1
|
Pardo I, Maezato AM, Callado GY, Gutfreund MC, Hsieh MK, Lin V, Kobayashi T, Salinas JL, Subramanian A, Edmond MB, Diekema DJ, Rizzo LV, Marra AR. Effectiveness of heterologous and homologous COVID-19 vaccination among immunocompromised individuals: a systematic literature review and meta-analysis. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2024; 4:e152. [PMID: 39346662 PMCID: PMC11427957 DOI: 10.1017/ash.2024.369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 10/01/2024]
Abstract
Objectives We assessed the effectiveness of heterologous vaccination strategy in immunocompromised individuals regarding COVID-19 outcomes, comparing it to homologous approaches. Design Systematic literature review/meta-analysis. Methods We searched PubMed, CINAHL, EMBASE, Cochrane Central Register of Controlled Trials, Scopus, and Web of Science from January 1, 2020 to September 29, 2023. We included studies that evaluated the heterologous vaccination strategy on immunocompromised individuals through outcomes related to COVID-19 (levels of anti-SARS-CoV-2 spike protein IgG, neutralizing antibodies, symptomatic COVID-19 infection, hospitalization, and death) in comparison to homologous schemes. We also used random-effect models to produce pooled odds ratio estimates. Heterogeneity was investigated with I2 estimation. Results Eighteen studies met the inclusion criteria for this systematic review. Fourteen of them provided quantitative data for inclusion in the meta-analysis on vaccine response, being four of them also included in the vaccine effectiveness meta-analysis. The vaccination strategies (heterologous vs homologous) showed no difference in the odds of developing anti-SARS-CoV-2 spike protein IgG (odds ratio 1.12 [95% Cl: 0.73-1.72]). Heterologous schemes also showed no difference in the production of neutralizing antibodies (odds ratio 1.48 [95% Cl: 0.72-3.05]) nor vaccine effectiveness in comparison to homologous schemes (odds ratio 1.52 [95% CI: 0.66-3.53]). Conclusions Alternative heterologous COVID-19 vaccinations have shown equivalent antibody response rates and vaccine effectiveness to homologous schemes, potentially aiding global disparity of vaccine distribution.
Collapse
Affiliation(s)
- Isabele Pardo
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Aline Miho Maezato
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Gustavo Yano Callado
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Maria Celidonio Gutfreund
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Mariana Kim Hsieh
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Vivian Lin
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Takaaki Kobayashi
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jorge L. Salinas
- Division of Infectious Diseases & Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Aruna Subramanian
- Division of Infectious Diseases & Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Michael B. Edmond
- Department of Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Daniel J. Diekema
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Luiz Vicente Rizzo
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Alexandre R. Marra
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Parikh UM, Heaps AL, Moisi D, Gordon KC, Mellors JW, Choudhary MC, Deo R, Moser C, Klekotka P, Landay AL, Currier JS, Eron JJ, Chew KW, Smith DM, Li JZ, Sieg SF. Comparison Study of the Bio-Plex and Meso Scale Multiplexed SARS-CoV-2 Serology Assays Reveals Evidence of Diminished Host Antibody Responses to SARS-CoV-2 after Monoclonal Antibody Treatment. Pathog Immun 2024; 9:58-78. [PMID: 39165724 PMCID: PMC11335343 DOI: 10.20411/pai.v9i2.715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/18/2024] [Indexed: 08/22/2024] Open
Abstract
Background Assessing the breadth and duration of antigen-specific binding antibodies provides valuable information for evaluating interventions to treat or prevent SARS-CoV-2 infection. Multiplex immunoassays are a convenient method for rapid measurement of antibody responses but can sometimes provide discordant results, and antibody positive percent agreement for COVID-19 diagnosis can vary depending on assay type, disease severity, and population sampled. Therefore, we compared two assays marked for research applications, MSD and Bio-Plex Pro, to evaluate qualitative interpretation of serostatus and quantitative detection of antibodies of varying isotypes (IgG, IgM, and IgA) against receptor binding domain (RBD) and nucleocapsid (N) antigens. Methods Specimens from ACTIV-2/A5401, a placebo-controlled clinical trial of the SARSCoV-2 monoclonal antibody (mAb) bamlanivimab to prevent COVID-19 disease progression, were used to evaluate the concordance of the Bio-Rad Bio-Plex Pro Human SARS-CoV-2 Serology Assay and the Meso Scale Discovery (MSD) V-PLEX COVID-19 Panel 1 serology assay in detecting and quantifying IgG, IgA, and IgM binding anti-SARS-CoV-2 antibody responses against the RBD and N antigens. Data were disaggregated by study arm, bamlanivimab dose, days post-enrollment, and