1
|
Augello M, Bono V, Rovito R, Tavelli A, Santoro A, Tincati C, Vergori A, Azzini AM, Righi E, Spiteri G, Porru S, Meschi S, Notari S, Maggi F, Antinori A, Tacconelli E, d'Arminio Monforte A, Marchetti G. Long-term immune responses to SARS-CoV-2 Omicron BA.4/5 mRNA booster in people living with HIV. COMMUNICATIONS MEDICINE 2025; 5:92. [PMID: 40148493 PMCID: PMC11950219 DOI: 10.1038/s43856-025-00799-6] [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: 05/17/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Variant-adapted vaccines are recommended in vulnerable populations to address the waning immunity and the emergence of immune-escaping SARS-CoV-2 variants, yet data about immune responses to such vaccines in people living with HIV (PLWH) are limited. We therefore aimed to assess long-term immune responses to an original-BA.4/5 mRNA booster in this population. METHODS In this prospective longitudinal study, PLWH receiving either an original-BA.4/5 bivalent booster or an original monovalent booster and HIV-negative healthcare workers (HCWs) receiving a bivalent booster were enrolled and sampled before (T0), 1 month (T1), and 4-9 months (T2) after the vaccine administration. SARS-CoV-2-specific T and B cells, RBD-binding antibodies, and RBD-blocking antibodies against both wild type (WT) and omicron BA.4/5 virus were determined. RESULTS The bivalent booster is able to transiently increase both humoral and polyfunctional T cell responses in PLWH, with humoral responses comparable to those observed in HCWs. While T cell responses are cross-reactive against viral variants and stable over time, humoral immunity is imprinted to the ancestral virus and wanes quickly. Furthermore, whilst previous SARS-CoV-2 infection does not affect the trajectory of vaccine-elicited immune responses, markers of HIV-related T cell dysfunction are associated with lower antibody peak responses and higher antibody waning. Lastly, the bivalent booster was superior to the monovalent one in inducing BA.4/5-reactive RBD-blocking antibodies. CONCLUSIONS The original-BA.4/5 bivalent booster is highly immunogenic in PLWH and superior to the monovalent one in inducing humoral responses against the BA.4/5 virus, although HIV-related T cell dysfunction markers are associated with blunted and less durable antibody immunity.
Collapse
Affiliation(s)
- Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Valeria Bono
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Roberta Rovito
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Andrea Santoro
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Camilla Tincati
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Alessandra Vergori
- Viral Immunodeficiencies Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Anna Maria Azzini
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Elda Righi
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Gianluca Spiteri
- Occupational Medicine Unit, Verona Hospital, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefano Porru
- Occupational Medicine Unit, Verona Hospital, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Stefania Notari
- Laboratory of Cellular Immunology and Pharmacology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Andrea Antinori
- Viral Immunodeficiencies Unit, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
2
|
Fredericks MN, Kolodner Z, Waalkes A, Sawatzki K, Hao L, Long DR, Penewit K, Midkiff CC, McCormick CJ, Beraki S, Edlefsen PT, Barrow J, Greninger AL, Gale M, Blair RV, Salipante SJ, Fuller DH, O’Connor MA. SIV/SARS-CoV-2 co-infection in rhesus macaques impacts viral shedding, host immunity, the microbiome, and viral evolution. RESEARCH SQUARE 2025:rs.3.rs-6033850. [PMID: 40195984 PMCID: PMC11975012 DOI: 10.21203/rs.3.rs-6033850/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
People living with HIV (PLWH) have an increased risk of severe COVID-19, including prolonged viral shedding and emergence of mutations. To investigate the simian immunodeficiency virus (SIV) macaque model for HIV/SARS-CoV-2 co-infection, seven SIV+ rhesus macaques were co-infected with SARS-CoV-2. COVID-19 in all macaques was mild. SARS-CoV-2 replication persisted in the upper, but not the lower respiratory tract for 14 days post-infection. Animals showed impaired generation of anti-SARS-CoV-2 antibodies and T-cells. Animals also displayed transient changes in microbial communities in the upper airway and gastrointestinal tract. Evidence of SARS-CoV-2 evolution was observed in the upper respiratory tract. This study demonstrates that SIV/SARS-CoV-2 co-infection in rhesus macaques recapitulates aspects of COVID-19 in PLWH. We show that SIV impairs anti-SARS-CoV-2 immunity, potentially leading to prolonged viral shedding, altered pathogenesis, and viral evolution. This highlights the importance of HIV status in COVID-19 and supports the use of this model for HIV/SARS-CoV-2 co-infection.
