1
|
Wang S, Tai Y, Yang X, Li P, Wang H, Tan Y, Gao T, Chu M, Liu M. Identification of potential biomarkers and drug of ischemic stroke in patients with COVID-19 through machine learning. Heliyon 2024; 10:e39039. [PMID: 39502238 PMCID: PMC11536010 DOI: 10.1016/j.heliyon.2024.e39039] [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: 06/06/2024] [Revised: 09/19/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
The relationship between COVID-19 and ischemic stroke (IS) has attracted significant attention, yet the precise mechanism at the gene level remains unclear. This study aims to reveal potential biomarkers and drugs for COVID-19-related IS through bioinformatics methods. We collected two gene expression profiling datasets, GSE16561 and GSE213313, and selected GSE179879 and GSE196822 as validation sets for analysis. Through analysis, we identified 77 differentially expressed genes (DEGs) shared between COVID-19 and IS. Further gene enrichment analysis revealed that these genes are primarily involved in immune regulation. By constructing a protein-protein interaction network, we screened out nine hub genes, including FCGR3A, KLRB1, IL2RB, CD2, IL7R, CCR7, CD3D, GZMK, and ITK. In LASSO regression analysis, we evaluated the ROC curve's area under the curve (AUC) scores of key genes to assess their diagnostic accuracy. Subsequently, we performed random forest (RF), Support Vector Machine Recursive Feature Elimination (SVM-RFE), and neural network construction on hub genes to ensure accurate diagnosis of IS. Finally, by intersecting the results of three algorithms (LASSO regression, random forest, and SVM), CD3D and ITK were identified as the ultimate key genes. Based on this, we predicted potential targeted drug Blinatumomab. These research findings provide clues for a deeper understanding of the biological mechanisms of COVID-19-related IS and offer new insights for exploring novel treatment approaches.
Collapse
Affiliation(s)
- Sixian Wang
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yuxing Tai
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xiaoqian Yang
- Changchun University of Chinese Medicine, Changchun 130117, China
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Peizhe Li
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Han Wang
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yi Tan
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianjiao Gao
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Mingrui Chu
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Mingjun Liu
- Changchun University of Chinese Medicine, Changchun 130117, China
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
2
|
Shen CJ, Hu SY, Hou CP, Shen CF, Cheng CM. T Cell Responses to SARS-CoV-2 in Vaccinated Pregnant Women: A Comparative Study of Pre-Pregnancy and During-Pregnancy Infections. Vaccines (Basel) 2024; 12:1208. [PMID: 39591111 PMCID: PMC11598868 DOI: 10.3390/vaccines12111208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/06/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
The COVID-19 pandemic has posed unprecedented challenges to global public health, particularly for vulnerable populations like pregnant women. This study delves into the T cell immune responses in pregnant women with confirmed SARS-CoV-2 infection, all of whom received three doses of a COVID-19 vaccine. Using the ELISpot assay, we measured T cell responses against SARS-CoV-2 spike S1 and nucleocapsid peptides in two groups: those infected before and during pregnancy. Our results showed weak to moderate correlations between T cell responses and neutralizing antibody levels, with no statistically significant differences between the two groups. T cell reactivity appeared to decrease over time post-diagnosis, regardless of infection timing. Intriguingly, over half of the participants maintained detectable T cell memory responses beyond one year post-infection, suggesting the long-term persistence of cellular immunity. These insights contribute to the understanding of COVID-19 immunology in pregnant women, highlighting the importance of considering both humoral and cellular immune responses in this high-risk population.
Collapse
Affiliation(s)
- Ching-Ju Shen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shu-Yu Hu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan;
| | | | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan;
| |
Collapse
|
3
|
Pasupuleti D, Bagwe P, Ferguson A, Uddin MN, D'Souza MJ, Zughaier SM. Evaluating Nanoparticulate Vaccine Formulations for Effective Antigen Presentation and T-Cell Proliferation Using an In Vitro Overlay Assay. Vaccines (Basel) 2024; 12:1049. [PMID: 39340079 PMCID: PMC11435973 DOI: 10.3390/vaccines12091049] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Inducing T lymphocyte (T-cell) activation and proliferation with specificity against a pathogen is crucial in vaccine formulation. Assessing vaccine candidates' ability to induce T-cell proliferation helps optimize formulation for its safety, immunogenicity, and efficacy. Our in-house vaccine candidates use microparticles (MPs) and nanoparticles (NPs) to enhance antigen stability and target delivery to antigen-presenting cells (APCs), providing improved immunogenicity. Typically, vaccine formulations are screened for safety and immunostimulatory effects using in vitro methods, but extensive animal testing is often required to assess immunogenic responses. We identified the need for a rapid, intermediate screening process to select promising candidates before advancing to expensive and time-consuming in vivo evaluations. In this study, an in vitro overlay assay system was demonstrated as an effective high-throughput preclinical testing method to evaluate the immunogenic properties of early-stage vaccine formulations. The overlay assay's effectiveness in testing particulate vaccine candidates for immunogenic responses has been evaluated by optimizing the carboxyfluorescein succinimidyl ester (CFSE) T-cell proliferation assay. DCs were overlaid with T-cells, allowing vaccine-stimulated DCs to present antigens to CFSE-stained T-cells. T-cell proliferation was quantified using flow cytometry on days 0, 1, 2, 4, and 6 upon successful antigen presentation. The assay was tested with nanoparticulate vaccine formulations targeting Neisseria gonorrhoeae (CDC F62, FA19, FA1090), measles, H1N1 flu prototype, canine coronavirus, and Zika, with adjuvants including Alhydrogel® (Alum) and AddaVax™. The assay revealed robust T-cell proliferation in the vaccine treatment groups, with variations between bacterial and viral vaccine candidates. A dose-dependent study indicated immune stimulation varied with antigen dose. These findings highlight the assay's potential to differentiate and quantify effective antigen presentation, providing valuable insights for developing and optimizing vaccine formulations.
Collapse
Affiliation(s)
- Dedeepya Pasupuleti
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Priyal Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Amarae Ferguson
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Mohammad N Uddin
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Martin J D'Souza
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Susu M Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2731, Qatar
| |
Collapse
|
4
|
Li J, Wang N, Mao G, Wang J, Xiang M, Zhang H, Zeng D, Ma H, Jiang J. Cuproptosis-associated lncRNA impact prognosis in patients with non-small cell lung cancer co-infected with COVID-19. J Cell Mol Med 2024; 28:e70059. [PMID: 39228012 PMCID: PMC11371660 DOI: 10.1111/jcmm.70059] [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: 01/31/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) patients infected with COVID-19 experience much worse prognosis. However, the specific mechanisms behind this phenomenon remain unclear. We conducted a multicentre study, collecting surgical tissue samples from a total of 36 NSCLC patients across three centres to analyse. Among the 36 lung cancer patients, 9 were infected with COVID-19. COVID-19 infection (HR = 21.62 [1.58, 296.06], p = 0.021) was an independent risk factor of progression-free survival (PFS). Analysis of RNA-seq data of these cancer tissues demonstrated significantly higher expression levels of cuproptosis-associated genes in COVID-19-infected lung cancer patients. Using Lasso regression and Cox regression analysis, we identified 12 long noncoding RNAs (lncRNA) regulating cuproptosis. A score based on these lncRNA were used to divide patients into high-risk and low-risk groups. The results showed that the high-risk group had lower overall survival and PFS compared to the low-risk group. Furthermore, Tumor Immune Dysfunction and Exclusion (TIDE) database revealed that the high-risk group benefited more from immunotherapy. Drug sensitivity analysis identified cetuximab and gefitinib as potentially effective treatments for the high-risk group. Cuproptosis plays a significant role NSCLC patients infected with COVID-19. Promisingly, cetuximab and gefitinib have shown potential effectiveness for managing these patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Respiratory and Critical Care MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
| | - Nan Wang
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
| | - Guocai Mao
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouJiangsuChina
| | - Jiantang Wang
- Department of Respiratory and Critical Care MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
| | - Mengqi Xiang
- Department of Medical OncologySichuan Cancer Hospital, Medical School of University of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Huachuan Zhang
- Department of Thoracic SurgerySichuan Cancer Hospital, Medical School of University of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Daxiong Zeng
- Department of Respiratory and Critical Care MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouJiangsuChina
| | - Haitao Ma
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouJiangsuChina
| | - Junhong Jiang
- Department of Respiratory and Critical Care MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Centre of Soochow UniversitySuzhouJiangsuChina
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
5
|
Xiong H, Meng X, Song Y, Zhong J, Liu S, Zhu X, Ye X, Zhong Y, Zhang D. Have Previous COVID-19 Vaccinations Shaped the Potential Enhancing Infection of Variant Strains? Vaccines (Basel) 2024; 12:567. [PMID: 38932296 PMCID: PMC11209312 DOI: 10.3390/vaccines12060567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the infection status of Omicron in the population and the association between COVID-19 vaccination and infection with Omicron. METHODS We conducted a cross-sectional study to openly recruit participants for a survey of SARS-CoV-2 infection by convenience sampling from 1 January to 15 January 2023 after a COVID-19 pandemic swept across China. Additionally, the binary logistic regression model was adopted to evaluate the association between COVID-19 vaccination and the infection outcomes or symptom severity, respectively. Meanwhile, the relations between the vaccination and duration of the symptoms were estimated via ordinal logistic analysis. RESULTS Of the 2007 participants, the prevalence of infection with Omicron was 82.9%. Compared with unvaccinated individuals, inactivated COVID-19 vaccination could increase the risk of Omicron infection (OR = 1.942, 95% CI: 1.093-3.448), and the receipt of at least one dose of non-inactivated COVID-19 vaccines was a protective factor against infection (OR = 0.428, 95% CI: 0.226-0.812). By contrast, no relations were observed in COVID-19 vaccination with the symptoms of infection and duration of symptoms (p > 0.05). CONCLUSIONS This cross-sectional study concluded that inactivated COVID-19 vaccination might increase the risk of Omicron infection, which should be a concern during COVID-19 vaccination and the treatment of variant infections in the future, and the receipt of at least one dose of non-inactivated COVID-19 vaccine was a protective factor against infection.
Collapse
Affiliation(s)
- Husheng Xiong
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (H.X.); (X.M.); (J.Z.); (S.L.)
| | - Xiang Meng
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (H.X.); (X.M.); (J.Z.); (S.L.)
| | - Yanqin Song
- The Fourth People’s Hospital of Foshan City, Foshan 528000, China;
| | - Jiayi Zhong
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (H.X.); (X.M.); (J.Z.); (S.L.)
| | - Shuang Liu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (H.X.); (X.M.); (J.Z.); (S.L.)
| | - Xun Zhu
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
| | - Xin Ye
- Tianhe District Center for Disease Control and Prevention, Guangzhou 510630, China;
| | - Yonghui Zhong
- The Fourth People’s Hospital of Foshan City, Foshan 528000, China;
| | - Dingmei Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (H.X.); (X.M.); (J.Z.); (S.L.)
| |
Collapse
|
6
|
Hartmann JA, Cardoso MR, Talarico MCR, Kenney DJ, Leone MR, Reese DC, Turcinovic J, O'Connell AK, Gertje HP, Marino C, Ojeda PE, De Paula EV, Orsi FA, Velloso LA, Cafiero TR, Connor JH, Ploss A, Hoelzemer A, Carrington M, Barczak AK, Crossland NA, Douam F, Boucau J, Garcia-Beltran WF. Evasion of NKG2D-mediated cytotoxic immunity by sarbecoviruses. Cell 2024; 187:2393-2410.e14. [PMID: 38653235 PMCID: PMC11088510 DOI: 10.1016/j.cell.2024.03.026] [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: 07/25/2023] [Revised: 01/30/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024]
Abstract
SARS-CoV-2 and other sarbecoviruses continue to threaten humanity, highlighting the need to characterize common mechanisms of viral immune evasion for pandemic preparedness. Cytotoxic lymphocytes are vital for antiviral immunity and express NKG2D, an activating receptor conserved among mammals that recognizes infection-induced stress ligands (e.g., MIC-A/B). We found that SARS-CoV-2 evades NKG2D recognition by surface downregulation of MIC-A/B via shedding, observed in human lung tissue and COVID-19 patient serum. Systematic testing of SARS-CoV-2 proteins revealed that ORF6, an accessory protein uniquely conserved among sarbecoviruses, was responsible for MIC-A/B downregulation via shedding. Further investigation demonstrated that natural killer (NK) cells efficiently killed SARS-CoV-2-infected cells and limited viral spread. However, inhibition of MIC-A/B shedding with a monoclonal antibody, 7C6, further enhanced NK-cell activity toward SARS-CoV-2-infected cells. Our findings unveil a strategy employed by SARS-CoV-2 to evade cytotoxic immunity, identify the culprit immunevasin shared among sarbecoviruses, and suggest a potential novel antiviral immunotherapy.
Collapse
Affiliation(s)
- Jordan A Hartmann
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | | | - Devin J Kenney
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Madison R Leone
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
| | - Dagny C Reese
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jacquelyn Turcinovic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Aoife K O'Connell
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Hans P Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Caitlin Marino
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
| | - Pedro E Ojeda
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
| | - Erich V De Paula
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil; Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | - Fernanda A Orsi
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil; Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | - Licio Augusto Velloso
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, Brazil
| | - Thomas R Cafiero
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - John H Connor
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Angelique Hoelzemer
- First Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Institute for Infection and Vaccine Development (IIRVD), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Research Department Virus Immunology, Leibniz Institute for Virology, Hamburg, Germany
| | - Mary Carrington
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Amy K Barczak
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nicholas A Crossland
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Julie Boucau
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA.
| | - Wilfredo F Garcia-Beltran
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Nakayama T, Todaka R, Sawada A, Ito T, Fujino M, Haga K, Katayama K. Different immunological responses following immunization with two mRNA vaccines. J Infect Chemother 2024; 30:439-449. [PMID: 38000497 DOI: 10.1016/j.jiac.2023.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023]
Abstract
INTRODUCTION Immunological responses were investigated following immunization with two mRNA vaccines: BNT162b2 and mRNA-1273. METHODS Neutralizing antibody (NAb) was assayed before, 2-4 weeks after, and 3 and 6 months after the primary immunization, and the same time-points after booster dose with 6- or 8-months interval. Whole-blood culture was stimulated with spike antigen, and cytokine production was assayed. RESULTS NAb was detected after primary immunization, NAb titers began to decrease three months after primary immunization with BNT162b2, lower than those after mRNA-1273, and elevated after booster immunization. The NAb level was 1/2 lower against δ variant, and 1/16 lower against omicron variant in comparison with that against α variant. Cytokine production following immunization with mRNA-1273 was maintained within three months at higher levels of Th1 (TNF-α), Th2 (IL-4 and IL-5), and inflammatory cytokines (IL-6 and IL-17) than that following immunization with BNT162b2, reflecting prominent levels of NAb following immunization with mRNA-1273. Cytokine production decreased six months after primary immunization in both vaccine recipients and was enhanced following booster doses. During the omicron outbreak, medical staff members in the outpatient office experienced asymptomatic infection, with a greater than 4-fold increase in NAb titers against omicron variant even after booster immunization. Asymptomatic infection enhanced the production of Th2 and inflammatory cytokines. CONCLUSION mRNA-1273 induced stronger NAb responses with wide-range cross-reactive antibodies against δ and omicron variants. mRNA-1273 induced higher levels of Th1, Th2, and inflammatory cytokines than BNT162b2 did, reflecting higher levels of NAb against variant strains.