presence of emerging resistance. Results We observed 90.5% (412 of 455 tests) concordance for anti-RBD IgG and 87% (396 of 455) concordance for anti-N IgG in classifying samples as negative or positive based on assay-defined cutoffs. Antibody levels converted to the WHO standard BAU/mL were significantly correlated for all isotypes (IgG, IgM, and IgA) and SARS-CoV-2 antigen targets (RBD and N) tested that were common between the two assays (Spearman r 0.65 to 0.92, P < 0.0001). Both assays uncovered evidence of diminished host-derived IgG immune responses in participants treated with bamlanivimab compared to placebo. Assessment of immune responses in the four individuals treated with the 700 mg of bamlanivimab with emerging mAb resistance demonstrated a stronger anti-N IgG response (MSD) at day 28 (median 2.18 log BAU/mL) compared to participants treated with bamlanivimab who did not develop resistance (median 1.55 log BAU/mL). Conclusions These data demonstrate the utility in using multiplex immunoassays for characterizing the immune responses with and without treatment in a study population and provide evidence that monoclonal antibody treatment in acute COVID-19 may have a modest negative impact on development of host IgG responses.
Collapse
Affiliation(s)
- Urvi M. Parikh
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Amy L. Heaps
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | | | - Manish C. Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Carlee Moser
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | | | - Alan L. Landay
- Department of Internal Medicine, Division of Geriatrics and Palliative Medicine, RUSH Medical College, Chicago, IL
| | - Judith S. Currier
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Davey M. Smith
- Department of Medicine, University of California, San Diego, CA
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Scott F. Sieg
- Case Western Reserve University and University Hospitals Cleveland, Cleveland, OH
| | - Team ACTIV-2/A5401 Study
- ACTIV-2/A5401 Study Team: David Smith, Kara Chew, Eric Daar, David Wohl, Judith Currier, Joseph Eron, Arzhang Cyrus Javan, Michael Hughes, Carlee Moser, Justin Ritz, Mark Giganti, Lara Hosey, Jhoanna Roa, Nilam Patel, Kelly Colsh, Irene Rwakazina, Justine Beck, Scott Sieg, Jonathan Li, Courtney Fletcher, William Fischer, Teresa Evering, Rachel Bender Ignacio, Sandra Cardoso, Katya Corado, Prasanna Jagannathan, Nikolaus Jilg, Alan Perelson, Sandy Pillay, Cynthia Riviere, Upinder Singh, Babafemi Taiwo, Joan Gottesman, Matthew Newell, Susan Pedersen, Joan Dragavon, Cheryl Jennings, Brian Greenfelder, William Murtaugh, Jan Kosmyna, Morgan Gapara, Akbar Shahkolahi
| |
Collapse
|
3
|
Gazeau S, Deng X, Brunet-Ratnasingham E, Kaufmann DE, Larochelle C, Morel PA, Heffernan JM, Davis CL, Smith AM, Jenner AL, Craig M. Using virtual patient cohorts to uncover immune response differences in cancer and immunosuppressed COVID-19 patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.605860. [PMID: 39131351 PMCID: PMC11312602 DOI: 10.1101/2024.08.01.605860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) resulted in millions of deaths globally. Adults with immunosuppression (e.g., solid organ transplant recipients) and those undergoing active cancer treatments experience worse infections and more severe COVID-19. It is difficult to conduct clinical studies in these populations, resulting in a restricted amount of data that can be used to relate mechanisms of immune dysfunction to COVID-19 outcomes in these vulnerable groups. To study immune dynamics after infection with SARS-CoV-2 and to investigate drivers of COVID-19 severity in individuals with cancer and immunosuppression, we adapted our mathematical model of the immune response during COVID-19 and generated virtual patient cohorts of cancer and immunosuppressed patients. The cohorts of plausible patients recapitulated available longitudinal clinical data collected from patients in Montréal, Canada area hospitals. Our model predicted that both cancer and immunosuppressed virtual patients with severe COVID-19 had decreased CD8+ T cells, elevated interleukin-6 concentrations, and delayed type I interferon peaks compared to those with mild COVID-19 outcomes. Additionally, our results suggest that cancer patients experience higher viral loads (however, with no direct relation with severity), likely because of decreased initial neutrophil counts (i.e., neutropenia), a frequent toxic side effect of anti-cancer therapy. Furthermore, severe cancer and immunosuppressed virtual patients suffered a high degree of tissue damage associated with elevated neutrophils. Lastly, parameter values associated with monocyte recruitment by infected cells were found to be elevated in severe cancer and immunosuppressed patients with respect to the COVID-19 reference group. Together, our study highlights that dysfunction in type I interferon and CD8+ T cells are key drivers of immune dysregulation in COVID-19, particularly in cancer patients and immunosuppressed individuals.