Collapse
Affiliation(s)
- Megan N. Fredericks
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle, Washington, United States of America
| | - Zohar Kolodner
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle, Washington, United States of America
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kaitlin Sawatzki
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Linhui Hao
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Cecily C. Midkiff
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Carter J. McCormick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Semira Beraki
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle, Washington, United States of America
| | - Paul T. Edlefsen
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jeana Barrow
- Washington National Primate Research Center, Seattle, Washington, United States of America
| | - Alexander L. Greninger
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Michael Gale
- Washington National Primate Research Center, Seattle, Washington, United States of America
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology and Immunology, University of Minnesota Twin Cities, United States
| | - Robert V. Blair
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Deborah H Fuller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle, Washington, United States of America
| | - Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
3
|
Liu WD, Lin MS, Sun HY, Shih MC, Chuang YC, Huang YS, Lin KY, Li GC, Wu PY, Chen LY, Liu WC, Su YC, He PC, Chen YT, Lin CY, Cheng YC, Yao Y, Yeh YC, Liu CC, Pan MY, Luo YZ, Chang HY, Wang JT, Sheng WH, Hsieh SM, Chang SY, Hung CC. Effectiveness and evolution of anti-SARS-CoV-2 spike protein titers after three doses of COVID-19 vaccination in people with HIV. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:554-563. [PMID: 38429206 DOI: 10.1016/j.jmii.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/20/2024] [Accepted: 02/16/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Real-world vaccine effectiveness following the third dose of vaccination against SARS-CoV-2 remains less investigated among people with HIV (PWH). METHODS PWH receiving the third dose of BNT162b2 and mRNA-1273 (either 50- or 100-μg) were enrolled. Participants were followed for 180 days until the fourth dose of COVID-19 vaccination, SARS-CoV-2 infection, seroconversion of anti-nucleocapsid IgG, death, or loss to follow-up. Anti-spike IgG was determined every 1-3 months. RESULTS Of 1427 participants undergoing the third-dose COVID-19 vaccination, 632 (44.3%) received 100-μg mRNA-1273, 467 (32.8%) 50-μg mRNA-1273, and 328 (23.0%) BNT162b2 vaccine and the respective rate of SARS-CoV-2 infection or seroconversion of anti-nucleocapsid IgG was 246.1, 280.8 and 245.2 per 1000 person-months of follow-up (log-rank test, p = 0.28). Factors associated with achieving anti-S IgG titers >1047 BAU/mL included CD4 count <200 cells/mm3 (adjusted odds ratio [aOR], 0.11; 95% CI, 0.04-0.31), plasma HIV RNA >200 copies/mL (aOR, 0.27; 95% CI, 0.09-0.80), having achieved anti-spike IgG >141 BAU/mL within 3 months after primary vaccination (aOR, 3.69; 95% CI, 2.68-5.07), receiving BNT162b2 vaccine as the third dose (aOR, 0.20; 95% CI, 0.10-0.41; reference, 100-μg mRNA-1273), and having previously received two doses of mRNA vaccine in primary vaccination (aOR, 2.46; 95% CI, 1,75-3.45; reference, no exposure to mRNA vaccine). CONCLUSIONS PWH receiving different types of the third dose of COVID-19 vaccine showed similar vaccine effectiveness against SARS-CoV-2 infection. An additional dose with 100-μg mRNA-1273 could generate a higher antibody response than with 50-μg mRNA-1273 and BNT162b2 vaccine.
