omicron variants, and 70% for Wuhan and Alpha variants. In an environment of widespread vaccination, these biomarkers could help medical practitioners determine if newly infected COVID-19 patients will improve and can be managed on an out-patient basis, or if they will deteriorate and require hospitalization.
Collapse
Affiliation(s)
- Kazuko Uno
- IFN and Host-Defense Research Laboratory, Louis Pasteur Center for Medical Research, Kyoto, Kyoto, 606-8225, Japan.
| | - Abu Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | - Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0781, Japan
| | - Rummana Rahim
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | | | | | - Shoji Hashimoto
- Osaka Prefectural Hospital Organization Osaka Habikino Medical Center, Habikino, Osaka, 583‑8588, Japan
| | - Toshio Tanaka
- Kinki Central Hospital, Itami, Hyogo, 664-8533, Japan
| | - Hisatake Matsumoto
- Trauma and Acute Critical Care Center, Osaka University, Suita, Osaka, 565‑0871, Japan
| | | | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0781, Japan
| | - Mizanur Rahman
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | - Kazuyuki Yoshizaki
- Department of Organic Fine Chemicals, Institute of Scientific and Industry Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
50
|
Alladina JW, Giacona FL, Haring AM, Hibbert KA, Medoff BD, Schmidt EP, Thompson T, Maron BA, Alba GA. Circulating Biomarkers of Endothelial Dysfunction Associated With Ventilatory Ratio and Mortality in ARDS Resulting From SARS-CoV-2 Infection Treated With Antiinflammatory Therapies. CHEST CRITICAL CARE 2024; 2:100054. [PMID: 39035722 PMCID: PMC11259037 DOI: 10.1016/j.chstcc.2024.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
BACKGROUND The association of plasma biomarkers and clinical outcomes in ARDS resulting from SARS-CoV-2 infection predate the evidence-based use of immunomodulators. RESEARCH QUESTION Which plasma biomarkers are associated with clinical outcomes in patients with ARDS resulting from SARS-CoV-2 infection treated routinely with immunomodulators? STUDY DESIGN AND METHODS We collected plasma from patients with ARDS resulting from SARS-CoV-2 infection within 24 h of admission to the ICU between December 2020 and March 2021 (N = 69). We associated 16 total biomarkers of inflammation (eg, IL-6), coagulation (eg, D-dimer), epithelial injury (eg, surfactant protein D), and endothelial injury (eg, angiopoietin-2) with the primary outcome of in-hospital mortality and secondary outcome of ventilatory ratio (at baseline and day 3). RESULTS Thirty patients (43.5%) died within 60 days. All patients received corticosteroids and 6% also received tocilizumab. Compared with survivors, nonsurvivors demonstrated a higher baseline modified Sequential Organ Failure Assessment score (median, 8.5 [interquartile range (IQR), 7-9] vs 7 [IQR, 5-8]); P = .004), lower Pao2 to Fio2 ratio (median, 153 [IQR, 118-182] vs 184 [IQR, 142-247]; P = .04), and higher ventilatory ratio (median, 2.0 [IQR, 1.9-2.3] vs 1.5 [IQR, 1.4-1.9]; P < .001). No difference was found in inflammatory, coagulation, or epithelial biomarkers between groups. Nonsurvivors showed higher median neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) levels (median, 8.4 ng/mL [IQR, 7.0-11.2 ng/mL] vs 6.9 ng/mL [IQR, 5.5-8.0 ng/mL]; P = .0025), von Willebrand factor domain A2 levels (8.7 ng/mL [IQR, 7.9-9.7 ng/mL] vs 6.5 ng/mL [IQR, 5.7-8.7 ng/mL]; P = .007), angiopoietin-2 levels (9.0 ng/mL [IQR, 7.9-14.1 ng/mL] vs 7.0 ng/mL [IQR, 5.6-10.6 ng/mL]; P = .01), and syndecan-1 levels (15.9 ng/mL [IQR, 14.5-17.5 ng/mL] vs 12.6 ng/mL [IQR, 10.5-16.1 ng/mL]; P = .01). Only NEDD9 level met the adjusted threshold for significance (P < .003). Plasma NEDD9 level was associated with 60-day mortality (adjusted OR, 9.7; 95% CI, 1.6-60.4; P = .015). Syndecan-1 level correlated with both baseline (ρ = 0.4; P = .001) and day 3 ventilatory ratio (ρ = 0.5; P < .001). INTERPRETATION Biomarkers of inflammation, coagulation, and epithelial injury were not associated with clinical outcomes in a small cohort of patients with ARDS uniformly treated with immunomodulators. However, endothelial biomarkers, including plasma NEDD9, were associated with 60-day mortality.
Collapse
Affiliation(s)
- Jehan W Alladina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Francesca L Giacona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alexis M Haring
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kathryn A Hibbert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Bradley A Maron
- Department of Medicine; University of Maryland School of Medicine, Baltimore, University of Maryland-Institute for Health Computing, Bethesda, MD
| | - George A Alba
- Division of Pulmonary and Critical Care Medicine, Bethesda, MD, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|