Collapse
Affiliation(s)
- Sonia Gazeau
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | - Xiaoyan Deng
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | | | - Daniel E. Kaufmann
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital (CHUV) and Université de Lausanne, Lausanne, Switzerland
| | - Catherine Larochelle
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jane M. Heffernan
- Centre for Disease Modelling, Department of Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Courtney L. Davis
- Natural Science Division, Pepperdine University, Malibu, California, USA
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Adrianne L. Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
4
|
Mateo-Urdiales A, Fabiani M, Mayer F, Sacco C, Belleudi V, Da Cas R, Fotakis EA, De Angelis L, Cutillo M, Petrone D, Morciano C, Cannone A, Del Manso M, Riccardo F, Bella A, Menniti-Ippolito F, Pezzotti P, Spila Alegiani S, Massari M. Risk of breakthrough infection and hospitalisation after COVID-19 primary vaccination by HIV status in four Italian regions during 2021. BMC Public Health 2024; 24:1569. [PMID: 38862939 PMCID: PMC11165887 DOI: 10.1186/s12889-024-19071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND As of 2024, vaccination remains the main mitigation measure against COVID-19, but there are contradictory results on whether people living with HIV (PLWH) are less protected by vaccines than people living without HIV (PLWoH). In this study we compared the risk of SARS-CoV-2 infection and COVID-19 hospitalisation following full vaccination in PLWH and PLWoH. METHODS We linked data from the vaccination registry, the COVID-19 surveillance system and from healthcare/pharmacological registries in four Italian regions. We identified PLWH fully vaccinated (14 days post completion of the primary cycle) and matched them at a ratio of 1:4 with PLWoH by week of vaccine administration, age, sex, region of residence and comorbidities. Follow-up started on January 24, 2021, and lasted for a maximum of 234 days. We used the Kaplan-Meier estimator to calculate the cumulative incidence of infection and COVID-19 hospitalisation in both groups, and we compared risks using risk differences and ratios taking PLWoH as the reference group. RESULTS We matched 42,771 PLWH with 171,084 PLWoH. The overall risk of breakthrough infection was similar in both groups with a rate ratio (RR) of 1.10 (95% confidence interval (CI):0.80-1.53). The absolute difference between groups at the end of the study period was 8.28 events per 10,000 person-days in the PLWH group (95%CI:-18.43-40.29). There was a non-significant increase the risk of COVID-19 hospitalisation among PLWH (RR:1.90; 95%CI:0.93-3.32) which corresponds to 6.73 hospitalisations per 10,000 individuals (95%CI: -0.57 to 14.87 per 10,000). CONCLUSIONS Our findings suggest PLWH were not at increased risk of breakthrough SARS-CoV-2 infection or COVID-19 hospitalisation following a primary cycle of mRNA vaccination.