Collapse
Affiliation(s)
- Wang-Da Liu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan.
| | - Meng-Shuan Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Hsin-Yun Sun
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Ming-Chieh Shih
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Yu-Chung Chuang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yu-Shan Huang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Kuan-Yin Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Center of Infection Control, National Taiwan University Hospital, Taipei, Taiwan.
| | - Guei-Chi Li
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pei-Ying Wu
- Center of Infection Control, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ling-Ya Chen
- Center of Infection Control, National Taiwan University Hospital, Taipei, Taiwan.
| | - Wen-Chun Liu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yi-Ching Su
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pu-Chi He
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yi-Ting Chen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chia-Yi Lin
- Department of Nursing, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yu-Chen Cheng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yi Yao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yi-Chen Yeh
- Department of Nursing, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chia-Chi Liu
- Department of Nursing, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Mei-Yan Pan
- Department of Nursing, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yu-Zhen Luo
- Center of Infection Control, National Taiwan University Hospital, Taipei, Taiwan.
| | - Hsi-Yen Chang
- Center of Infection Control, National Taiwan University Hospital, Taipei, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan.
| | - Wang-Huei Sheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; School of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Szu-Min Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chien-Ching Hung
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County, Taiwan; Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
4
|
Jongkees MJ, Tan NH, Geers D, de Vries RD, GeurtsvanKessel CH, Hensley KS, Sablerolles RS, Bogers S, Gommers L, Blakaj B, Miranda Afonso P, Hansen BE, Rijnders BJ, Brinkman K, van der Kuy PHM, Roukens AH, Rokx C. Immunogenicity of a bivalent BA.1 COVID-19 booster vaccine in people with HIV in the Netherlands. AIDS 2024; 38:1355-1365. [PMID: 38788210 PMCID: PMC11216395 DOI: 10.1097/qad.0000000000003933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE We evaluated the immunogenicity of a bivalent BA.1 COVID-19 booster vaccine in people with HIV (PWH). DESIGN Prospective observational cohort study. METHODS PWH aged ≥45 years received Wuhan-BA.1 mRNA-1273.214 and those <45 years Wuhan-BA.1 BNT162b2. Participants were propensity score-matched 1 : 2 to people without HIV (non-PWH) by age, primary vaccine platform (mRNA-based or vector-based), number of prior COVID-19 boosters and SARS-CoV-2 infections, and spike (S1)-specific antibodies on the day of booster administration. The primary endpoint was the geometric mean ratio (GMR) of ancestral S1-specific antibodies from day 0 to 28 in PWH compared to non-PWH. Secondary endpoints included humoral responses, T-cell responses and cytokine responses up to 180 days post-vaccination. RESULTS Forty PWH received mRNA-1273.214 ( N = 35) or BNT162b2 ( N = 5) following mRNA-based ( N = 29) or vector-based ( N = 11) primary vaccination. PWH were predominantly male (87% vs. 26% of non-PWH) and median 57 years [interquartile range (IQR) 53-59]. Their median CD4 + T-cell count was 775 (IQR 511-965) and the plasma HIV-RNA load was <50 copies/ml in 39/40. The GMR of S1-specific antibodies by 28 days post-vaccination was comparable between PWH [4.48, 95% confidence interval (CI) 3.24-6.19] and non-PWH (4.07, 95% CI 3.42-4.83). S1-specific antibody responses were comparable between PWH and non-PWH up to 180 days, and T-cell responses up to 90 days post-vaccination. Interferon-γ, interleukin (IL)-2, and IL-4 cytokine concentrations increased 28 days post-vaccination in PWH. CONCLUSION A bivalent BA.1 booster vaccine was immunogenic in well treated PWH, eliciting comparable humoral responses to non-PWH. However, T-cell responses waned faster after 90 days in PWH compared to non-PWH.