Collapse
Affiliation(s)
- Tetsuo Nakayama
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan.
| | - Reiko Todaka
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan.
| | - Akihito Sawada
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan.
| | - Takashi Ito
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan; Department of Pediatrics, Kitasato University Hospital, Sagamihara, Kanagawa, 252-0329, Japan.
| | - Motoko Fujino
- Department of Pediatrics, Saiseikai Central Hospital Tokyo, Tokyo, 108-0073, Japan.
| | - Kei Haga
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan.
| | - Kazuhiko Katayama
- Laboratory of Viral Infection, Ömura Satoshi Memorial Institute, Tokyo, 108-8641, Japan.
| |
Collapse
|
8
|
Wang X, Zhang M, Wei K, Li C, Yang J, Jiang S, Zhao C, Zhao X, Qiao R, Cui Y, Chen Y, Li J, Cai G, Liu C, Yu J, Zhang W, Xie F, Wang P, Zhang Y. Longitudinal Analysis of Humoral and Cellular Immune Response up to 6 Months after SARS-CoV-2 BA.5/BF.7/XBB Breakthrough Infection and BA.5/BF.7-XBB Reinfection. Vaccines (Basel) 2024; 12:464. [PMID: 38793715 PMCID: PMC11125724 DOI: 10.3390/vaccines12050464] [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: 03/25/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
The rapid mutation of SARS-CoV-2 has led to multiple rounds of large-scale breakthrough infection and reinfection worldwide. However, the dynamic changes of humoral and cellular immunity responses to several subvariants after infection remain unclear. In our study, a 6-month longitudinal immune response evaluation was conducted on 118 sera and 50 PBMC samples from 49 healthy individuals who experienced BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection. By studying antibody response, memory B cell, and IFN-γ secreting CD4+/CD8+ T cell response to several SARS-CoV-2 variants, we observed that each component of immune response exhibited distinct kinetics. Either BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection induces relatively high level of binding and neutralizing antibody titers against Omicron subvariants at an early time point, which rapidly decreases over time. Most of the individuals at 6 months post-breakthrough infection completely lost their neutralizing activities against BQ.1.1, CH.1.1, BA.2.86, JN.1 and XBB subvariants. Individuals with BA.5/BF.7-XBB reinfection exhibit immune imprinting shifting and recall pre-existing BA.5/BF.7 neutralization antibodies. In the BA.5 breakthrough infection group, the frequency of BA.5 and XBB.1.16-RBD specific memory B cells, resting memory B cells, and intermediate memory B cells gradually increased over time. On the other hand, the frequency of IFN-γ secreting CD4+/CD8+ T cells induced by WT/BA.5/XBB.1.16 spike trimer remains stable over time. Overall, our research indicates that individuals with breakthrough infection have rapidly declining antibody levels but have a relatively stable cellular immunity that can provide some degree of protection from future exposure to new antigens.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Meng Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Kaifeng Wei
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jinghui Yang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Changyi Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jizhen Yu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Wenhong Zhang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200437, China;
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| |
Collapse
|
9
|
Moguem Soubgui AF, Ndeme Mboussi WS, Kojom Foko LP, Embolo Enyegue EL, Koanga Mogtomo ML. Serological surveillance reveals a high exposure to SARS-CoV-2 and altered immune response among COVID-19 unvaccinated Cameroonian individuals. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0002380. [PMID: 38346064 PMCID: PMC10861046 DOI: 10.1371/journal.pgph.0002380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024]
Abstract
Surveillance of COVID-19/SARS-CoV-2 dynamics is crucial to understanding natural history and providing insights into the population's exposure risk and specific susceptibilities. This study investigated the seroprevalence of SARS-CoV-2 antibodies, its predictors, and immunological status among unvaccinated patients in Cameroon. A multicentre cross-sectional study was conducted between January and September 2022 in the town of Douala. Patients were consecutively recruited, and data of interest were collected using a questionnaire. Blood samples were collected to determine Immunoglobin titres (IgM and IgG), interferon gamma (IFN- γ) and interleukin-6 (IL-6) by ELISA, and CD4+ cells by flow cytometry. A total of 342 patients aged 41.5 ± 13.9 years were included. Most participants (75.8%) were asymptomatic. The overall crude prevalence of IgM and IgG was 49.1% and 88.9%, respectively. After adjustment, the seroprevalence values were 51% for IgM and 93% for IgM. Ageusia and anosmia have displayed the highest positive predictive values (90.9% and 82.4%) and specificity (98.9% and 98.3%). The predictors of IgM seropositivity were being diabetic (aOR = 0.23, p = 0.01), frequently seeking healthcare (aOR = 1.97, p = 0.03), and diagnosed with ageusia (aOR = 20.63, p = 0.005), whereas those of IgG seropositivity included health facility (aOR = 0.15, p = 0.01), age of 40-50 years (aOR = 8.78, p = 0.01), married (aOR = 0.21, p = 0.02), fever (aOR = 0.08, p = 0.01), and ageusia (aOR = 0.08, p = 0.01). CD4+, IFN-γ, and IL-6 were impaired in seropositive individuals, with a confounding role of socio-demographic factors or comorbidities. Although the WHO declared the end of COVID-19 as a public health emergency, the findings of this study indicate the need for continuous surveillance to adequately control the disease in Cameroon.
Collapse
Affiliation(s)
- Arlette Flore Moguem Soubgui
- Faculty of Science, Department of Biochemistry, The University of Douala, Douala, Cameroon
- Centre de Recherche et d’Expertise en Biologie, Douala, Cameroon
| | - Wilfred Steve Ndeme Mboussi
- Faculty of Science, Department of Biochemistry, The University of Douala, Douala, Cameroon
- Centre de Recherche et d’Expertise en Biologie, Douala, Cameroon
| | - Loick Pradel Kojom Foko
- Centre de Recherche et d’Expertise en Biologie, Douala, Cameroon
- Department of Animal Biology, Faculty of Science, The University of Douala, Douala, Cameroon
| | - Elisée Libert Embolo Enyegue
- Center for Research on Health and Priority Diseases, Ministry of Scientific Research and Innovation, Yaoundé, Centre Region, Cameroon
| | - Martin Luther Koanga Mogtomo
- Faculty of Science, Department of Biochemistry, The University of Douala, Douala, Cameroon
- Centre de Recherche et d’Expertise en Biologie, Douala, Cameroon
| |
Collapse
|
10
|
Peng W, Deng R, Wu S, Zhao J, Fu J, Zhang Y, Chen W, Wang W. Increased rates of indeterminate results in QuantiFERON-TB gold in-tube during the COVID-19 epidemic in Hunan province, China. Diagn Microbiol Infect Dis 2024; 108:116131. [PMID: 37976555 DOI: 10.1016/j.diagmicrobio.2023.116131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/18/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Increased rates of indeterminate QuantiFERON-TB Gold In-Tube (QFT-GIT) results have been reported since the COVID-19 epidemic in Hunan Province, China. The indeterminate result (ITR) rate of QFT increased from an average of 5.2% to 12.4%, paralleling the first COVID-19 pandemic wave in the region. QFT-GIT results of 243 hospitalized patients with COVID-19 from January 2022 to April 2023 at Xiangya Hospital of Central South University were analyzed. Of the 243 patients, 71 (29.2%) had ITRs due to reduced interferon-gamma production in the positive control. Multiple factors are associated with ITRs, such as disease severity, respiratory failure incidence, immunosuppressant use, and prognosis. Additionally, interferon-gamma (Mitogen-Nil) levels differed significantly depending upon disease severity, prognosis, immunosuppressant use, sepsis symptoms, respiratory failure, or hyperlipidemia. An abnormal increase in the ITR rate in the QFT was observed after the COVID-19 pandemic, and an optimal machine learning predictive model for indeterminate QFT results was established.
Collapse
Affiliation(s)
- Wanchan Peng
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Rihui Deng
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siyu Wu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Juan Zhao
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Jinfang Fu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Yunli Zhang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Wei Chen
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Wei Wang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
| |
Collapse
|
11
|
Sophonmanee R, Preampruchcha P, Ongarj J, Seeyankem B, Intapiboon P, Surasombatpattana S, Uppanisakorn S, Sangsupawanich P, Chusri S, Pinpathomrat N. Intradermal Fractional ChAdOx1 nCoV-19 Booster Vaccine Induces Memory T Cells: A Follow-Up Study. Vaccines (Basel) 2024; 12:109. [PMID: 38400093 PMCID: PMC10891531 DOI: 10.3390/vaccines12020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
The administration of viral vector and mRNA vaccine booster effectively induces humoral and cellular immune responses. Effector T cell responses after fractional intradermal (ID) vaccination are comparable to those after intramuscular (IM) boosters. Here, we quantified T cell responses after booster vaccination. ChAdOx1 nCoV-19 vaccination induced higher numbers of S1-specific CD8+ memory T cells, consistent with the antibody responses. Effector memory T cell phenotypes elicited by mRNA vaccination showed a similar trend to those elicited by the viral vector vaccine booster. Three months post-vaccination, cytokine responses remained detectable, confirming effector T cell responses induced by both vaccines. The ID fractional dose of ChAdOx1 nCoV-19 elicited higher effector CD8+ T cell responses than IM vaccination. This study confirmed that an ID dose-reduction vaccination strategy effectively stimulates effector memory T cell responses. ID injection could be an improved approach for effective vaccination programs.
Collapse
Affiliation(s)
- Ratchanon Sophonmanee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (R.S.); (P.P.); (J.O.); (B.S.)
| | - Perawas Preampruchcha
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (R.S.); (P.P.); (J.O.); (B.S.)
| | - Jomkwan Ongarj
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (R.S.); (P.P.); (J.O.); (B.S.)
| | - Bunya Seeyankem
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (R.S.); (P.P.); (J.O.); (B.S.)
| | - Porntip Intapiboon
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.I.); (S.C.)
| | | | - Supattra Uppanisakorn
- Clinical Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (S.U.); (P.S.)
| | - Pasuree Sangsupawanich
- Clinical Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (S.U.); (P.S.)
| | - Sarunyou Chusri
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.I.); (S.C.)
| | - Nawamin Pinpathomrat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (R.S.); (P.P.); (J.O.); (B.S.)
| |
Collapse
|
12
|
Wang X, Jiang S, Ma W, Li X, Wei K, Xie F, Zhao C, Zhao X, Wang S, Li C, Qiao R, Cui Y, Chen Y, Li J, Cai G, Liu C, Yu J, Li J, Hu Z, Zhang W, Jiang S, Li M, Zhang Y, Wang P. Enhanced neutralization of SARS-CoV-2 variant BA.2.86 and XBB sub-lineages by a tetravalent COVID-19 vaccine booster. Cell Host Microbe 2024; 32:25-34.e5. [PMID: 38029742 DOI: 10.1016/j.chom.2023.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Emerging SARS-CoV-2 sub-lineages like XBB.1.5, XBB.1.16, EG.5, HK.3 (FLip), and XBB.2.3 and the variant BA.2.86 have recently been identified. Understanding the efficacy of current vaccines on these emerging variants is critical. We evaluate the serum neutralization activities of participants who received COVID-19 inactivated vaccine (CoronaVac), those who received the recently approved tetravalent protein vaccine (SCTV01E), or those who had contracted a breakthrough infection with BA.5/BF.7/XBB virus. Neutralization profiles against a broad panel of 30 sub-lineages reveal that BQ.1.1, CH.1.1, and all the XBB sub-lineages exhibit heightened resistance to neutralization compared to previous variants. However, despite their extra mutations, BA.2.86 and the emerging XBB sub-lineages do not demonstrate significantly increased resistance to neutralization over XBB.1.5. Encouragingly, the SCTV01E booster consistently induces higher neutralizing titers against all these variants than breakthrough infection does. Cellular immunity assays also show that the SCTV01E booster elicits a higher frequency of virus-specific memory B cells. Our findings support the development of multivalent vaccines to combat future variants.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China
| | - Wentai Ma
- Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiangnan Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China
| | - Kaifeng Wei
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shidi Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Changyi Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jizhen Yu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jixi Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Zixin Hu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China; Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mingkun Li
- Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China.