Collapse
Affiliation(s)
| | - Massimo Fabiani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Flavia Mayer
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Sacco
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- European Programme on Intervention Epidemiology Training (EPIET), European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Valeria Belleudi
- Department of Epidemiology, Lazio Regional Health Service, ASL Roma 1, Rome, Italy
| | - Roberto Da Cas
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Emmanouil Alexandros Fotakis
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- European Programme on Intervention Epidemiology Training (EPIET), European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Luigi De Angelis
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Cutillo
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Petrone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Morciano
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cannone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Del Manso
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Flavia Riccardo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Antonino Bella
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Patrizio Pezzotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Marco Massari
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
5
|
Kamboj M, Bohlke K, Baptiste DM, Dunleavy K, Fueger A, Jones L, Kelkar AH, Law LY, LeFebvre KB, Ljungman P, Miller ED, Meyer LA, Moore HN, Soares HP, Taplitz RA, Woldetsadik ES, Kohn EC. Vaccination of Adults With Cancer: ASCO Guideline. J Clin Oncol 2024; 42:1699-1721. [PMID: 38498792 PMCID: PMC11095883 DOI: 10.1200/jco.24.00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 03/20/2024] Open
Abstract
PURPOSE To guide the vaccination of adults with solid tumors or hematologic malignancies. METHODS A systematic literature review identified systematic reviews, randomized controlled trials (RCTs), and nonrandomized studies on the efficacy and safety of vaccines used by adults with cancer or their household contacts. This review builds on a 2013 guideline by the Infectious Disease Society of America. PubMed and the Cochrane Library were searched from January 1, 2013, to February 16, 2023. ASCO convened an Expert Panel to review the evidence and formulate recommendations. RESULTS A total of 102 publications were included in the systematic review: 24 systematic reviews, 14 RCTs, and 64 nonrandomized studies. The largest body of evidence addressed COVID-19 vaccines. RECOMMENDATIONS The goal of vaccination is to limit the severity of infection and prevent infection where feasible. Optimizing vaccination status should be considered a key element in the care of patients with cancer. This approach includes the documentation of vaccination status at the time of the first patient visit; timely provision of recommended vaccines; and appropriate revaccination after hematopoietic stem-cell transplantation, chimeric antigen receptor T-cell therapy, or B-cell-depleting therapy. Active interaction and coordination among healthcare providers, including primary care practitioners, pharmacists, and nursing team members, are needed. Vaccination of household contacts will enhance protection for patients with cancer. Some vaccination and revaccination plans for patients with cancer may be affected by the underlying immune status and the anticancer therapy received. As a result, vaccine strategies may differ from the vaccine recommendations for the general healthy adult population vaccine.Additional information is available at www.asco.org/supportive-care-guidelines.
Collapse
Affiliation(s)
- Mini Kamboj
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY
| | - Kari Bohlke
- American Society of Clinical Oncology, Alexandria, VA
| | | | - Kieron Dunleavy
- MedStar Georgetown University Hospital, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC
| | - Abbey Fueger
- The Leukemia and Lymphoma Society, Rye Brook, NY
| | - Lee Jones
- Fight Colorectal Cancer, Arlington, VA
| | - Amar H Kelkar
- Harvard Medical School, Dana Farber Cancer Institute, Boston, MA
| | | | | | - Per Ljungman
- Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Eric D Miller
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Larissa A Meyer
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Heloisa P Soares
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| | | | | | - Elise C Kohn
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, MD
| |
Collapse