Collapse
Affiliation(s)
- Marlou J. Jongkees
- Department of Internal Medicine, Section Infectious Diseases, and Department of Medical Microbiology and Infectious Diseases
| | | | | | | | | | - Kathryn S. Hensley
- Department of Internal Medicine, Section Infectious Diseases, and Department of Medical Microbiology and Infectious Diseases
| | | | | | | | | | - Pedro Miranda Afonso
- Department of Biostatistics and Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Bettina E. Hansen
- Department of Biostatistics and Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Institute of Health Policy, Management and Evaluation, University of Toronto
- Toronto Centre for Liver Disease, Toronto General Hospital University Health Network, Toronto, Canada
| | - Bart J.A. Rijnders
- Department of Internal Medicine, Section Infectious Diseases, and Department of Medical Microbiology and Infectious Diseases
| | - Kees Brinkman
- Department of Internal Medicine and Infectious Diseases, OLVG Hospital, Amsterdam
| | | | - Anna H.E. Roukens
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, the Netherlands
| | - Casper Rokx
- Department of Internal Medicine, Section Infectious Diseases, and Department of Medical Microbiology and Infectious Diseases
| |
Collapse
|
5
|
Huygens S, GeurtsvanKessel C, Gharbharan A, Bogers S, Worp N, Boter M, Bax HI, Kampschreur LM, Hassing RJ, Fiets RB, Levenga H, Afonso PM, Koopmans M, Rijnders BJA, Oude Munnink BB. Clinical and Virological Outcome of Monoclonal Antibody Therapies Across SARS-CoV-2 Variants in 245 Immunocompromised Patients: A Multicenter Prospective Cohort Study. Clin Infect Dis 2024; 78:1514-1521. [PMID: 38445721 PMCID: PMC11175671 DOI: 10.1093/cid/ciae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Immunocompromised patients (ICPs) have an increased risk for a severe and prolonged COVID-19. SARS-CoV-2 monoclonal antibodies (mAbs) were extensively used in these patients, but data from randomized trials that focus on ICPs are lacking. We evaluated the clinical and virological outcome of COVID-19 in ICPs treated with mAbs across SARS-CoV-2 variants. METHODS In this multicenter prospective cohort study, we enrolled B-cell- and/or T-cell-deficient patients treated with casirivimab/imdevimab, sotrovimab, or tixagevimab/cilgavimab. SARS-CoV-2 RNA was quantified and sequenced weekly, and time to viral clearance, viral genome mutations, hospitalization, and death rates were registered. RESULTS Two hundred and forty five patients infected with the Delta (50%) or Omicron BA.1, 2, or 5 (50%) variant were enrolled. Sixty-seven percent were vaccinated; 78 treated as outpatients, of whom 2 required hospital admission, but both survived. Of the 159 patients hospitalized at time of treatment, 43 (27%) required mechanical ventilation or died. The median time to viral clearance was 14 days (interquartile range, 7-22); however, it took >30 days in 15%. Resistance-associated spike mutations emerged in 9 patients in whom the median time to viral clearance was 63 days (95% confidence interval, 57-69; P < .001). Spike mutations were observed in 1 of 42 (2.4%) patients after treatment with 2 active mAbs, in 5 of 34 (14.7%) treated with actual monotherapy (sotrovimab), and 3 of 20 (12%) treated with functional monotherapy (ie, tixagevimab/cilgavimab against tixagevimab-resistant variant). CONCLUSIONS Despite treatment with mAbs, morbidity and mortality of COVID-19 in ICPs remained substantial. Combination antiviral therapy should be further explored and may be preferred in severely ICPs.