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Farah R, Khamisy-Farah R, Dziedzic K, Skrzypek E, Pruc M, Bragazzi NL, Szarpak L, Jomah M. The role of the neutrophil-lymphocyte ratio in predicting poor outcomes in COVID-19 patients. Cardiol J 2023; 31:374-380. [PMID: 38149489 PMCID: PMC11229800 DOI: 10.5603/cj.98214] [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: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND This study examines how the neutrophil-lymphocyte ratio (NLR) predicts coronavirus disease 2019 (COVID-19) hospitalization, severity, length, and mortality in adult patients. METHODS A study was done using a retrospective, single-center, observational design. A total of 400 patients who were admitted to the Ziv Medical Center (Safed, Israel) from April 2020 to December 2021 with a confirmed diagnosis of COVID-19 through RT-PCR testing were included in the analysis. Two complete blood count laboratory tests were conducted for each patient. The first test was administered upon admission to the hospital, while the second test was conducted prior to the patient's discharge from the hospital or a few days before their death. RESULTS Four hundred patients were included in the study, 206 males (51.5%) and 194 females (48.5%). The mean age was 64.5 ± 17.1 years. In the group of cases, there were 102 deaths, and 296 survivors were recorded, with a fatality rate of 25.5%. The median NLR was 6.9 ± 5.8 at the beginning of hospitalization and 15.1 ± 32.9 at the end of hospitalization (p < 0.001). The median length of hospital stay was 9.4 ± 8.8 days. NLR in the fatality group was 34.0 ± 49.9 compared to 8.4 ± 20.4 in the survivor group (p < 0.001). Comparison between the NLR at the time of admission of the patient and before discharge/death was 6.9 ± 5.8 vs. 15.1 ± 32.9 (p < 0.001). CONCLUSIONS The analyses conducted revealed a statistically significant correlation between the NLR and the severity, mortality rates, and the duration of hospitalization. The consideration of NLR should commence during the initial phases of the disease when assessing individuals afflicted with COVID-19.
Collapse
Affiliation(s)
- Raymond Farah
- Department of Medicine B, Ziv Medical Center, Safed, Israel
- Azrieli Faculty of Medicine, Safed, Bar-Ilan University, Israel
| | - Rola Khamisy-Farah
- Azrieli Faculty of Medicine, Safed, Bar-Ilan University, Israel
- Clalit Health Service, Akko, Israel
| | - Kacper Dziedzic
- Department of Clinical Research and Development, LUXMED Group, Warsaw, Poland
| | - Ewa Skrzypek
- Department of Medical Ethics and History of Medicine, Medical University of Warsaw, Poland.
| | - Michal Pruc
- Department of Public Health, International European University, Kyiv, Ukraine
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
| | | | - Lukasz Szarpak
- Department of Clinical Research and Development, LUXMED Group, Warsaw, Poland
- Henry JN Taub Department of Emergency Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Mahmoud Jomah
- Azrieli Faculty of Medicine, Safed, Bar-Ilan University, Israel
| |
Collapse
|
14
|
Jia S, Zhang J, Wang X, Zhang Z, Wang B, Zhang J, Jiang H, Guo G, Wang Y, Wan J, Wang W, Hou L, Zhu F. Safety and Immunogenicity of Homologous Recombinant Adenovirus Type 5-Vectored COVID-19 Vaccine Booster Dose in Healthy Adults Aged 18-60 Years: a Single-Center, Open-Label Trial. Infect Dis Ther 2023; 12:2757-2769. [PMID: 38038866 PMCID: PMC10746682 DOI: 10.1007/s40121-023-00892-0] [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: 09/07/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION The waning antibody levels several months after prime vaccination and the persistent epidemics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) around the world have generated great interest in the evaluation of a booster dose. We aimed to assess the safety and immunogenicity of a homologous booster dose of the recombinant adenovirus type 5-vectored coronavirus disease 2019 (COVID-19) vaccine (Ad5-nCoV). METHODS In this trial, we recruited healthy adults aged 18-60 years who had received one dose of Ad5-nCoV vaccine (low, middle, or high dose) in the previous phase 1 trial approximately 6 months earlier, and then all participants received a booster dose of 5 × 1010 viral particles (low dose) intramuscularly. The primary outcome was the incidence of adverse reactions within 14 days after booster vaccination. The specific binding antibodies were measured by enzyme-linked immunosorbent assay and the neutralizing antibody responses were assessed with live SARS-CoV-2 and pseudovirus neutralization assay. The cellular immune responses were analyzed by enzyme-linked immunospot assay and intracellular cytokine staining. RESULTS From September 26 to 28, 2020, 108 volunteers were recruited and 89 eligible participants (52% male) were enrolled and received a booster dose of Ad5-nCoV vaccine: 28 (31%) had received a low prime dose, 30 (34%) a middle prime dose, and 31 (35%) a high prime dose in the previous phase 1 trial. All participants were included in the safety analysis and immunogenicity was assessed in 88 (99%) participants. Twenty-three (82%) participants in the low prime dose group, 23 (77%) participants in the middle prime dose group, and 26 (84%) participants in the high prime dose group reported at least one adverse reaction within the first 14 days post booster. Pain at the injection site and fatigue were the most common adverse reactions. Most adverse reactions were mild or moderate in all groups and no vaccine-related severe adverse event was noted within 12 months after booster vaccination. Neutralizing antibodies increased moderately at day 14 and peaked at 28 days post booster. T cell responses were also boosted at 14 days after vaccination. CONCLUSIONS A homologous booster of Ad5-nCoV vaccine is well tolerated and immunogenic in healthy adults aged 18-60 years who had received a priming dose of Ad5-nCoV 6 months previously. The neutralizing antibodies against SARS-CoV-2 peaked at day 28 and specific T cell responses were noted at day 14 after booster. Ad5-nCoV vaccine can be considered as a homologous booster 6 months after a priming dose. TRIAL REGISTRATION ClinicalTrials.gov, NCT04568811.
Collapse
Affiliation(s)
- Siyue Jia
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
| | | | - Xue Wang
- CanSino Biologics, Tianjin, China
| | - Zhe Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Busen Wang
- Beijing Institute of Biotechnology, Beijing, China
| | - Jun Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Hudachuan Jiang
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
| | - Ge Guo
- CanSino Biologics, Tianjin, China
| | | | | | - Wenjuan Wang
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China.
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China.
| | - Fengcai Zhu
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China.
- Center for Global Health, CGH, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Moliva JI, Andrew SF, Flynn BJ, Wagner DA, Foulds KE, Gagne M, Flebbe DR, Lamb E, Provost S, Marquez J, Mychalowych A, Lorag CG, Honeycutt CC, Burnett MR, McCormick L, Henry AR, Godbole S, Davis-Gardner ME, Minai M, Bock KW, Nagata BM, Todd JPM, McCarthy E, Dodson A, Kouneski K, Cook A, Pessaint L, Ry AV, Valentin D, Young S, Littman Y, Boon ACM, Suthar MS, Lewis MG, Andersen H, Alves DA, Woodward R, Leuzzi A, Vitelli A, Colloca S, Folgori A, Raggiolli A, Capone S, Nason MC, Douek DC, Roederer M, Seder RA, Sullivan NJ. Durable immunity to SARS-CoV-2 in both lower and upper airways achieved with a gorilla adenovirus (GRAd) S-2P vaccine in non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.567930. [PMID: 38076895 PMCID: PMC10705562 DOI: 10.1101/2023.11.22.567930] [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: 12/23/2023]
Abstract
SARS-CoV-2 continues to pose a global threat, and current vaccines, while effective against severe illness, fall short in preventing transmission. To address this challenge, there's a need for vaccines that induce mucosal immunity and can rapidly control the virus. In this study, we demonstrate that a single immunization with a novel gorilla adenovirus-based vaccine (GRAd) carrying the pre-fusion stabilized Spike protein (S-2P) in non-human primates provided protective immunity for over one year against the BA.5 variant of SARS-CoV-2. A prime-boost regimen using GRAd followed by adjuvanted S-2P (GRAd+S-2P) accelerated viral clearance in both the lower and upper airways. GRAd delivered via aerosol (GRAd(AE)+S-2P) modestly improved protection compared to its matched intramuscular regimen, but showed dramatically superior boosting by mRNA and, importantly, total virus clearance in the upper airway by day 4 post infection. GrAd vaccination regimens elicited robust and durable systemic and mucosal antibody responses to multiple SARS-CoV-2 variants, but only GRAd(AE)+S-2P generated long-lasting T cell responses in the lung. This research underscores the flexibility of the GRAd vaccine platform to provide durable immunity against SARS-CoV-2 in both the lower and upper airways.
Collapse
Affiliation(s)
- Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Danielle A Wagner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Dillon R Flebbe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Evan Lamb
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Samantha Provost
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Josue Marquez
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Anna Mychalowych
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Cynthia G Lorag
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Matthew R Burnett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Lauren McCormick
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Sucheta Godbole
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Meredith E Davis-Gardner
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, 30322, United States of America
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20892, United States of America
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20892, United States of America
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20892, United States of America
| | - John-Paul M Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Alan Dodson
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Katelyn Kouneski
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Anthony Cook
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Laurent Pessaint
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Alex Van Ry
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Daniel Valentin
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Steve Young
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Yoav Littman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, 63110, United States of America
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, 30322, United States of America
| | - Mark G Lewis
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Hanne Andersen
- Bioqual, Inc., Rockville, Maryland, 20850, United States of America
| | - Derron A Alves
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20892, United States of America
| | - Ruth Woodward
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | | | | | | | | | | | | | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892, United States of America
- Correspondence: and
| | - Nancy J Sullivan
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
- Correspondence: and
- Lead contact
| |
Collapse
|
16
|
Pathakumari B, Marty PK, Shah M, Van Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity Is Highly Heterogenous after SARS-CoV-2 Infection. J Clin Med 2023; 12:7136. [PMID: 38002748 PMCID: PMC10672050 DOI: 10.3390/jcm12227136] [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: 10/08/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, we recruited 30 unvaccinated convalescent donors who had previously been infected with COVID-19 and 7 unexposed asymptomatic controls. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG via semiquantitative ELISA, and T-cell immunity against S1 and S2 subunits were studied via IFN-γ enzyme-linked immunosorbent spot (ELISpot) and flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2-positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and the determination of optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARS-CoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Paige K. Marty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Maleeha Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Virginia P. Van Keulen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Courtney L. Erskine
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Matthew S. Block
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Pedro Arias-Sanchez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Patricio Escalante
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Tobias Peikert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| |
Collapse
|
17
|
Liang Z, Wang N, Fan C, Shang L, Zhang Y, Gao C, Luo J. Disturbance of Adaptive Immunity System Was Accompanied by a Decrease in Plasma Short-Chain Fatty Acid for Patients Hospitalized During SARS-CoV-2 Infection After COVID-19 Vaccination. J Inflamm Res 2023; 16:5261-5272. [PMID: 38026252 PMCID: PMC10656857 DOI: 10.2147/jir.s434860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to disorders of immune function and a decrease in the diversity of intestinal flora. We aimed to explore the changes of circulating immune cell subsets and the plasma level of intestinal short-chain fatty acids (SCFAs) in patients with Coronavirus disease 2019 (COVID-19), further understanding the pathogenesis of COVID-19. Methods The study included 83 newly diagnosed COVID-19 patients and 39 non-COVID-19 controls. All have completed a full course of vaccination against SARS-CoV-2. The levels of peripheral lymphocyte subsets and plasma cytokines were detected by flow cytometry. Targeted metabolomics was used to explore the level of SCFAs in plasma. Results Compared with the non-COVID-19 group, COVID-19 patients showed a decrease in CD19+B cells, CD4+T cells, CD8+T cells, NK cells, CD4+CD8+T cells and CD4-CD8-T cells (all p<0.001) and concomitantly an increase in sIL-2R, IL-6 and IL-10 (all p<0.005). These alterations were more pronounced in those critical patients. In addition, COVID-19 patients had lower levels of propanoic acid (PA), butyric acid (BA), isobutyric acid (IBA) and isohexanoic acid (ICA) (all p<0.01). Among them, the level of ICA is positively correlated with the absolute number of immune cells. Conclusion Our study suggests the immune cell subsets in COVID-19 patients who had completed vaccination were still severely disturbed and concomitantly lower SCFAs, especially in severe patients with poor prognosis. Lower levels of plasma SCFAs may contribute to lymphopenia in COVID-19. The potential relationship between plasma SCFAs and immune cell reduction provides a new direction for the treatment of COVID-19.
Collapse
Affiliation(s)
- Zhaojun Liang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Nan Wang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chunxue Fan
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Lili Shang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yaping Zhang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chong Gao
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Luo
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
18
|
Familiar-Macedo D, Vieira Damasco P, Fiestas Solórzano VE, Carnevale Rodrigues J, Sampaio de Lemos ER, Barreto Dos Santos F, Agudo Mendonça Teixeira de Siqueira M, Leal de Azeredo E, de-Oliveira-Pinto LM. Inflammatory and cytotoxic mediators in COVID-19 patients and in ChAdOx1 nCoV-19 (AZD1222) vaccine recipients. Cytokine 2023; 171:156350. [PMID: 37672863 DOI: 10.1016/j.cyto.2023.156350] [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: 07/14/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
Immunological and cytotoxic mediators are induced in natural infection and are essential for the effectiveness of vaccination. Vaccination is useful to prevent the spread of SARS-CoV-2 and limit the morbidity/mortality of COVID-19. ChAdOx1 nCoV-19 is one of the most widespread vaccines in the world. We compared the detection of anti-S1 SARS-CoV2 IgG and the profile of inflammatory and cytotoxic responses of patients who developed different clinical outcomes of COVID-19 with individuals previously exposed or not to the virus received the first and booster doses of ChAdOx1 nCoV-19. Plasma from 35 patients with COVID-19 and 11 vaccinated were evaluated by multiplex assay. Here, no vaccinated subjects had serious adverse effects. Those vaccinated with a booster dose had higher anti-S1 IgG than mild/moderate and recovered patients. Critically ill and deceased patients had IgG levels like those immunized. By univariate analysis, IL-2, IL-17, and perforin do not differentiate between patients and vaccinated individuals. Granzyme A increased at dose 1, while patients had their levels reduced. High levels of granulysin, sFas, and IL-6 were detected in the deaths, but after vaccination, all were declined. The multivariate analysis supports the role of IL-6 and granulysin as associated and non-confounding variables related to the worst clinical outcome of COVID-19, but not sFas. Our data confirm the ability of the ChAdOx1 vaccine to produce specific antibody levels up to booster time. Furthermore, our data suggest that the vaccine can regulate both the hyper-production and the kinetics of the production of inflammatory and cytotoxic mediators involved in the cytokine storm, such as granulysin and IL-6.