|
6
|
Yadegarynia D, Keyvanfar A, Keyvani H, Tehrani S, Sali S, Abolghasemi S. Immunogenicity and safety of a quadrivalent recombinant influenza vaccine manufactured in Iran (FluGuard) in volunteers aged 18-60 years: A double-blind, non-inferiority, randomized controlled trial. Vaccine 2024; 42:2254-2259. [PMID: 38423811 DOI: 10.1016/j.vaccine.2024.02.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/17/2024] [Accepted: 02/24/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND This study aimed to evaluate the non-inferiority of the FluGuard (a quadrivalent recombinant vaccine manufactured by Nivad Pharmed Salamat Company in Iran) by comparing its immunogenicity and safety with Vaxigrip Tetra (a quadrivalent inactivated vaccine manufactured by Sanofi Pasteur in France). MATERIALS AND METHODS In this double-blind, randomized controlled trial, eligible volunteers aged 18-60 were randomized to receive either FluGuard or Vaxigrip Tetra vaccines. Immunogenicity was evaluated using the Hemagglutination Inhibition (HAI) assay and reported with the geometric mean titer (GMT), seroprotection, and seroconversion. In addition, vaccine safety was assessed by interviewing participants through phone calls. RESULTS Out of 110 randomized volunteers, 51 and 53 were entered into the final analysis in the Vaxigrip and FluGuard groups, respectively. Vaxigrip had a higher seroprotection rate for the H1N1 strain compared with FluGuard (98 % vs. 91 %). Besides, FluGuard had higher seroprotection rates for H3N2 (74 % vs. 69 %), B-Yamagata (87 % vs. 84 %), and B-Victoria (66 % vs. 41 %) strains compared with Vaxigrip. In all four strains, FluGuard was non-inferior to Vaxigrip with the upper bounds of the 95 % CI on the ratio of the GMTs < 1.5: H1N1 (1.25), H3N2 (0.94), B-Yamagata (0.62), and B-Victoria (0.59). Furthermore, FluGuard was non-inferior to Vaxigrip with the upper bounds of the 95 % CI on the difference between the seroconversion rates < 10 %: H1N1 (2 %), H3N2 (10 %), B-Yamagata (-10 %), and B-Victoria (-29 %). The prevalence of solicited adverse drug reactions did not differ between groups. Furthermore, participants did not experience serious adverse events. CONCLUSION Our findings support the non-inferiority of the FluGuard vaccine to the Vaxigrip vaccine regarding immunogenicity and safety. CLINICAL TRIAL REGISTRY The study protocol was approved by the Iranian Registry of Clinical Trials (IRCT20210901052358N5).
Collapse
Affiliation(s)
- Davood Yadegarynia
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Keyvanfar
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Tehrani
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahnaz Sali
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Abolghasemi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Mercolini F, Abram N, Cesaro S. Managing acute COVID-19 in immunocompromised pediatric patients. Expert Rev Clin Immunol 2024; 20:349-357. [PMID: 38099388 DOI: 10.1080/1744666x.2023.2295982] [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: 05/14/2023] [Accepted: 12/13/2023] [Indexed: 03/16/2024]
Abstract
INTRODUCTION SARS-CoV-2 infection is a potentially life-threatening infection in immunocompromised pediatric patients, and its management has rapidly evolved during the pandemic. To control SARS-CoV-2 infection over time, the scenario changed for the better with the introduction of specific treatments such as antiviral drugs, vaccines, and monoclonal antibodies, together with drugs blocking the inflammatory cytokine cascade and improvements in supportive care. AREAS COVERED This paper discusses the therapeutic strategies to apply for patients affected by COVID-19 in the pediatric population, with a focus on the immunocompromised patients. EXPERT OPINION Treatment in pediatric patients retraces the therapies investigated and approved in adults and must be calibrated on the basis of the severity of the infection (anti-spike monoclonal antibody, antivirals, anti-inflammatory drugs, and immunomodulators). Transmission prevention policies and vaccination reduce the risk of infection, while early intervention in the immunocompromised patients at high-risk of progression to severe-critical COVID-19 may reduce the period of viral shedding and the need for hospitalization, intensive care admission, and death. In hemato-oncological patients, the delayed treatment of SARS-CoV-2 infection or COVID-19 disease represents a frequent complication and its impact on the patient outcome remains a matter of research for the next few years.