Collapse
Affiliation(s)
- Sammy Huygens
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Corine GeurtsvanKessel
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Arvind Gharbharan
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Susanne Bogers
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Nathalie Worp
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marjan Boter
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Hannelore I Bax
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Linda M Kampschreur
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Robert-Jan Hassing
- Department of Internal Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Roel B Fiets
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - Henriette Levenga
- Department of Internal Medicine, Groene Hart Gouda, Gouda, The Netherlands
| | - Pedro Miranda Afonso
- Department of Biostatistics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Bas B Oude Munnink
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Montesi G, Augello M, Polvere J, Marchetti G, Medaglini D, Ciabattini A. Predicting humoral responses to primary and booster SARS-CoV-2 mRNA vaccination in people living with HIV: a machine learning approach. J Transl Med 2024; 22:432. [PMID: 38715088 PMCID: PMC11077794 DOI: 10.1186/s12967-024-05147-1] [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: 01/24/2024] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND SARS-CoV-2 mRNA vaccines are highly immunogenic in people living with HIV (PLWH) on effective antiretroviral therapy (ART). However, whether viro-immunologic parameters or other factors affect immune responses to vaccination is debated. This study aimed to develop a machine learning-based model able to predict the humoral response to mRNA vaccines in PLWH and to assess the impact of demographic and clinical variables on antibody production over time. METHODS Different machine learning algorithms have been compared in the setting of a longitudinal observational study involving 497 PLWH, after primary and booster SARS-CoV-2 mRNA vaccination. Both Generalized Linear Models and non-linear Models (Tree Regression and Random Forest) were trained and tested. RESULTS Non-linear algorithms showed better ability to predict vaccine-elicited humoral responses. The best-performing Random Forest model identified a few variables as more influential, within 39 clinical, demographic, and immunological factors. In particular, previous SARS-CoV-2 infection, BMI, CD4 T-cell count and CD4/CD8 ratio were positively associated with the primary cycle immunogenicity, yet their predictive value diminished with the administration of booster doses. CONCLUSIONS In the present work we have built a non-linear Random Forest model capable of accurately predicting humoral responses to SARS-CoV-2 mRNA vaccination, and identifying relevant factors that influence the vaccine response in PLWH. In clinical contexts, the application of this model provides promising opportunities for predicting individual vaccine responses, thus facilitating the development of vaccination strategies tailored for PLWH.
Collapse
Affiliation(s)
- Giorgio Montesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| |
Collapse
|
7
|
Ferrari L, Ruggiero A, Stefani C, Benedetti L, Piermatteo L, Andreassi E, Caldara F, Zace D, Pagliari M, Ceccherini-Silberstein F, Jones C, Iannetta M, Geretti AM. Utility of accessible SARS-CoV-2 specific immunoassays in vaccinated adults with a history of advanced HIV infection. Sci Rep 2024; 14:8337. [PMID: 38594459 PMCID: PMC11003986 DOI: 10.1038/s41598-024-58597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/01/2024] [Indexed: 04/11/2024] Open
Abstract
Accessible SARS-CoV-2-specific immunoassays may inform clinical management in people with HIV, particularly in case of persisting immunodysfunction. We prospectively studied their application in vaccine recipients with HIV, purposely including participants with a history of advanced HIV infection. Participants received one (n = 250), two (n = 249) or three (n = 42) doses of the BNT162b2 vaccine. Adverse events were documented through questionnaires. Sample collection occurred pre-vaccination and a median of 4 weeks post-second dose and 14 weeks post-third dose. Anti-spike and anti-nucleocapsid antibodies were measured with the Roche Elecsys chemiluminescence immunoassays. Neutralising activity was evaluated using the GenScript cPass surrogate virus neutralisation test, following validation against a Plaque Reduction Neutralization Test. T-cell reactivity was assessed with the Roche SARS-CoV-2 IFNγ release assay. Primary vaccination (2 doses) was well tolerated and elicited measurable anti-spike antibodies in 202/206 (98.0%) participants. Anti-spike titres varied widely, influenced by previous SARS-CoV-2 exposure, ethnicity, intravenous drug use, CD4 counts and HIV viremia as independent predictors. A third vaccine dose significantly boosted anti-spike and neutralising responses, reducing variability. Anti-spike titres > 15 U/mL correlated with neutralising activity in 136/144 paired samples (94.4%). Three participants with detectable anti-S antibodies did not develop cPass neutralising responses post-third dose, yet displayed SARS-CoV-2 specific IFNγ responses. SARS-CoV-2 vaccination is well-tolerated and immunogenic in adults with HIV, with responses improving post-third dose. Anti-spike antibodies serve as a reliable indicator of neutralising activity. Discordances between anti-spike and neutralising responses were accompanied by detectable IFN-γ responses, underlining the complexity of the immune response in this population.