Collapse
Affiliation(s)
- Débora Familiar-Macedo
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Paulo Vieira Damasco
- Rede Casa Hospital Rio Laranjeiras e Rio Botafogo, Rio de Janeiro 22240-000, Brazil
| | - Victor Edgar Fiestas Solórzano
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Joyce Carnevale Rodrigues
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil
| | - Elba Regina Sampaio de Lemos
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Flávia Barreto Dos Santos
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Marilda Agudo Mendonça Teixeira de Siqueira
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Elzinandes Leal de Azeredo
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Luzia Maria de-Oliveira-Pinto
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
19
|
Neto TAP, Sidney J, Grifoni A, Sette A. Correlative CD4 and CD8 T-cell immunodominance in humans and mice: Implications for preclinical testing. Cell Mol Immunol 2023; 20:1328-1338. [PMID: 37726420 PMCID: PMC10616275 DOI: 10.1038/s41423-023-01083-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Antigen-specific T-cell recognition is restricted by Major Histocompatibility Complex (MHC) molecules, and differences between CD4 and CD8 immunogenicity in humans and animal species used in preclinical vaccine testing are yet to be fully understood. In this study, we addressed this matter by analyzing experimentally identified epitopes based on published data curated in the Immune Epitopes DataBase (IEDB) database. We first analyzed SARS-CoV-2 spike (S) and nucleoprotein (N), which are two common targets of the immune response and well studied in both human and mouse systems. We observed a weak but statistically significant correlation between human and H-2b mouse T-cell responses (CD8 S specific (r = 0.206, p = 1.37 × 10-13); CD4 S specific (r = 0.118, p = 2.63 × 10-5) and N specific (r = 0.179, p = 2.55 × 10-4)). Due to intrinsic differences in MHC molecules across species, we also investigated the association between the immunodominance of common Human Leukocyte Antigen (HLA) alleles for which HLA transgenic mice are available, namely, A*02:01, B*07:02, DRB1*01:01, and DRB1*04:01, and found higher significant correlations for both CD8 and CD4 (maximum r = 0.702, p = 1.36 × 10-31 and r = 0.594, p = 3.04-122, respectively). Our results further indicated that some regions are commonly immunogenic between humans and mice (either H-2b or HLA transgenic) but that others are human specific. Finally, we noted a significant correlation between CD8 and CD4 S- (r = 0.258, p = 7.33 × 1021) and N-specific (r = 0.369, p = 2.43 × 1014) responses, suggesting that discrete protein subregions can be simultaneously recognized by T cells. These findings were confirmed in other viral systems, providing general guidance for the use of murine models to test T-cell immunogenicity of viral antigens destined for human use.
Collapse
Affiliation(s)
- Tertuliano Alves Pereira Neto
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| |
Collapse
|
20
|
Lopez-Gomez A, Pelaez-Prestel HF, Juarez I. Approaches to evaluate the specific immune responses to SARS-CoV-2. Vaccine 2023; 41:6434-6443. [PMID: 37770298 DOI: 10.1016/j.vaccine.2023.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
The SARS-CoV-2 pandemic has a huge impact on public health and global economy, meaning an enormous scientific, political, and social challenge. Studying how infection or vaccination triggers both cellular and humoral responses is essential to know the grade and length of protection generated in the population. Nowadays, scientists and authorities around the world are increasingly concerned about the arrival of new variants, which have a greater spread, due to the high mutation rate of this virus. The aim of this review is to summarize the different techniques available for the study of the immune responses after exposure or vaccination against SARS-CoV-2, showing their advantages and limitations, and proposing suitable combinations of different techniques to achieve extensive information in these studies. We wish that the information provided here will helps other scientists in their studies of the immune response against SARS-CoV-2 after vaccination with new vaccine candidates or infection with upcoming variants.
Collapse
Affiliation(s)
- Ana Lopez-Gomez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Hector F Pelaez-Prestel
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| | - Ignacio Juarez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
21
|
Marty PK, Pathakumari B, Shah M, Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity is Highly Heterogenous after SARS-CoV-2 Infection. RESEARCH SQUARE 2023:rs.3.rs-3222112. [PMID: 37674707 PMCID: PMC10479471 DOI: 10.21203/rs.3.rs-3222112/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, between April 23, 2020, to May 11, 2020, we recruited 30 COVID-19 unvaccinated convalescent donors and 7 unexposed asymptomatic donors. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG semiquantitative ELISA and T cell immunity against S1 and S2 subunits were studied by IFN-γ Enzyme-Linked Immune absorbent Spot (ELISpot), flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2 positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARSCoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
|
22
|
España PP, Bilbao-González A, Larrea N, Castillo-Sintes I, García-Gutiérrez S, Portuondo J, Villanueva A, Uranga A, Legarreta MJ, Gascon M, Quintana JM. Impact of prior SARS-COV-2 infection and vaccination on COVID-19 hospital admission and mortality amongst nursing home residents. Aging Clin Exp Res 2023; 35:1771-1778. [PMID: 37249860 PMCID: PMC10228436 DOI: 10.1007/s40520-023-02446-3] [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: 03/13/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Nursing home residents (NHRs) have experienced disproportionately high risk of severe outcomes due to COVID-19 infection. AIM We investigated the impact of COVID-19 vaccinations and previous SARS-CoV-2 episodes in preventing hospitalization and mortality in NHRs. METHODS Retrospective study of a cohort of all NHRs in our area who were alive at the start of the vaccination campaign. The first three doses of SARS-CoV-2 vaccine and prior COVID-19 infections were registered. The main outcomes were hospital admission and mortality during each follow up. Random effects time-varying Cox models adjusted for age, sex, and comorbidities were fitted to estimate hazard ratios (HRs) according to vaccination status. RESULTS COVID-19 hospitalization and death rates for unvaccinated NHRs were respectively 2.39 and 1.42 per 10,000 person-days, falling after administration of the second dose (0.37 and 0.34) and rising with the third dose (1.08 and 0.8). Rates were much lower amongst people who had previously had COVID-19. Adjusted HRs indicated a significant decrease in hospital admission amongst those with a two- and three-dose status; those who had had a previous COVID-19 infection had even lower hospital admission rates. Death rates decreased as NHRs received two and three doses, and the probability of death was much lower among those who had previously had the infection. CONCLUSIONS The effectiveness of current vaccines against severe COVID-19 disease in NHRs remains high and SARS-CoV-2 episodes prior to vaccination entail a major reduction in hospitalization and mortality rates. The protection conferred by vaccines appears to decline in the following months. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04463706.
Collapse
Affiliation(s)
- Pedro P España
- Respiratory Service, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Respiratory Unit, Galdakao-Usansolo University Hospital, Galdakao, Spain.
| | - Amaia Bilbao-González
- Research and Innovation Unit, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Nere Larrea
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Idoia Castillo-Sintes
- Research and Innovation Unit, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Susana García-Gutiérrez
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Janire Portuondo
- Osakidetza Basque Health Service, Sub-Directorate for Primary Care Coordination, Vitoria-Gasteiz, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Ane Villanueva
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Ane Uranga
- Respiratory Service, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Maria J Legarreta
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Maria Gascon
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Jose M Quintana
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| |
Collapse
|
23
|
SUN W, ZHAO Y, LIAO L, ZHAO Z, CHEN S, YAN X, WANG X, CHAO G, ZHOU J. Effectiveness and safety of Xuebijing injection for patients with coronavirus disease 2019: a systematic review and Metaanalysis. J TRADIT CHIN MED 2023; 43:631-639. [PMID: 37454247 PMCID: PMC10320442 DOI: 10.19852/j.cnki.jtcm.20230517.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/08/2022] [Indexed: 07/18/2023]
Abstract
OBJECTIVE To evaluate the effectiveness and safety of Xuebijing injection (XBJ) on coronavirus disease 2019 (COVID-19) in patients. METHODS Related studies on multiple biological databases and websites were searched up to December 11, 2021 without language and publication time restrictions. Review Manager V.5.3 and Stata 14 software were used for data analysis. RESULTS Seven studies were finally included. The Metaanalysis showed that compared with the routine treatment alone, XBJ combined with the routine treatment can reduce the 28day mortality ( = 0.3, 95% : 0.12, 0.74), Creactive protein ( = -12.8, 95% : -23.13, 3.46), erythrocyte sedimentation rate ( = -9.32, 95% : -14.66, -3.98) and interleukin-6 (S = -0.6, 95% : -1.04, -0.17) levels and increase the leukocyte ( = 0.73, 95% : 0.42, 1.04) and lymphocyte count ( = 0.18, 95% : 0.07, 0.29) in peripheral blood; additionally, it has no obvious side effects ( = 1.11, 95% : 0.65, 1.9). There was no evidence that the XBJ combined therapy can improve the nucleic acid conversion rate and computed tomography improvement rate of COVID19 patients. CONCLUSIONS Preliminary evidence suggests that XBJ combined with routine treatment seems to be more effective than routine treatment for patients with COVID19. Limited by the number and quality of included papers, this finding still needs further validation by more studies.
Collapse
Affiliation(s)
- Wu SUN
- 1 Department of Ophthalmology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
- 2 Beijing University of Chinese Medicine, Beijing 100029, China
- 3 Department of Retina, Eye Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Yuwei ZHAO
- 2 Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liang LIAO
- 4 Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Zhonghui ZHAO
- 2 Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shiqi CHEN
- 2 Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoling YAN
- 4 Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Xueyao WANG
- 3 Department of Retina, Eye Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Guojun CHAO
- 3 Department of Retina, Eye Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Jian ZHOU
- 2 Beijing University of Chinese Medicine, Beijing 100029, China
- 4 Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| |
Collapse
|
24
|
Luo H, Yan J, Zhang D, Zhou X. Identification of cuproptosis-related molecular subtypes and a novel predictive model of COVID-19 based on machine learning. Front Immunol 2023; 14:1152223. [PMID: 37533853 PMCID: PMC10393044 DOI: 10.3389/fimmu.2023.1152223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
Background To explicate the pathogenic mechanisms of cuproptosis, a newly observed copper induced cell death pattern, in Coronavirus disease 2019 (COVID-19). Methods Cuproptosis-related subtypes were distinguished in COVID-19 patients and associations between subtypes and immune microenvironment were probed. Three machine algorithms, including LASSO, random forest, and support vector machine, were employed to identify differentially expressed genes between subtypes, which were subsequently used for constructing cuproptosis-related risk score model in the GSE157103 cohort to predict the occurrence of COVID-19. The predictive values of the cuproptosis-related risk score were verified in the GSE163151 cohort, GSE152418 cohort and GSE171110 cohort. A nomogram was created to facilitate the clinical use of this risk score, and its validity was validated through a calibration plot. Finally, the model genes were validated using lung proteomics data from COVID-19 cases and single-cell data. Results Patients with COVID-19 had higher significantly cuproptosis level in blood leukocytes compared to patients without COVID-19. Two cuproptosis clusters were identified by unsupervised clustering approach and cuproptosis cluster A characterized by T cell receptor signaling pathway had a better prognosis than cuproptosis cluster B. We constructed a cuproptosis-related risk score, based on PDHA1, PDHB, MTF1 and CDKN2A, and a nomogram was created, which both showed excellent predictive values for COVID-19. And the results of proteomics showed that the expression levels of PDHA1 and PDHB were significantly increased in COVID-19 patient samples. Conclusion Our study constructed and validated an cuproptosis-associated risk model and the risk score can be used as a powerful biomarker for predicting the existence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hong Luo
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
| | - Jisong Yan
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
| | - Dingyu Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, China
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Xia Zhou
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
25
|
Lautem A, Boedecker-Lips SC, Schneider E, Runkel S, Feist C, Lang H, Weinmann-Menke J, Koch M. The Cellular and Humoral Immune Response to SARS-CoV-2 Messenger RNA Vaccines Is Significantly Better in Liver Transplant Patients Compared with Kidney Transplant Patients. Pathogens 2023; 12:910. [PMID: 37513757 PMCID: PMC10383075 DOI: 10.3390/pathogens12070910] [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: 03/10/2023] [Revised: 06/09/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Patients after organ transplantation have impaired immune response after vaccination against the SARS-CoV-2 virus. So far, published studies have reported quite different response rates to SARS-CoV-2 vaccination, ranging from 15-79% in liver and kidney transplant recipients. Up to one year after the first vaccine dose, we analyzed the humoral and cellular immune response of 21 liver transplant (LTX) patients after vaccination with mRNA vaccines compared with 28 kidney transplant (KTX) patients. We evaluated IgG against the SARS-CoV-2 spike protein as well as SARS-CoV-2 specific T cells using an ELISpot assay that detected IFN-γ- and/or IL-2-expressing T cells. We found a cellular and/or humoral immune response in 100% of the LTX patients compared with 68% of the KTX patients. Antibody titers against the spike protein of SARS-CoV-2 were significantly higher in the LTX group, and significantly more LTX patients had detectable specific IL-2-producing T cells. The immunosuppression applied in our LTX cohort was lower compared with the KTX cohort (14% triple therapy in LTX patients vs. 79% in KTX patients). One year after the first vaccination, breakthrough infections could be detected in 41% of all organ transplant patients. None of those patients suffered from a severe course of COVID-19 disease, indicating that a partial vaccination response seemed to offer protection to immunosuppressed patients. The better immune response of LTX patients after SARS-CoV-2 vaccination might be due to less intense immunosuppressive therapy compared with KTX patients.