Collapse
Affiliation(s)
- Federico Mercolini
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS, Azienda Ospedaliero-Universitaria di Bologna institution, Bologna, Italy
| | - Nicoletta Abram
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS, Azienda Ospedaliero-Universitaria di Bologna institution, Bologna, Italy
| | - Simone Cesaro
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
8
|
Meeraus W, Joy M, Ouwens M, Taylor KS, Venkatesan S, Dennis J, Tran TN, Dashtban A, Fan X, Williams R, Morris T, Carty L, Kar D, Hoang U, Feher M, Forbes A, Jamie G, Hinton W, Sanecka K, Byford R, Anand SN, Hobbs FDR, Clifton DA, Pollard AJ, Taylor S, de Lusignan S. AZD1222 effectiveness against severe COVID-19 in individuals with comorbidity or frailty: The RAVEN cohort study. J Infect 2024; 88:106129. [PMID: 38431156 DOI: 10.1016/j.jinf.2024.106129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES Despite being prioritized during initial COVID-19 vaccine rollout, vulnerable individuals at high risk of severe COVID-19 (hospitalization, intensive care unit admission, or death) remain underrepresented in vaccine effectiveness (VE) studies. The RAVEN cohort study (NCT05047822) assessed AZD1222 (ChAdOx1 nCov-19) two-dose primary series VE in vulnerable populations. METHODS Using the Oxford-Royal College of General Practitioners Clinical Informatics Digital Hub, linked to secondary care, death registration, and COVID-19 datasets in England, COVID-19 outcomes in 2021 were compared in vaccinated and unvaccinated individuals matched on age, sex, region, and multimorbidity. RESULTS Over 4.5 million AZD1222 recipients were matched (mean follow-up ∼5 months); 68% were ≥50 years, 57% had high multimorbidity. Overall, high VE against severe COVID-19 was demonstrated, with lower VE observed in vulnerable populations. VE against hospitalization was higher in the lowest multimorbidity quartile (91.1%; 95% CI: 90.1, 92.0) than the highest quartile (80.4%; 79.7, 81.1), and among individuals ≥65 years, higher in the 'fit' (86.2%; 84.5, 87.6) than the frailest (71.8%; 69.3, 74.2). VE against hospitalization was lowest in immunosuppressed individuals (64.6%; 60.7, 68.1). CONCLUSIONS Based on integrated and comprehensive UK health data, overall population-level VE with AZD1222 was high. VEs were notably lower in vulnerable groups, particularly the immunosuppressed.
Collapse
Affiliation(s)
- Wilhelmine Meeraus
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Mark Joy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Mario Ouwens
- Medical & Payer Evidence Statistics, BioPharmaceuticals Medical, AstraZeneca, Mölndal, Sweden
| | - Kathryn S Taylor
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Sudhir Venkatesan
- Medical & Payer Evidence Statistics, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | | | - Trung N Tran
- Biopharmaceutical Medicine Respiratory and Immunology, AstraZeneca, Gaithersburg, MD, USA
| | - Ashkan Dashtban
- Institute of Health Informatics, University College London, London, UK
| | - Xuejuan Fan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Robert Williams
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Tamsin Morris
- Medical and Scientific Affairs, BioPharmaceuticals Medical, AstraZeneca, London, UK
| | - Lucy Carty
- Medical & Payer Evidence Statistics, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Debasish Kar
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Uy Hoang
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Feher
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Anna Forbes
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Gavin Jamie
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - William Hinton
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Kornelia Sanecka
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Warsaw, Poland
| | - Rachel Byford
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Sneha N Anand
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - F D Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - David A Clifton
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sylvia Taylor
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; Royal College of General Practitioners Research and Surveillance Centre, London, UK.