Collapse
Affiliation(s)
- Ludovica Ferrari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Department of Infectious Diseases, Fondazione PTV, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Alessandra Ruggiero
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, Verona, Italy
| | - Chiara Stefani
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, Verona, Italy
| | - Livia Benedetti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Eleonora Andreassi
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Federica Caldara
- Department of Infectious Diseases, Fondazione PTV, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Drieda Zace
- Department of Infectious Diseases, Fondazione PTV, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Matteo Pagliari
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale Delle Venezie, Legnaro, Italy
| | | | - Christopher Jones
- Department of Primary Care and Public Health, Brighton and Sussex Medical School, Falmer, UK
| | - Marco Iannetta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Department of Infectious Diseases, Fondazione PTV, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Anna Maria Geretti
- Department of Infectious Diseases, Fondazione PTV, University of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy.
- Department of Infection, North Middlesex University Hospital, London, UK.
- School of Immunity & Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
8
|
Knudsen ML, Nielsen SD, Heftdal LD. Immune responses to mRNA-based vaccines given as a third COVID-19 vaccine dose in people living with HIV-a literature review. APMIS 2024; 132:236-244. [PMID: 38275143 DOI: 10.1111/apm.13379] [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: 05/10/2023] [Accepted: 01/03/2024] [Indexed: 01/27/2024]
Abstract
People living with HIV (PLWH) were not included in the first efficacy studies of mRNA vaccines against SARS-CoV-2. In this literature review, we investigate evidence of humoral and cellular immunity after a third dose of an mRNA vaccine in PLWH. We performed a literature search in PubMed, Embase, Web of Science and SCOPUS published between 1 January 2020 and 31 December 2022. Selection criteria were studies on immunological responses in PLWH, who were given an mRNA-based vaccine as a third vaccine dose against SARS-CoV-2. Eight articles complied with our selection criteria. All studies found a strong humoral response after the third dose. Five studies investigated cellular immunity and found an increased cellular response after the third vaccine dose in PLWH. No difference in humoral response was observed between PLWH and controls after three doses. However, some of the studies suggested a weaker cellular response among PLWH than in controls, which was associated with lower nadir or current CD4+ T-cell counts. In conclusion, we found evidence of strong humoral immunity in PLWH after receiving an mRNA-based COVID-19 vaccine as a third dose, while the cellular immunity may be impaired compared to controls.
Collapse
Affiliation(s)
- Maria Lodberg Knudsen
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Line Dam Heftdal
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Swan CL, Dushimiyimana V, Ndishimye P, Buchanan R, Yourkowski A, Semafara S, Nsanzimana S, Francis ME, Thivierge B, Lew J, Facciuolo A, Gerdts V, Falzarano D, Sjaarda C, Kelvin DJ, Bitunguhari L, Kelvin AA. Third COVID-19 vaccine dose boosts antibody function in Rwandans with high HIV viral load. iScience 2023; 26:107959. [PMID: 37810226 PMCID: PMC10558770 DOI: 10.1016/j.isci.2023.107959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/18/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) causing COVID-19 (coronavirus disease 2019) poses a greater health risk to immunocompromized individuals including people living with HIV (PLWH). However, most studies on PLWH have been conducted in higher-income countries. We investigated the post-vaccination antibody responses of PLWH in Rwanda by collecting peripheral blood from participants after receiving a second or third COVID-19 vaccine. Virus-binding antibodies as well as antibody neutralization ability against all major SARS-CoV-2 variants of concern were analyzed. We found that people with high HIV viral loads and two COVID-19 vaccine doses had lower levels of binding antibodies that were less virus neutralizing and less cross-reactive compared to control groups. A third vaccination increased neutralizing antibody titers. Our data suggest that people with high HIV viral loads require a third dose of vaccine to neutralize SARS-CoV-2 virus and new variants as they emerge.