Collapse
Affiliation(s)
- Anja Lautem
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Simone Cosima Boedecker-Lips
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Elisa Schneider
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Stefan Runkel
- Blood Transfusion Center, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Christina Feist
- Department of Internal Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Julia Weinmann-Menke
- Department of Nephrology, I. Department of Medicine, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Martina Koch
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| |
Collapse
|
26
|
Oliveira DCD, Spiri BS, Schluga YC, Justus JLP, Lopes Neto FDN, Azambuja APD. Evaluation of lymphocyte count, T-cell subsets and neutrophil-to-lymphocyte ratio as early predictors for severity and outcome of COVID-19 disease-a report from a highly complex hospital in Brazil. Hematol Transfus Cell Ther 2023; 45:330-337. [PMID: 35782909 PMCID: PMC9239916 DOI: 10.1016/j.htct.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/19/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Lymphopenia is a laboratory marker of poor prognosis and severity of disease in the SARS-CoV-2 infection. This study aims to describe the immune profile of a Brazilian population. METHODS A total of 121 consecutive patients with severe acute respiratory syndrome (SARS) were analyzed between April and June 2020. Routine peripheral blood counts and multiparametric flow cytometry were performed on admission to assess lymphocytes and subsets (CD3, CD4, CD8). Demographic, clinical and laboratory data were collected from hospital sources. RESULTS The total of 116 patients included 63 (54.3%) males; 76 (62.8%) COVID-19 patients were divided, based on clinical characteristics and mechanical ventilation (MV) use, into moderate (n = 41; no MV) and severe (n = 35; MV) groups. The control group (n = 40) was comprised of patients with SARS of different etiologies. All patients had lymphopenia, with overall lymphocyte counts and their subsets considerably lower in severe patients, when compared to the moderate and controls. Patients with a high neutrophil-to-lymphocyte ratio (> 15.2) and T-cell lymphopenia (CD3 < 593 cells/µL, CD4 < 326 cells/µL, CD8 < 121 cells/µL) had a higher risk of being intubated and progressing to death. A total of 39 patients (95.1%) in the moderate group and 54.3% (n = 19) in the severe group were discharged; 28 patients died. CONCLUSION Laboratory assessment of the neutrophil/lymphocyte ratio and T-cell subsets may be predictive of mortality and may be useful for stratifying COVID-19 patients.
Collapse
Affiliation(s)
| | - Beatriz Sanada Spiri
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| | - Yara Carolina Schluga
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| | | | | | - Ana Paula de Azambuja
- Hospital de Clínicas da Universidade Federal do Paraná (HC UFPR), Curitiba, PR, Brazil
| |
Collapse
|
27
|
Souan L, Abdel-Razeq H, Al Zughbieh M, Al Badr S, Sughayer MA. Comparative Assessment of the Kinetics of Cellular and Humoral Immune Responses to COVID-19 Vaccination in Cancer Patients. Viruses 2023; 15:1439. [PMID: 37515127 PMCID: PMC10383486 DOI: 10.3390/v15071439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVE The kinetics of immune responses to various SARS-CoV-2 vaccines in cancer patients were investigated. METHODS In total, 57 cancer patients who received BNT162b2-RNA or BBIBP-CorV vaccines were enrolled. Cellular and humoral immunity were assessed at three-time points, before the first vaccine dose and 14-21 days after the first and second doses. Chemiluminescent microparticle immunoassay was used to evaluate SARS-CoV-2 anti-spike IgG response, and QuantiFERON® SARS-CoV-2 kit assessed T-cell response. RESULTS Data showed that cancer patients' CD4+ and CD8+ T cell-median IFN-γ secretion of SARS-CoV-2 antigens increased after the first and second vaccine doses (p = 0.027 and p = 0.042). BNT162b2 vaccinees had significantly higher IFN-γ levels to CD4+ and CD8+ T cell epitopes than BBIBP-CorV vaccinees (p = 0.028). There was a positive correlation between IgG antibody titer and T cell response regardless of vaccine type (p < 0.05). CONCLUSIONS This study is one of the first to investigate cellular and humoral immune responses to SARS-CoV-2 immunization in cancer patients on active therapy after each vaccine dose. COVID-19 immunizations helped cancer patients develop an effective immune response. Understanding the cellular and humoral immune response to COVID-19 in cancer patients undergoing active treatment is necessary to improve vaccines and avoid future SARS pandemics.
Collapse
Affiliation(s)
- Lina Souan
- Laboratory Medicine, Department of Pathology, King Hussein Cancer Center, Amman 11941, Jordan
| | | | - Muna Al Zughbieh
- Laboratory Medicine, Department of Pathology, King Hussein Cancer Center, Amman 11941, Jordan
| | - Sara Al Badr
- Laboratory Medicine, Department of Pathology, King Hussein Cancer Center, Amman 11941, Jordan
| | - Maher A Sughayer
- Laboratory Medicine, Department of Pathology, King Hussein Cancer Center, Amman 11941, Jordan
| |
Collapse
|
28
|
Aleksova M, Todorova Y, Emilova R, Baymakova M, Yancheva N, Andonova R, Zasheva A, Grifoni A, Weiskopf D, Sette A, Nikolova M. Virus-Specific Stem Cell Memory CD8+ T Cells May Indicate a Long-Term Protection against Evolving SARS-CoV-2. Diagnostics (Basel) 2023; 13:diagnostics13071280. [PMID: 37046496 PMCID: PMC10093371 DOI: 10.3390/diagnostics13071280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/15/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Immune memory to SARS-CoV-2 is key for establishing herd immunity and limiting the spread of the virus. The duration and qualities of T-cell-mediated protection in the settings of constantly evolving pathogens remain an open question. We conducted a cross-sectional study of SARS-CoV-2-specific CD4+ and CD8+ T-cell responses at several time points over 18 months (30–750 days) post mild/moderate infection with the aim to identify suitable methods and biomarkers for evaluation of long-term T-cell memory in peripheral blood. Included were 107 samples from 95 donors infected during the periods 03/2020–07/2021 and 09/2021–03/2022, coinciding with the prevalence of B.1.1.7 (alpha) and B.1.617.2 (delta) variants in Bulgaria. SARS-CoV-2-specific IFNγ+ T cells were measured in ELISpot in parallel with flow cytometry detection of AIM+ total and stem cell-like memory (TSCM) CD4+ and CD8+ T cells after in vitro stimulation with peptide pools corresponding to the original and delta variants. We show that, unlike IFNγ+ T cells, AIM+ virus-specific CD4+ and CD8+ TSCM are more adequate markers of T cell memory, even beyond 18 months post-infection. In the settings of circulating and evolving viruses, CD8+ TSCM is remarkably stable, back-differentiated into effectors, and delivers immediate protection, regardless of the initial priming strain.
Collapse
|
29
|
Pavia G, Spagnuolo R, Quirino A, Marascio N, Giancotti A, Simeone S, Cosco C, Tino E, Carrabetta F, Di Gennaro G, Nobile C, Bianco A, Matera G, Doldo P. COVID-19 Vaccine Booster Shot Preserves T Cells Immune Response Based on Interferon-Gamma Release Assay in Inflammatory Bowel Disease (IBD) Patients on Anti-TNFα Treatment. Vaccines (Basel) 2023; 11:vaccines11030591. [PMID: 36992175 DOI: 10.3390/vaccines11030591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Immune-modifying treatment in inflammatory bowel disease (IBD) impairs the humoral response. The role of T lymphocytes in this setting is still unclear. This study aims to assess if a booster shot (third dose) of BNT162b2 mRNA COVID-19 vaccine enhanced the humoral response and elicited cellular immunity in IBD patients on different immuno-therapy regimens compared to healthy controls (HCs). Five months after a booster dose, serological and T-cell responses were assessed. The measurements were described using geometric means with 95% confidence intervals. The differences between study groups were assessed by Mann–Whitney tests. Seventy-seven subjects (n = 53 IBD patients and n = 24 HCs), who were fully vaccinated and not previously SARS-CoV-2 infected, were recruited. Regarding the IBD patients, 19 were affected by Crohn’s disease and 34 by ulcerative colitis. During the vaccination cycle, half of the patients (53%) were on stable treatment with aminosalicylates, and 32% were on biological therapy. No differences in antibody concentrations between IBD patients and HCs, nor T-cell responses, were found. Stratifying IBD patients based on the type of treatment (anti-TNFα agents vs. other treatment regimens), a decrease only in antibody titer (p = 0.008), but not in cellular response, was observed. Even after the COVID-19 vaccine booster dose, the TNFα inhibitors selectively decreased the humoral immune response compared to patients on other treatment regimens. The T-cell response was preserved in all study groups. These findings highlight the importance of evaluating T-cell immune responses following COVID-19 vaccination in a routine diagnostic setting, particularly for immunocompromised cohorts.
Collapse
Affiliation(s)
- Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Rocco Spagnuolo
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Aida Giancotti
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Silvio Simeone
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Cristina Cosco
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Elena Tino
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Federico Carrabetta
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Gianfranco Di Gennaro
- Department of Health Sciences, School of Medicine, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Carmelo Nobile
- Department of Health Sciences, School of Medicine, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Aida Bianco
- Department of Health Sciences, School of Medicine, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Patrizia Doldo
- Unit of Gastroenterology, Department of Clinical and Experimental Medicine, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
30
|
Peterson DR, Baran AM, Bhattacharya S, Branche AR, Croft DP, Corbett AM, Walsh EE, Falsey AR, Mariani TJ. Gene Expression Risk Scores for COVID-19 Illness Severity. J Infect Dis 2023; 227:322-331. [PMID: 34850892 PMCID: PMC8767880 DOI: 10.1093/infdis/jiab568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The correlates of coronavirus disease 2019 (COVID-19) illness severity following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are incompletely understood. METHODS We assessed peripheral blood gene expression in 53 adults with confirmed SARS-CoV-2 infection clinically adjudicated as having mild, moderate, or severe disease. Supervised principal components analysis was used to build a weighted gene expression risk score (WGERS) to discriminate between severe and nonsevere COVID-19. RESULTS Gene expression patterns in participants with mild and moderate illness were similar, but significantly different from severe illness. When comparing severe versus nonsevere illness, we identified >4000 genes differentially expressed (false discovery rate < 0.05). Biological pathways increased in severe COVID-19 were associated with platelet activation and coagulation, and those significantly decreased with T-cell signaling and differentiation. A WGERS based on 18 genes distinguished severe illness in our training cohort (cross-validated receiver operating characteristic-area under the curve [ROC-AUC] = 0.98), and need for intensive care in an independent cohort (ROC-AUC = 0.85). Dichotomizing the WGERS yielded 100% sensitivity and 85% specificity for classifying severe illness in our training cohort, and 84% sensitivity and 74% specificity for defining the need for intensive care in the validation cohort. CONCLUSIONS These data suggest that gene expression classifiers may provide clinical utility as predictors of COVID-19 illness severity.
Collapse
Affiliation(s)
- Derick R Peterson
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| | - Andrea M Baran
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York, USA
| | - Angela R Branche
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, New York, USA
| | - Daniel P Croft
- Division of Pulmonary and Critical Care, Department of Medicine, University of Rochester, Rochester, New York, USA
| | - Anthony M Corbett
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| | - Edward E Walsh
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, New York, USA
- Department of Medicine, Rochester General Hospital, Rochester, New York, USA
| | - Ann R Falsey
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, New York, USA
- Department of Medicine, Rochester General Hospital, Rochester, New York, USA
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York, USA
| |
Collapse
|
31
|
Matkowska-Kocjan A, Owoc-Lempach J, Ludwikowska K, Szenborn F, Moskwa N, Kurek K, Kałwak K, Szenborn L, Ussowicz M. COVID-19 mRNA Vaccine Tolerance and Immunogenicity in Hematopoietic Stem Cell Transplantation Recipients Aged 5-11 Years Old-Non-Randomized Clinical Trial. Vaccines (Basel) 2023; 11:vaccines11010195. [PMID: 36680039 PMCID: PMC9866698 DOI: 10.3390/vaccines11010195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
The SARS-CoV-2 pandemic had a devastating impact on the world’s population in the years 2020−2022. The rapid development of vaccines enabled a reduction in the mortality and morbidity of COVID-19, but there are limited data about their effects on immunocompromised children. The aim of this prospective study was to evaluate the safety and efficacy of the mRNA BNT162b2 (Pfizer/Biontech) vaccine in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. Material and methods: Two cohorts of 34 children after allo-HSCT and 35 healthy children aged 5−11 years were vaccinated with two doses of the mRNA BNT162b2 (10 µg) vaccine. All children were evaluated for adverse effects with electronic surveys and the immunogenicity of the vaccine was assessed with anti-SARS-CoV-2 IgG titer measurements. Results: All reported adverse events (AEs) were classified as mild. The most common AE was pain at the injection site. All the other AEs (both local and systemic) were rarely reported (<15% patients). Both groups showed a similar response in anti-SARS-CoV-2 IgG production. Patients after allo-HSCT that were undergoing immunosuppressive treatment presented a poorer immunological response than patients off of treatment. Time since HSCT, patient age, lymphocyte count, and total IgG concentration did not correlate with initial/post-vaccination anti-SARS-CoV-2 IgG titers. Most patients who were eligible for a third dose of the vaccine had an excellent humoral response observed after two vaccine doses. Conclusions: The COVID-19 mRNA BNT162b2 vaccine is very well tolerated and highly immunogenic in 5−11-year-old children after HSCT. Children >2 years of age after HSCT who did not receive immunosuppressive treatment presented excellent antibody production after two doses of the vaccine, but children on immunosuppression may require a more intense vaccination schedule.
Collapse
Affiliation(s)
- Agnieszka Matkowska-Kocjan
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
- Correspondence:
| | - Joanna Owoc-Lempach
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Kamila Ludwikowska
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Filip Szenborn
- Faculty of Electronics, Wroclaw University of Science and Technology, 50-370 Wrocław, Poland
| | - Natalia Moskwa
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Katarzyna Kurek
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Krzysztof Kałwak
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Leszek Szenborn
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Marek Ussowicz
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| |
Collapse
|
32
|
Montaño-Armendáriz N, Zamudio-Cuevas Y, Fernández-Torres J, Martínez-Flores K, Luján-Juárez IA. [Importance of autoimmunity induced by SARS-CoV-2 and development of post-vaccination autoimmune diseases]. REVISTA ALERGIA MÉXICO 2023; 69:78-88. [PMID: 36928248 DOI: 10.29262/ram.v69i2.1153] [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: 07/26/2022] [Accepted: 10/23/2022] [Indexed: 01/06/2023] Open
Abstract
SARS-CoV-2, a virus belonging to the large family of coronavirus, aroused great interest following the outbreak of this new strain reported in 2019, in Wuhan China. Its clinical spectrum is highly variable, ranging from a self-limited disease to an acute respiratory distress syndrome with systemic clinical manifestations (COVID-19), in which the immune system plays a key role in the pathophysiology of this disease and in its severity; several studies show the prevalence of some autoimmune markers suggesting that they may lead to autoimmune states. The most important strategy worldwide to protect the population was the development of vaccines to induce immunity to severe COVID-19; however, vaccines have also been shown to have the ability to produce autoimmune states in a small percentage of the world's population; nevertheless, the best strategy remains vaccination. The aim of this review is to show the current overview of the mechanisms of SARS-CoV-2-induced autoimmunity and post-vaccination for a better understanding and identification of these in the population. Publications from 2019 to 2022 were reviewed in PubMed as the primary search source.