| |
Collapse
|
9
|
Cherry N, Adisesh A, Burstyn I, Charlton C, Chen Y, Durand-Moreau Q, Labrèche F, Ruzycki S, Turnbull L, Zadunayski T, Yasui Y. Determinants of SARS-CoV-2 IgG response and decay in Canadian healthcare workers: A prospective cohort study. Vaccine 2024; 42:1168-1178. [PMID: 38278628 DOI: 10.1016/j.vaccine.2024.01.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Healthcare workers (HCWs) from an interprovincial Canadian cohort gave serial blood samples to identify factors associated with anti-receptor binding domain (anti-RBD) IgG response to the SARS-CoV-2 virus. METHODS Members of the HCW cohort donated blood samples four months after their first SARS-CoV-2 immunization and again at 7, 10 and 13 months. Date and type of immunizations and dates of SARS-CoV-2 infection were collected at each of four contacts, together with information on immunologically-compromising conditions and current therapies. Blood samples were analyzed centrally for anti-RBD IgG and anti-nucleocapsid IgG (Abbott Architect, Abbott Diagnostics). Records of immunization and SARS-CoV-2 testing from public health agencies were used to assess the impact of reporting errors on estimates from the random-effects multivariable model fitted to the data. RESULTS 2752 of 4567 vaccinated cohort participants agreed to donate at least one blood sample. Modelling of anti-RBD IgG titer from 8903 samples showed an increase in IgG with each vaccine dose and with first infection. A decrease in IgG titer was found with the number of months since vaccination or infection, with the sharpest decline after the third dose. An immunization regime that included mRNA1273 (Moderna) resulted in higher anti-RBD IgG. Participants reporting multiple sclerosis, rheumatoid arthritis or taking selective immunosuppressants, tumor necrosis factor inhibitors, calcineurin inhibitors and antineoplastic agents had lower anti-RBD IgG. Supplementary analyses showed higher anti-RBD IgG in those reporting side-effects of vaccination, no relation of anti-RBD IgG to obesity and lower titers in women immunized in early or mid-pregnancy. Sensitivity analysis results suggested no important bias in the self-report data. CONCLUSION Creation of a prospective cohort was central to the credibility of results presented here. Serial serology assessments, with longitudinal analysis, provided effect estimates with enhanced accuracy and a clearer understanding of medical and other factors affecting response to vaccination.
Collapse
Affiliation(s)
- Nicola Cherry
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada.
| | - Anil Adisesh
- Division2 Division of Occupational Medicine, Department of Medicine, University of Toronto, C. David Naylor Building, 6 Queen's Park Crescent West, Toronto, ON M5S 3H2, Canada
| | - Igor Burstyn
- Department of Environmental and Occupational Health, Drexel University, Philadelphia, PA 19104, USA
| | - Carmen Charlton
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Yan Chen
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| | - Quentin Durand-Moreau
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - France Labrèche
- Research Department, Institut de recherche Robert-Sauvé en santé et en sécurité du travail, 505 de Maisonneuve Blvd, West Montreal, QC H3A 3C2, Canada
| | - Shannon Ruzycki
- Department of Medicine, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada
| | - LeeAnn Turnbull
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Tanis Zadunayski
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - Yutaka Yasui
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| |
Collapse
|
10
|
Sulaiman SK, Musa MS, Tsiga-Ahmed FI, Sulaiman AK, Bako AT. A systematic review and meta-analysis of the global prevalence and determinants of COVID-19 vaccine acceptance and uptake in people living with HIV. Nat Hum Behav 2024; 8:100-114. [PMID: 37904021 PMCID: PMC10810755 DOI: 10.1038/s41562-023-01733-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/26/2023] [Indexed: 11/01/2023]
Abstract
People living with HIV (PLHIV) are at higher risk of poor outcomes of SARS-CoV-2 infection. Here we report the pooled prevalence of COVID-19 vaccine acceptance/uptake and determinants among this vulnerable population of PLHIV based on a systematic review and meta-analysis of studies published by 25 August 2023. Among the 54 included studies (N = 167,485 participants), 53 (N = 166,455) provided data on vaccine acceptance rate, while 27 (N = 150,926) provided uptake data. The global prevalences of COVID-19 vaccine acceptance and uptake were 67.0% and 56.6%, respectively. Acceptance and uptake rates were 86.6% and 90.1% for the European Region, 74.9% and 71.6% for the Region of the Americas, 62.3% and 78.9% for the South-East Asian Region, 64.6% and 19.3% for the Eastern Mediterranean Region, 58.0% and 35.5% for the African Region, and 57.4% and 44.0% for the Western Pacific Region. The acceptance rate increased from 65.9% in 2020 to 71.0% in 2022, and the uptake rate increased from 55.9% in 2021 to 58.1% in 2022. Men, PLHIV aged ≥40 years and those who had recently received the influenza vaccine were more likely to accept and receive the COVID-19 vaccine. Factors associated with lower uptake included Black race, other races (Latinx/Hispanic/mixed race), low education level and being unemployed. Vaccine-related factors associated with higher acceptance included belief in vaccine effectiveness, vaccine trust, perceived high susceptibility to SARS-CoV-2 infection and fear of potential COVID-19 effect in PLHIV. Sustained efforts and targeted interventions are needed to reduce regional disparities in COVID-19 vaccine uptake among PLHIV.