Collapse
Affiliation(s)
- Cynthia L. Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | | | - Pacifique Ndishimye
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- African Institute for Mathematical Sciences, Kigali, Rwanda
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Sage Semafara
- Rwanda Network of the People living with HIV (RRP+), Kigali, Rwanda
| | | | - Magen E. Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Brittany Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Jocelyne Lew
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Antonio Facciuolo
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Calvin Sjaarda
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
- Queen’s Genomics Lab at Ongwanada (Q-GLO), Ongwanada Resource Centre, Kingston, ON K7M 8A6, Canada
| | - David J. Kelvin
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
10
|
Cheng MQ, Li R, Weng ZY, Song G. Immunogenicity and effectiveness of COVID-19 booster vaccination among people living with HIV: a systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1275843. [PMID: 37877024 PMCID: PMC10591097 DOI: 10.3389/fmed.2023.1275843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Background The effect of booster vaccinations with the coronavirus virus disease (COVID-19) vaccine on people living with HIV (PLWH) remains unknown. In this study, we aimed to investigate the immunogenicity and effectiveness of booster doses of the COVID-19 vaccine in PLWH. Methods Literature research was done through the PubMed, Embase, Cochrane Review, and Web of Science databases up to 4 July 2023. Pooled estimates were calculated and compared using the DerSimonian and Laird method for a random effects model. Randomized control trials and observational studies were both considered for inclusion. Results We included 35 eligible studies covering 30,154 PLWH. The pooled immune response rate (IRR) of PLWH after the COVID-19 booster vaccination was 97.25% (95% confidence interval [CI], 93.81-99.49), and similar to healthy control (HC) (risk ratio [RR] = 0.98, 95% CI, 0.96-1.00). The pooled IRR for PLWH with CD4+ T-cell counts ≤ 200 was 86.27 (95% CI, 65.35-99.07). For Omicron variants, the pooled IRR for PLWH after booster dose was 74.07% (95% CI, 58.83-89.30), and the risk of IRR was reduced by 10% in PLWH compared with HC (RR = 0.90, 95% CI, 0.80-1.00). The T-cell immune response of PLWH was found to be comparable to HC (p ≥ 0.05). Subgroup analyses revealed that mRNA vaccines produced a relatively high IRR in PLWH compared to other vaccines. In addition, the results showed that booster vaccination appeared to further reduce the risk of COVID-19-related infections, hospitalizations, and deaths compared with the primary vaccination. Conclusion It was shown that booster vaccination with the COVID-19 vaccine provided a high IRR in PLWH and still produced a desirable moderate IRR in PLWH with a CD4+ T-cell count of ≤ 200. Importantly, the humoral and T-cell responses to booster vaccination in PLWH were comparable to HC, and similar results were observed with the SARS-CoV-2 Omicron variant. Our review strongly emphasizes the effect of mRNA vaccine booster vaccination in PLWH on eliciting desirable protective IRR. Furthermore, booster vaccination appears to further reduce the risk of COVID-19 infection, hospitalization, and death in PLWH compared to primary vaccination. However, more evidence is needed to confirm its effectiveness.