Collapse
Affiliation(s)
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Ciudad de México, México
| | - Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Ciudad de México, México
| | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Ciudad de México, México
| | | |
Collapse
|
33
|
Dhawan M, Rabaan AA, Fawarah MMA, Almuthree SA, Alsubki RA, Alfaraj AH, Mashraqi MM, Alshamrani SA, Abduljabbar WA, Alwashmi ASS, Ibrahim FA, Alsaleh AA, Khamis F, Alsalman J, Sharma M, Emran TB. Updated Insights into the T Cell-Mediated Immune Response against SARS-CoV-2: A Step towards Efficient and Reliable Vaccines. Vaccines (Basel) 2023; 11:101. [PMID: 36679947 PMCID: PMC9861463 DOI: 10.3390/vaccines11010101] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
The emergence of novel variants of SARS-CoV-2 and their abilities to evade the immune response elicited through presently available vaccination makes it essential to recognize the mechanisms through which SARS-CoV-2 interacts with the human immune response. It is essential not only to comprehend the infection mechanism of SARS-CoV-2 but also for the generation of effective and reliable vaccines against COVID-19. The effectiveness of the vaccine is supported by the adaptive immune response, which mainly consists of B and T cells, which play a critical role in deciding the prognosis of the COVID-19 disease. T cells are essential for reducing the viral load and containing the infection. A plethora of viral proteins can be recognized by T cells and provide a broad range of protection, especially amid the emergence of novel variants of SARS-CoV-2. However, the hyperactivation of the effector T cells and reduced number of lymphocytes have been found to be the key characteristics of the severe disease. Notably, excessive T cell activation may cause acute respiratory distress syndrome (ARDS) by producing unwarranted and excessive amounts of cytokines and chemokines. Nevertheless, it is still unknown how T-cell-mediated immune responses function in determining the prognosis of SARS-CoV-2 infection. Additionally, it is unknown how the functional perturbations in the T cells lead to the severe form of the disease and to reduced protection not only against SARS-CoV-2 but many other viral infections. Hence, an updated review has been developed to understand the involvement of T cells in the infection mechanism, which in turn determines the prognosis of the disease. Importantly, we have also focused on the T cells' exhaustion under certain conditions and how these functional perturbations can be modulated for an effective immune response against SARS-CoV-2. Additionally, a range of therapeutic strategies has been discussed that can elevate the T cell-mediated immune response either directly or indirectly.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, Punjab, India
- Trafford College, Altrincham, Manchester WA14 5PQ, UK
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Mahmoud M. Al Fawarah
- Microbiology Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Souad A. Almuthree
- Department of Infectious Disease, King Abdullah Medical City, Makkah 43442, Saudi Arabia
| | - Roua A. Alsubki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Mutaib M. Mashraqi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Saleh A. Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Wesam A. Abduljabbar
- Department of Medical Laboratory Sciences, Fakeeh College for Medical Science, Jeddah 21134, Saudi Arabia
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Fatimah Al Ibrahim
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Abdulmonem A. Alsaleh
- Clinical Laboratory Science Department, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Faryal Khamis
- Infection Diseases Unit, Department of Internal Medicine, Royal Hospital, Muscat 1331, Oman
| | - Jameela Alsalman
- Infection Disease Unit, Department of Internal Medicine, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 435, Bahrain
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| |
Collapse
|
34
|
Le Nouën C, Nelson CE, Liu X, Park HS, Matsuoka Y, Luongo C, Santos C, Yang L, Herbert R, Castens A, Moore IN, Wilder-Kofie T, Moore R, Walker A, Zhang P, Lusso P, Johnson RF, Garza NL, Via LE, Munir S, Barber DL, Buchholz UJ. Intranasal pediatric parainfluenza virus-vectored SARS-CoV-2 vaccine is protective in monkeys. Cell 2022; 185:4811-4825.e17. [PMID: 36423629 PMCID: PMC9684001 DOI: 10.1016/j.cell.2022.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/07/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022]
Abstract
Pediatric SARS-CoV-2 vaccines are needed that elicit immunity directly in the airways as well as systemically. Building on pediatric parainfluenza virus vaccines in clinical development, we generated a live-attenuated parainfluenza-virus-vectored vaccine candidate expressing SARS-CoV-2 prefusion-stabilized spike (S) protein (B/HPIV3/S-6P) and evaluated its immunogenicity and protective efficacy in rhesus macaques. A single intranasal/intratracheal dose of B/HPIV3/S-6P induced strong S-specific airway mucosal immunoglobulin A (IgA) and IgG responses. High levels of S-specific antibodies were also induced in serum, which efficiently neutralized SARS-CoV-2 variants of concern of alpha, beta, and delta lineages, while their ability to neutralize Omicron sub-lineages was lower. Furthermore, B/HPIV3/S-6P induced robust systemic and pulmonary S-specific CD4+ and CD8+ T cell responses, including tissue-resident memory cells in the lungs. Following challenge, SARS-CoV-2 replication was undetectable in airways and lung tissues of immunized macaques. B/HPIV3/S-6P will be evaluated clinically as pediatric intranasal SARS-CoV-2/parainfluenza virus type 3 vaccine.
Collapse
Affiliation(s)
- Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Christine E Nelson
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong-Su Park
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, MD 20837, USA
| | - Ashley Castens
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, MD 20837, USA
| | - Ian N Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Temeri Wilder-Kofie
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rashida Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - April Walker
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peng Zhang
- Viral Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paolo Lusso
- Viral Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole L Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura E Via
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel L Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ursula J Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Hui AM, Li J, Zhu L, Tang R, Ye H, Lin M, Ge L, Wang X, Peng F, Wu Z, Guo X, Shi Y, Pan H, Zhu J, Song Z, Qiu J, Wang W, Zheng J, Ozhelvaci O, Shpyro S, Bushway M, Derhovanessian E, Kühnle MC, Luxemburger U, Muik A, Shishkova Y, Khondker Z, Hu S, Lagkadinou E, Şahin U, Türeci Ö, Zhu F. Immunogenicity and safety of BNT162b2 mRNA vaccine in Chinese adults: A phase 2 randomised clinical trial. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 29:100586. [PMID: 36120090 PMCID: PMC9472569 DOI: 10.1016/j.lanwpc.2022.100586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND BNT162b2, an mRNA vaccine against COVID-19, is being utilised worldwide, but immunogenicity and safety data in Chinese individuals are limited. METHODS This phase 2, randomised, double-blind, placebo-controlled trial included healthy or medically stable individuals aged 18-85 years enrolled at two clinical sites in China. Participants were stratified by age (≤55 or >55 years) and randomly assigned (3:1) by an independent randomisation professional to receive two doses of intramuscular BNT162b2 30 μg or placebo, administered 21 days apart. Study participants, study personnel, investigators, statisticians, and the sponsor's study management team were blinded to treatment assignment. Primary immunogenicity endpoints were the geometric mean titers (GMTs) of neutralising antibodies to live severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and seroconversion rates (SCR) 1 month after the second dose. Safety assessments included reactogenicity within 14 days of vaccination, adverse events (AEs), and clinical laboratory parameters. Randomised participants who received at least one dose were included in the efficacy and safety analyses on a complete case basis (incomplete/missing data not imputed). Results up to 6 months after the second dose are reported. FINDINGS Overall, 959 participants (all of Han ethnicity) who were recruited between December 5th, 2020 and January 9th, 2021 received at least one injection (BNT162b2, n=720; placebo, n=239). At 1 month after the second dose, the 50% neutralising antibody GMT was 294.4 (95% CI; 281.1-308.4) in the BNT162b2 group and 5.0 (95% CI; 5.0-5.0) in the placebo group. SCRs were 99.7% (95% CI; 99.0%-100.0%) and 0% (95% CI; 0.0%-1.5%), respectively (p<0.0001 vs placebo). Although the GMT of neutralising antibodies in the BNT162b2 group was greatly reduced at 6 months after the second dose, the SCR still remained at 58.8%. BNT162b2-elicited sera neutralised SARS-CoV-2 variants of concern. T-cell responses were detected in 58/73 (79.5%) BNT162b2 recipients. Reactogenicity was mild or moderate in severity and resolved within a few days after onset. Unsolicited AEs were uncommon at 1 month following vaccine administration, and there were no vaccine-related serious AEs at 1 month or 6 months after the second dose. INTERPRETATION BNT162b2 vaccination induced a robust immune response with acceptable tolerability in Han Chinese adults. However, follow-up duration was relatively short and COVID-19 rates were not assessed. Safety data collection is continuing until 12 months after the second dose. FUNDING BioNTech - sponsored the trial. Shanghai Fosun Pharmaceutical Development Inc. (Fosun Pharma) - conducted the trial, funded medical writing. CLINICALTRIALSGOV REGISTRATION NUMBER NCT04649021. Trial status: Completed.
Collapse
Affiliation(s)
| | - Jingxin Li
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
| | - Li Zhu
- Taizhou People's Hospital, Taizhou, China
| | - Rong Tang
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
| | - Huayue Ye
- Taizhou Vaccine Clinical Research Centre, Taizhou, China
| | - Mei Lin
- Taizhou People's Hospital, Taizhou, China
| | - Lei Ge
- Fosun Pharma, Beijing, China
| | | | - Fuzhong Peng
- Taizhou Vaccine Clinical Research Centre, Taizhou, China
| | | | - Xiling Guo
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
| | - Yunfeng Shi
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
| | - Hongxing Pan
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
| | - Jiahong Zhu
- Lianshui Center for Disease Control and Prevention, Lianshui, China
| | - Zhizhou Song
- Lianshui Center for Disease Control and Prevention, Lianshui, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Fengcai Zhu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Centre for Disease Control and Prevention, Nanjing, China
- Centre for Global Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
36
|
Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. Int J Mol Sci 2022; 23:ijms232315122. [PMID: 36499448 PMCID: PMC9737069 DOI: 10.3390/ijms232315122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
COVID-19, a significant global health threat, appears to be an immune-related disease. Failure of effective immune responses in initial stages of infection may contribute to development of cytokine storm and systemic inflammation with organ damage, leading to poor clinical outcomes. Disease severity and the emergence of new SARS-CoV-2 variants highlight the need for new preventative and therapeutic strategies to protect the immunocompromised population. Available data indicate that these people may benefit from adoptive transfer of allogeneic SARS-CoV-2-specific T cells isolated from convalescent individuals. This review first provides an insight into the mechanism of cytokine storm development, as it is directly related to the exhaustion of T cell population, essential for viral clearance and long-term antiviral immunity. Next, we describe virus-specific T lymphocytes as a promising and efficient approach for the treatment and prevention of severe COVID-19. Furthermore, other potential cell-based therapies, including natural killer cells, regulatory T cells and mesenchymal stem cells are mentioned. Additionally, we discuss fast and effective ways of producing clinical-grade antigen-specific T cells which can be cryopreserved and serve as an effective "off-the-shelf" approach for rapid treatment of SARS-CoV-2 infection in case of sudden patient deterioration.
Collapse
|
37
|
Monroe JM, Haralambieva IH, Warner ND, Grill DE, Quach HQ, Kennedy RB. Longitudinal antibody titer, avidity, and neutralizing responses after SARS-CoV-2 infection. Heliyon 2022; 8:e11676. [PMID: 36439767 PMCID: PMC9675084 DOI: 10.1016/j.heliyon.2022.e11676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/05/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
While waning immunity and SARS-CoV-2 variant immune escape continue to result in high infection rates worldwide, associations between longitudinal quantitative, qualitative, and functional humoral immune responses after SARS-CoV-2 infection remain unclear. In this study, we found significant waning of antibody against Spike S1 (R = -0.32, p = 0.035) and N protein (R = -0.39, p = 0.008), while RBD antibody moderately decreased (R = -0.19, p = 0.203). Likewise, neutralizing antibody titer (ND50) waned over time (R = -0.46, p = 0.001). In contrast, antibody avidity increased significantly over time for Spike S1 (R = 0.62, p = 6.0e-06), RBD (R = 0.54, p = 2.0e-04), and N (R = 0.33, p = 0.025) antibodies. Across all humoral responses, ND50 strongly associated with Spike S1 (R = 0.85, p = 2.7e-13) and RBD (R = 0.78, p = 2.9e-10) antibodies. Our findings provide longitudinal insight into humoral immune responses after infection and imply the potential of Spike S1/RBD antibody titer as surrogate correlates of protection.
Collapse
Affiliation(s)
- Jonathon M. Monroe
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Diane E. Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Anatolou D, Dovrolis N, Ragia G, Kolios G, Manolopoulos VG. Unpacking COVID-19 Systems Biology in Lung and Whole Blood with Transcriptomics and miRNA Regulators. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:608-621. [PMID: 36269619 DOI: 10.1089/omi.2022.0104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
COVID-19 is a systemic disease affecting tissues and organs, including and beyond the lung. Apart from the current pandemic context, we also have vastly inadequate knowledge of consequences of repeated exposures to SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), the virus causing COVID-19, in multiple organ systems and the whole organism scales when the disease evolves from a pandemic to an endemic state. This calls for a systems biology and systems medicine approach and unpacking the effects of COVID-19 in lung as well as other tissues. We report here original findings from transcriptomics analyses and differentially expressed genes (DEGs) in lung samples from 60 patients and 27 healthy controls, and in whole blood samples from 255 patients and 103 healthy individuals. A total of 11 datasets with RNA-seq transcriptomic data were obtained from the Gene Expression Omnibus and the European Nucleotide Archive. The identified DEGs were used to construct protein interaction and functional networks and to identify related pathways and miRNAs. We found 35 DEGs common between lung and the whole blood, and importantly, 2 novel genes, namely CYP1B1 and TNFAIP6, which have not been previously implicated with COVID-19. We also identified four novel miRNA potential regulators, hsa-mir-192-5p, hsa-mir-221-3p, hsa-mir-4756-3p, and hsa-mir-10a-5p, implicated in lung or other diseases induced by coronaviruses. In summary, these findings offer new molecular leads and insights to unpack COVID-19 systems biology in a whole organism context and might inform future antiviral drug, diagnostics, and vaccine discovery efforts.
Collapse
Affiliation(s)
- Dimitra Anatolou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Alexandroupolis, Greece
| |
Collapse
|
39
|
Kombe Kombe AJ, Biteghe FAN, Ndoutoume ZN, Jin T. CD8 + T-cell immune escape by SARS-CoV-2 variants of concern. Front Immunol 2022; 13:962079. [PMID: 36389664 PMCID: PMC9647062 DOI: 10.3389/fimmu.2022.962079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/03/2022] [Indexed: 07/30/2023] Open
Abstract
Despite the efficacy of antiviral drug repositioning, convalescent plasma (CP), and the currently available vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the worldwide coronavirus disease 2019 (COVID-19) pandemic is still challenging because of the ongoing emergence of certain new SARS-CoV-2 strains known as variants of concern (VOCs). Mutations occurring within the viral genome, characterized by these new emerging VOCs, confer on them the ability to efficiently resist and escape natural and vaccine-induced humoral and cellular immune responses. Consequently, these VOCs have enhanced infectivity, increasing their stable spread in a given population with an important fatality rate. While the humoral immune escape process is well documented, the evasion mechanisms of VOCs from cellular immunity are not well elaborated. In this review, we discussed how SARS-CoV-2 VOCs adapt inside host cells and escape anti-COVID-19 cellular immunity, focusing on the effect of specific SARS-CoV-2 mutations in hampering the activation of CD8+ T-cell immunity.
Collapse
Affiliation(s)
- Arnaud John Kombe Kombe
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Zélia Nelly Ndoutoume
- The Second Clinical School, Medical Imaging, Chongqing Medical University, Chongqing, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Laboratory of Structural Immunology, Chinese Academic of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Chinese Academic of Sciences (CAS) Center for Excellence in Molecular Cell Science, Chinese Academy of Science, Shanghai, China
| |
Collapse
|
40
|
Yoo JJ, Yon DK, Lee SW, Shin JI, Kim BK. Humoral Immunogenicity to SARS-CoV-2 Vaccination in Liver Transplant Recipients: A Systematic Review and Meta-Analysis. Int J Biol Sci 2022; 18:5849-5857. [PMID: 36263175 PMCID: PMC9576515 DOI: 10.7150/ijbs.77030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/20/2022] [Indexed: 01/12/2023] Open
Abstract
Solid organ transplant recipients generally show reduced immunogenicity to various vaccines. We aimed to assess the immunogenicity of the immune response among orthotopic liver transplant (OLT) recipients after the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. A systematic search was performed to evaluate immunogenicity or adverse events reported after SARS-CoV-2 vaccination. The pooled analysis of 20 studies showed a humoral immune response rate of 0.70 (95% confidence interval [CI], 0.63-0.77) after SARS-CoV-2 vaccination among OLT recipients. The immunogenicity among OLT recipients was significantly lower compared to the overall population and healthy controls, with odds ratios (ORs) of 0.80 and 0.69. However, it was significantly higher than that of patients receiving other organ transplants, especially kidneys, with an OR of 1.50. Male sex, old age, chronic kidney disease, obesity, and multiple or high immunosuppressant doses significantly increased the risk of unresponsiveness in patients with OLT. The overall incidence of any adverse event after vaccination was 0.68 (95% CI, 0.55-0.81), similar to that of control. OLT recipients had an overall humoral immune response rate of 70% after SARS-CoV-2 vaccination, which is lower than that of healthy controls but favourable compared to those of other solid organ transplant recipients.
Collapse
Affiliation(s)
- Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyaung University College of Medicine, Bucheon, Republic of Korea
| | - Dong Keon Yon
- Department of Pediatrics, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Seung Won Lee
- Department of Data Science, Sejong University College of Software Convergence, Seoul, Republic of Korea.,Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea.,✉ Corresponding authors: contributed equally to this work. Jae Il Shin, M.D., Ph.D. Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea. 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. E-mail: ; Beom Kyung Kim, M.D., Ph.D. Department of Internal Medicine, Yonsei University College of Medicine. 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Tel.: +82-(0)2-2228-1930, Fax: +82-(0) 2-393-6884, E-mail:
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Yonsei Liver Center, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea.,✉ Corresponding authors: contributed equally to this work. Jae Il Shin, M.D., Ph.D. Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea. 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. E-mail: ; Beom Kyung Kim, M.D., Ph.D. Department of Internal Medicine, Yonsei University College of Medicine. 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea. Tel.: +82-(0)2-2228-1930, Fax: +82-(0) 2-393-6884, E-mail:
| |
Collapse
|
41
|
McCafferty S, Haque AKMA, Vandierendonck A, Weidensee B, Plovyt M, Stuchlíková M, François N, Valembois S, Heyndrickx L, Michiels J, Ariën KK, Vandekerckhove L, Abdelnabi R, Foo CS, Neyts J, Sahu I, Sanders NN. A dual-antigen self-amplifying RNA SARS-CoV-2 vaccine induces potent humoral and cellular immune responses and protects against SARS-CoV-2 variants through T cell-mediated immunity. Mol Ther 2022; 30:2968-2983. [PMID: 35450821 PMCID: PMC9020838 DOI: 10.1016/j.ymthe.2022.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023] Open
Abstract
Self-amplifying RNA vaccines may induce equivalent or more potent immune responses at lower doses compared to non-replicating mRNA vaccines via amplified antigen expression. In this paper, we demonstrate that 1 μg of an LNP-formulated dual-antigen self-amplifying RNA vaccine (ZIP1642), encoding both the S-RBD and N antigen, elicits considerably higher neutralizing antibody titers against Wuhan-like Beta B.1.351 and Delta B.1.617.2 SARS-CoV-2 variants compared to those of convalescent patients. In addition, ZIP1642 vaccination in mice expanded both S- and N-specific CD3+CD4+ and CD3+CD8+ T cells and caused a Th1 shifted cytokine response. We demonstrate that the induction of such dual antigen-targeted cell-mediated immune response may provide better protection against variants displaying highly mutated Spike proteins, as infectious viral loads of both Wuhan-like and Beta variants were decreased after challenge of ZIP1642 vaccinated hamsters. Supported by these results, we encourage redirecting focus toward the induction of multiple antigen-targeted cell-mediated immunity in addition to neutralizing antibody responses to bypass waning antibody responses and attenuate infectious breakthrough and disease severity of future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Sean McCafferty
- Ziphius Vaccines NV, B-9820 Merelbeke, Belgium; Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
| | | | | | | | | | | | - Nathalie François
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium
| | | | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Johan Michiels
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, B-2000 Antwerp, Belgium
| | - Linos Vandekerckhove
- HIV Cure and Research Center, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Rana Abdelnabi
- University of Leuven, Department of Microbiology, Immunology, and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Caroline S Foo
- University of Leuven, Department of Microbiology, Immunology, and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Johan Neyts
- University of Leuven, Department of Microbiology, Immunology, and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium; Global Virus Network (GVN), Baltimore, MD, USA
| | | | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium; Cancer Research Institute (CRIG), Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
42
|
Fan Z, Duan S, Liu F, Shi W, Yang Z, Bai R, Li T, Chen J, Xie H, Li J, Tang Y. SARS-CoV-2 vaccination in androgen sensitive phenotypes – A study on associated factors for SARS-CoV-2 vaccination and its adverse effects among androgenetic alopecia and benign prostate hyperplasia patients. Front Immunol 2022; 13:919958. [PMID: 36119091 PMCID: PMC9478654 DOI: 10.3389/fimmu.2022.919958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAndrogen sensitivity, which was established as the leading etiology of androgenetic alopecia (AGA) and benign prostatic hyperplasia (BPH), plays an important role in SARS-CoV-2 infection. Vaccination is essential for AGA and BPH patients in view of the high risk from SARS-CoV-2 infection.PurposeWe aimed to investigate the associated factors for SARS-CoV-2 vaccination and its side effects in populations with AGA and BPH.MethodWe collected the data on SARS-CoV-2 vaccination and adverse reactions of male AGA and BPH patients visited the outpatient of Xiangya hospital by telephone and web-based questionnaires. Vaccination rate and adverse reactions were compared by different vaccine types and use of anti-androgen therapy.ResultA total of 457 AGA patients and 397 BPH patients were recruited in this study. Among which, 92.8% AGA patients and 61.0% BPH patients had at least the first dose of SARS-CoV-2 vaccination (p < 0.001). Having comorbidities and use of anti-androgen therapy increased the risk of un-vaccination among AGA by 2.875 and 3.729 times, respectively (p < 0.001). Around 31.1% AGA patients and 9.5% BPH patients presented adverse reactions, which were mostly mild. Anti-androgen therapy increased the inclination of injection site pain after vaccination (18.7% vs 11.9%; OR: 1.708, 95% CI: 1.088-2.683, p = 0.019).ConclusionCo-existence of other systemic diseases and anti-androgen therapy were the limiting factors for SARS-CoV-2 unvaccination, especially in AGA patients. The importance of SARS-CoV-2 vaccines should be strengthened and popularized in androgen sensitive phenotypes.
Collapse
Affiliation(s)
- Zhihua Fan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shixin Duan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Fangfen Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziye Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruiyang Bai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingxian Chen
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yan Tang,
| |
Collapse
|
43
|
Immunogenicity and Safety of the BNT162b2 mRNA COVID-19 Vaccine in Patients with Melanoma Treated with Immunotherapy. Cancers (Basel) 2022; 14:cancers14153791. [PMID: 35954454 PMCID: PMC9367332 DOI: 10.3390/cancers14153791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/25/2022] [Accepted: 07/31/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The efficacy and safety of the BNT126b2 vaccine against SARS-CoV-2 has not been thoroughly studied in cancer patients treated with immunotherapy. This research aims to investigate the efficacy and safety of the vaccine in patients with melanoma under immunotherapy; at the same time, through the immunophenotyping of T cells and myeloid cells of the peripheral blood, it will be possible to look for changes in the subpopulations of such cells after vaccinations. The results of the study help establish the efficacy and safety of the vaccine in this population, especially since a theoretical concern exists about the vaccine triggering irAEs. Abstract The BNT162b2 vaccine against SARS-CoV-2 has a proven efficacy and a favorable safety profile. In cancer patients under immunotherapy in the form of immune-checkpoint inhibitors (ICIs), the efficacy of the vaccine has not been thoroughly studied, while a theoretical concern has also been raised about triggering immune-related adverse events (irAEs) by the vaccine. We conducted a prospective, non-interventional study on the immunogenicity and safety of the BNT162b2 vaccine in patients with advanced or metastatic melanoma treated with ICIs. Blood samples were obtained 0–4 days before the first dose and 12–21 days after the second dose of the vaccine for the quantification of the SARS-CoV-2 anti-spike antibody using an ELISA and immunophenotyping of the T and myeloid cell subpopulations. The active recording of AEs for a two-month period was conducted. Forty patients were included in the study. All but one (97.3%) achieved seroconversion after two doses of the vaccine and no correlations of the antibody titers with any of the studied parameters (age, gender, stage and duration of the disease, type of ICI, previous treatment, etc.) were found. Moreover, no differences in the subpopulations of the T cells (including the T-regulatory cells) or the myeloid cells were found pre- and post-vaccination. All AEs were low-grade, while one case of arthritis exacerbation was noted. The seroconversion rate in the studied population was high and was comparable to that of healthy subjects, while no major safety issues were raised during the safety follow-up. Finally, no derangements in the subpopulations of T cells or myeloid cells were noted. This is the first study focusing on the immunogenicity, safety, and effect of anti-SARS-CoV-2 vaccines on the blood-cell immunophenotype status of patients with melanoma treated with ICIs.
Collapse
|
44
|
Li JX, Wu SP, Guo XL, Tang R, Huang BY, Chen XQ, Chen Y, Hou LH, Liu JX, Zhong J, Pan HX, Shi FJ, Xu XY, Li ZP, Zhang XY, Cui LB, Tan WJ, Chen W, Zhu FC. Safety and immunogenicity of heterologous boost immunisation with an orally administered aerosolised Ad5-nCoV after two-dose priming with an inactivated SARS-CoV-2 vaccine in Chinese adults: a randomised, open-label, single-centre trial. THE LANCET RESPIRATORY MEDICINE 2022; 10:739-748. [PMID: 35605625 PMCID: PMC9122540 DOI: 10.1016/s2213-2600(22)00087-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 11/15/2022]
Abstract
Background Due to waning immunity and protection against infection with SARS-CoV-2, a third dose of a homologous or heterologous COVID-19 vaccine has been proposed by health agencies for individuals who were previously primed with two doses of an inactivated COVID-19 vaccine. Methods We did a randomised, open-label, controlled trial to evaluate the safety and immunogenicity of heterologous boost immunisation with an orally administered aerosolised adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCoV) in Chinese adults (≥18 years old) who had previously received two doses of an inactivated SARS-CoV-2 vaccine—Sinovac CoronaVac. Eligible participants were randomly assigned (1:1:1) to receive a heterologous booster vaccination with a low dose (1·0 × 1011 viral particles per mL; 0·1 mL; low dose group), or a high dose (1·0 × 1011 viral particles per mL; 0·2 mL; high dose group) aerosolised Ad5-nCoV, or a homologous intramuscular vaccination with CoronaVac (0·5 mL). Only laboratory staff were masked to group assignment. The primary endpoint for safety was the incidence of adverse reactions within 14 days after the booster dose. The primary endpoint for immunogenicity was the geometric mean titres (GMTs) of serum neutralising antibodies (NAbs) against live SARS-CoV-2 virus 14 days after the booster dose. This study was registered with ClinicalTrials.gov, NCT05043259. Findings Between Sept 14 and 16, 2021, 420 participants were enrolled: 140 (33%) participants per group. Adverse reactions were reported by 26 (19%) participants in the low dose group and 33 (24%) in the high dose group within 14 days after the booster vaccination, significantly less than the 54 (39%) participants in the CoronaVac group (p<0·0001). The low dose group had a serum NAb GMT of 744·4 (95% CI 520·1–1065·6) and the high dose group had a GMT of 714·1 (479·4–1063·7) 14 days after booster dose, significantly higher than the GMT in the CoronaVac group (78·5 [60·5–101·7]; p<0·0001). Interpretation We found that a heterologous booster vaccine with an orally administered aerosolised Ad5-nCoV is safe and highly immunogenic in adults who have previously received two doses of CoronaVac as the primary series vaccination. Funding National Natural Science Foundation of China and Jiangsu Provincial Key Research and Development Program.