Collapse
Affiliation(s)
- Meng-Qun Cheng
- Department of Reproductive Medicine, The Puer People's Hospital, Pu'er, China
| | - Rong Li
- Department of Pharmacy, The Puer People's Hospital, Pu'er, China
| | - Zhi-Ying Weng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Gao Song
- Department of Pharmacy, The Puer People's Hospital, Pu'er, China
| |
Collapse
|
11
|
Wang Y, Qiao Y, Huo Y, Wang L, Liang S, Yu M, Lan X, Song M, Zhang X, Yan Y, Xu J. The safety and immunogenicity of a two-dose schedule of CoronaVac, and the immune persistence of vaccination for six months, in people living with HIV: A multicenter prospective cohort study. Front Immunol 2023; 14:1129651. [PMID: 36993947 PMCID: PMC10040764 DOI: 10.3389/fimmu.2023.1129651] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Background People living with HIV (PLWH) are more vulnerable to SARS-CoV-2. However, evidence on the immunogenicity of coronavirus disease 2019 (COVID-19) vaccines in this population is insufficient. The objective of this study is to assess the immunogenicity and safety of the two-dose schedule of Sinovac CoronaVac for 6 months postvaccination in PLWH. Methods We conducted a multicenter prospective cohort study among PLWH and HIV-negative adults in China. Participants who received two doses of CoronaVac prior to the recruitment were allocated into two groups and followed up for 6 months. The neutralizing antibodies (nAbs), immunoglobulin G against the receptor-binding domain of the spike protein (S-IgG), and gamma-interferon (IFN-γ) were measured to assess the associations among CoronaVac immunogenicity and related factors. Adverse reactions were collected to evaluate the safety profile of vaccination. Results A total of 203 PLWH and 100 HIV-negative individuals were enrolled. A small portion of participants reported mild or moderate adverse reactions without serious adverse events. Median nAbs level in PLWH (31.96 IU/mL, IQR: 12.34-76.40) was lower than that in the control group (46.52 IU/mL, IQR: 29.08-77.30) at the 2-4 weeks postvaccination (P=0.002), and the same trend was presented for median S-IgG titer (37.09 vs. 60.02 IU/ml) (both P <0.05). The nAbs seroconversion rate in the PLWH group was also lower than in the control group (75.86% vs. 89.00%). After then, the immune responses reduced over time in term of only 23.04% of PLWH and 36.00% of HIV-negative individuals had a positive seroconversion for nAbs at 6-month. The multivariable generalized estimating equation analysis showed that PLWH with CD4+T count≥350 cells/µL presented higher immune response than PLWH with CD4+T count <350 cells/µL in terms of antibody seroconversion and titers. The immunogenicity did not differ in participants with low or high HIV viral load. The S-antigen specific IFN-γ immunity was generally stable and had a slow attenuation in both two groups for 6 months postvaccination. Conclusion The Sinovac CoronaVac was generally safe and immunogenic in PLWH, but the immunity response was inferior and the antibodies vanished faster compared to HIV-negative individuals. This study suggested a shorter than 6-month interval of prime-boost vaccination for PLWH to ensure a better protection.
Collapse
Affiliation(s)
- Yuxiao Wang
- Clinical Research Academy, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Ying Qiao
- Department of infection, The Second Hospital of Huhhot, Huhhot, China
| | - Yuqi Huo
- Translational Medicine Research Center, The Sixth People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Li Wang
- Department of Infection, Heilongjiang Provincial Hospital, Heilongjiang, Harbin, China
| | - Shijie Liang
- Department of infectious disease prevention, Zhengzhou Centers for Disease Control and Prevention, Zhengzhou, China
| | - Maohe Yu
- Department of HIV prevention, Tianjin Centers for Disease Control and Prevention, Tianjin, China
| | - Xinquan Lan
- Department of Epidemiology, China Medical University, Shenyang, China
| | - Moxin Song
- Department of Epidemiology, China Medical University, Shenyang, China
| | - Xiangjun Zhang
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Junjie Xu
- Clinical Research Academy, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|