Collapse
Affiliation(s)
- Jing-Xin Li
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China; School of Public Health, Nanjing Medical University, Nanjing, China; Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shi-Po Wu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Xi-Ling Guo
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Rong Tang
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Bao-Ying Huang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Qin Chen
- Donghai County Center for Disease Control and Prevention, Donghai, China
| | - Yin Chen
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Li-Hua Hou
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Jing-Xian Liu
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jin Zhong
- Donghai County Center for Disease Control and Prevention, Donghai, China
| | - Hong-Xing Pan
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Feng-Juan Shi
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | | | - Zhuo-Pei Li
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiao-Yin Zhang
- School of Public Health, Southeast University, Nanjing, China
| | - Lun-Biao Cui
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Wen-Jie Tan
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wei Chen
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China.
| | - Feng-Cai Zhu
- National Health Commission (NHC) Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China; School of Public Health, Nanjing Medical University, Nanjing, China; Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China; School of Public Health, Southeast University, Nanjing, China.
| |
Collapse
|
45
|
Chen Z, Zhang Y, Wang M, Islam MS, Liao P, Hu Y, Chen X. Humoral and Cellular Immune Responses of COVID-19 vaccines against SARS-Cov-2 Omicron variant: a systemic review. Int J Biol Sci 2022; 18:4629-4641. [PMID: 35874952 PMCID: PMC9305266 DOI: 10.7150/ijbs.73583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/21/2022] [Indexed: 12/13/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has undergone multiple mutations since its emergence, and its latest variant, Omicron (B.1.1.529), is the most contagious variant of concern (VOC) which poses a major and imminent threat to public health. Since firstly reported by World Health Organization (WHO) in November 2021, Omicron variant has been spreading rapidly and has become the dominant variant in many countries worldwide. Omicron is the most mutated variant so far, containing 60 mutations in its genome, including 37 mutations in the S-protein. Since all current COVID-19 vaccines in use were developed based on ancestral SARS-CoV-2 strains, whether they are protective against Omicron is a critical question which has been the center of study currently. In this article, we systemically reviewed the studies regarding the effectiveness of 2- or 3-dose vaccines delivered in either homologous or heterologous manner. The humoral and cellular immune responses elicited by various vaccine regimens to protect against Omicron variant are discussed. Current understanding of the molecular basis underlying immune escape of Omicron was also analyzed. These studies indicate that two doses of vaccination are insufficient to elicit neutralizing antibody responses against Omicron variant. Nevertheless, Omicron-specific humoral immune responses can be enhanced by booster dose of almost all type vaccines in certain degree, and heterologous vaccination strategy may represent a better choice than homogenous regimens. Intriguingly, results of studies indicate that all current vaccines are still able to elicit robust T cell response against Omicron. Future focus should be the development of Omicron variant vaccine, which may induce potent humoral as well as cellular immune responses simultaneously against all known variants of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
46
|
Chong P, Yoon BJ, Lai D, Carlson M, Lee J, He S. Looking back on forward-looking COVID models. PATTERNS (NEW YORK, N.Y.) 2022; 3:100492. [PMID: 35845843 PMCID: PMC9278499 DOI: 10.1016/j.patter.2022.100492] [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] [Received: 12/07/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022]
Abstract
Covid Act Now (CAN) developed an epidemiological model that takes various non-pharmaceutical interventions (NPIs) into account and predicts viral spread and subsequent health outcomes. In this study, the projections of the model developed by CAN were back-tested against real-world data, and it was found that the model consistently overestimated hospitalizations and deaths by 25%-100% and 70%-170%, respectively, due in part to an underestimation of the efficacy of NPIs. Other COVID models were also back-tested against historical data, and it was found that all models generally captured the potential magnitude and directionality of the pandemic in the short term. There are limitations to epidemiological models, but understanding these limitations enables these models to be utilized as tools for data-driven decision-making in viral outbreaks. Further, it can be valuable to have multiple, independently developed models to mitigate the inaccuracies of or to correct for the incorrect assumptions made by a particular model.
Collapse
Affiliation(s)
- Paul Chong
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Byung-Jun Yoon
- Texas A&M University, Department of Electrical and Computer Engineering, College Station, TX 77843, USA
- Brookhaven National Laboratory, Computational Science Initiative, Upton, NY 11973-5000, USA
| | - Debbie Lai
- Act Now Coalition, 340 S Lemon Avenue 6683, Walnut, CA 91789-2706, USA
- University College of London, Division of Psychology and Language Sciences, London WC1E 6BT, England, UK
| | - Michael Carlson
- Act Now Coalition, 340 S Lemon Avenue 6683, Walnut, CA 91789-2706, USA
- Northwell Health, New York, NY 10075, USA
| | - Jarone Lee
- Departments of Surgery and Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shuhan He
- Lab of Computer Science, Department of Internal Medicine, Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
47
|
Tarke A, Potesta M, Varchetta S, Fenoglio D, Iannetta M, Sarmati L, Mele D, Dentone C, Bassetti M, Montesano C, Mondelli MU, Filaci G, Grifoni A, Sette A. Early and Polyantigenic CD4 T Cell Responses Correlate with Mild Disease in Acute COVID-19 Donors. Int J Mol Sci 2022; 23:ijms23137155. [PMID: 35806161 PMCID: PMC9267033 DOI: 10.3390/ijms23137155] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022] Open
Abstract
We assessed SARS-CoV-2-specific CD4+ and CD8+ T cell responses in samples from 89 acute COVID-19 patients, utilizing blood samples collected during the first wave of COVID-19 in Italy. The goal of the study was to examine correlations between SARS-CoV-2-specific T cell responses in the early phase comparing mild, moderate, or severe COVID-19 disease outcomes. T cell responses to the spike (S) and non-S proteins were measured in a combined activation-induced marker (AIM) and intracellular cytokine staining (ICS) assay. Early CD4+ T cell responses to SARS-CoV-2 S correlated with milder disease by both AIM and IFNγ ICS readouts. The correlation of S-specific CD4+ T cell responses with milder disease severity was most striking within the first two weeks of symptom onset compared to later time points. Furthermore, donors with milder disease were associated with polyantigenic CD4+ T cell responses that recognized more prominently non-S proteins in addition to S, while severe acute COVID-19 was characterized by lower magnitudes of CD4+ T cell responses and a narrower repertoire. In conclusion, this study highlights that both the magnitude and breadth of early SARS-CoV-2-specific CD4+ T cell responses correlated with milder disease outcomes in acute COVID-19 patients.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Center of Excellence for Biomedical Research (CEBR), Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
| | - Marina Potesta
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.P.); (C.M.)
| | - Stefania Varchetta
- Division of Clinical Immunology and Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.V.); (D.M.); (M.U.M.)
| | - Daniela Fenoglio
- Center of Excellence for Biomedical Research (CEBR), Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (D.F.); (G.F.)
- Bioterapy Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marco Iannetta
- Department of System Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (L.S.)
| | - Loredana Sarmati
- Department of System Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (L.S.)
| | - Dalila Mele
- Division of Clinical Immunology and Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.V.); (D.M.); (M.U.M.)
| | - Chiara Dentone
- Infectious Diseases Unit, Polyclinic San Martino Hospital-IRCCS, 16132 Genoa, Italy; (C.D.); (M.B.)
| | - Matteo Bassetti
- Infectious Diseases Unit, Polyclinic San Martino Hospital-IRCCS, 16132 Genoa, Italy; (C.D.); (M.B.)
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy
| | - Carla Montesano
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.P.); (C.M.)
| | - Mario U. Mondelli
- Division of Clinical Immunology and Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.V.); (D.M.); (M.U.M.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Gilberto Filaci
- Center of Excellence for Biomedical Research (CEBR), Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (D.F.); (G.F.)
- Bioterapy Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Correspondence: (A.G.); (A.S.)
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Correspondence: (A.G.); (A.S.)
| |
Collapse
|
48
|
Trajectory patterns of SARS-CoV-2 neutralising antibody response in convalescent COVID-19 patients. COMMUNICATIONS MEDICINE 2022; 2:53. [PMID: 35603297 PMCID: PMC9120513 DOI: 10.1038/s43856-022-00119-2] [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: 10/01/2021] [Accepted: 04/26/2022] [Indexed: 11/22/2022] Open
Abstract
Background The adaptive immune responses of COVID-19 patients contributes to virus clearance, restoration of health and protection from re-infection. The patterns of and the associated characteristics with longitudinal neutralising antibody (NAb) response following SARS-CoV-2 infection are important in their potential association with the population risks of re-infection. Methods This is a longitudinal study with blood samples and clinical data collected in adults aged 18 or above following diagnosis of SARS-CoV-2 infection. NAb levels were measured by the SARS-CoV-2 surrogate virus neutralisation test (sVNT). Anonymous clinical and laboratory data were matched with surveillance data for each subject for enabling analyses and applying latent class mixed models for trajectory delineation. Logistic regression models were performed to compare the characteristics between the identified classes. Results In 2020–2021, 368 convalescent patients in Hong Kong are tested for NAb. Their seroconversion occur within 3 months in 97% symptomatic patients, the level of which are maintained at 97% after 9 months. The NAb trajectories of 200 symptomatic patients are classified by the initial response and subsequent trend into high-persistent and waning classes in latent class mixed models. High-persistent (15.5%) class patients are older and most have chronic illnesses. Waning class patients (84.5%) are largely young adults who are mildly symptomatic including 2 who serorevert after 10 months. Conclusions Characteristic sub-class variabilities in clinical pattern are noted especially among patients with waning NAb. The heterogeneity of the NAb trajectory patterns and their clinical association can be important for informing vaccination strategy to prevent re-infection. Neutralising antibodies are produced by the immune system and help to defend against viruses like SARS-CoV-2, which causes COVID-19. Declining levels of these antibodies over time might be linked to risk of re-infection with the virus. Here, we look at changes in neutralising antibody levels over time in people who have had COVID-19. We define two classes of people: those with persistently high levels of antibodies over time, who are more likely to be older and have chronic illnesses, and those with declining antibody levels, who are younger and had mild COVID-19 symptoms. Understanding differences in how these antibodies are maintained over time in different groups of people might help to guide vaccination strategies to prevent re-infection. Wong, Lee et al. analyse trajectory patterns in the neutralising antibody response to SARS-CoV-2 in convalescent COVID-19 patients. The authors identify two major classes of patients—high-persistent and waning—reporting specific clinical characteristics of each class, which could help with targeted vaccination strategies.
Collapse
|
49
|
Nouën CL, Nelson CE, Liu X, Park HS, Matsuoka Y, Luongo C, Santos C, Yang L, Herbert R, Castens A, Moore IN, Wilder-Kofie T, Moore R, Walker A, Zhang P, Lusso P, Johnson RF, Garza NL, Via LE, Munir S, Barber D, Buchholz UJ. Intranasal pediatric parainfluenza virus-vectored SARS-CoV-2 vaccine candidate is protective in macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.21.492923. [PMID: 35665011 PMCID: PMC9164439 DOI: 10.1101/2022.05.21.492923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pediatric SARS-CoV-2 vaccines are needed that elicit immunity directly in the airways, as well as systemically. Building on pediatric parainfluenza virus vaccines in clinical development, we generated a live-attenuated parainfluenza virus-vectored vaccine candidate expressing SARS-CoV-2 prefusion-stabilized spike (S) protein (B/HPIV3/S-6P) and evaluated its immunogenicity and protective efficacy in rhesus macaques. A single intranasal/intratracheal dose of B/HPIV3/S-6P induced strong S-specific airway mucosal IgA and IgG responses. High levels of S-specific antibodies were also induced in serum, which efficiently neutralized SARS-CoV-2 variants of concern. Furthermore, B/HPIV3/S-6P induced robust systemic and pulmonary S-specific CD4+ and CD8+ T-cell responses, including tissue-resident memory cells in lungs. Following challenge, SARS-CoV-2 replication was undetectable in airways and lung tissues of immunized macaques. B/HPIV3/S-6P will be evaluated clinically as pediatric intranasal SARS-CoV-2/parainfluenza virus type 3 vaccine.
Collapse
Affiliation(s)
- Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Christine E. Nelson
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Hong-Su Park
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Poolesville, MD 20837, USA
| | - Ashley Castens
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Poolesville, MD 20837, USA
| | - Ian N. Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- Current address: Division of Pathology, Yerkes National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - Temeri Wilder-Kofie
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- Current Address: Division of Assurances, Office of Laboratory Animal Welfare, National Institutes of Health, MD 20892, USA
| | - Rashida Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- Current address: Yerkes National Primate Research Center, Environmental Health and Safety Office, Emory University; Atlanta, GA, 30322, USA
| | - April Walker
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Peng Zhang
- Viral Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Paolo Lusso
- Viral Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Reed F. Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Nicole L. Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Laura E. Via
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Daniel Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Ursula J. Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
- These authors contributed equally to this work
| |
Collapse
|
50
|
Valarezo-Sevilla D, Sarzosa-Terán V. The enemy is the virus, not the vaccine. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.02.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
If we remember very superficially a little about the complex human immune system, a part of this immune system is made up of leukocytes (especially phagocytes and lymphocytes) that are responsible for detecting invaders and sending them defensive markers that adhere to them (the antibodies) and also to destroy the invaders identified by the system; In addition to the already mentioned leukocytes, our immune system is made up of various other elements such as cells, proteins, tissues and organs, which defend us against germs and microorganisms. 1, 2
Speaking specifically about the immune system and COVID-19, the events mainly involved in the immunopathogenesis of COVID-19 and the dysregulation of the immune response include lymphopenia and increased neutrophil-to-lymphocyte ratio, cytokines, lymphocyte depletion and dysfunction, antibody-dependent enhancement, and monocyte and granulocyte abnormalities. It must be emphasized that lymphopenia is an essential finding in most patients with COVID-19, particularly in those with a severe phenotype.3 It has been shown that multiple viral proteins induce T cells after infection and that T cells that have antiviral signatures related to safety and protection can be achieved by vaccination.4
Collapse
|