Collapse
Affiliation(s)
| | - Muhammad Sale Musa
- Department of Medicine, Yobe State University Teaching Hospital, Damaturu, Nigeria
| | | | - Abdulwahab Kabir Sulaiman
- Department of Medicine, Murtala Muhammad Specialist Hospital, Kano, Nigeria
- Kwanar Dawaki COVID-19 Isolation Center, Kano, Nigeria
| | | |
Collapse
|
11
|
Morgans HA, Bradley T, Flebbe-Rehwaldt L, Selvarangan R, Bagherian A, Barnes AP, Bass J, Cooper AM, Fischer R, Kleiboeker S, Lee BR, LeMaster C, Markus K, Morrison S, Myers A, Myers D, Payne E, Schuster JE, Standley S, Wieser A, Warady B. Humoral and cellular response to the COVID-19 vaccine in immunocompromised children. Pediatr Res 2023; 94:200-205. [PMID: 36376507 PMCID: PMC9662120 DOI: 10.1038/s41390-022-02374-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND A suboptimal response to the 2-dose COVID-19 vaccine series in the immunocompromised population prompted recommendations for a 3rd primary dose. We aimed to determine the humoral and cellular immune response to the 3rd COVID-19 vaccine in immunocompromised children. METHODS Prospective cohort study of immunocompromised participants, 5-21 years old, who received 2 prior doses of an mRNA COVID-19 vaccine. Humoral and CD4/CD8 T-cell responses were measured to SARS-CoV-2 spike antigens prior to receiving the 3rd vaccine dose and 3-4 weeks after the 3rd dose was given. RESULTS Of the 37 participants, approximately half were solid organ transplant recipients. The majority (86.5%) had a detectable humoral response after the 2nd and 3rd vaccine doses, with a significant increase in antibody levels after the 3rd dose. Positive T-cell responses increased from being present in 86.5% to 100% of the cohort after the 3rd dose. CONCLUSIONS Most immunocompromised children mount a humoral and cellular immune response to the 2-dose COVID-19 vaccine series, which is significantly augmented after receiving the 3rd vaccine dose. This supports the utility of the 3rd vaccine dose and the rationale for ongoing emphasis for vaccination against COVID-19 in this population. IMPACT Most immunocompromised children mount a humoral and cellular immune response to the 2-dose COVID-19 vaccine series, which is significantly augmented after receiving the 3rd vaccine dose. This is the first prospective cohort study to analyze both the humoral and T-cell immune response to the 3rd COVID-19 primary vaccine dose in children who are immunocompromised. The results of this study support the utility of the 3rd vaccine dose and the rationale for ongoing emphasis for vaccination against COVID-19 in the immunosuppressed pediatric population.
Collapse
Affiliation(s)
- Heather A Morgans
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA.
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA.
| | - Todd Bradley
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | | | | | | | - Aliessa P Barnes
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Julie Bass
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Ashley M Cooper
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Ryan Fischer
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | | | - Brian R Lee
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Cas LeMaster
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Kelsey Markus
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | | | - Angela Myers
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Doug Myers
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Erin Payne
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Jennifer E Schuster
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Sarah Standley
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Andrea Wieser
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Bradley Warady
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| |
Collapse
|
12
|
Meredith RT, Bermingham MD, Bentley K, Agah S, Aboagye-Odei A, Yarham RAR, Mills H, Shaikh M, Hoye N, Stanton RJ, Chadwick DR, Oliver MA. Differential cellular and humoral immune responses in immunocompromised individuals following multiple SARS-CoV-2 vaccinations. Front Cell Infect Microbiol 2023; 13:1207313. [PMID: 37424787 PMCID: PMC10327606 DOI: 10.3389/fcimb.2023.1207313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction The heterogeneity of the immunocompromised population means some individuals may exhibit variable, weak or reduced vaccine-induced immune responses, leaving them poorly protected from COVID-19 disease despite receiving multiple SARS-CoV-2 vaccinations. There is conflicting data on the immunogenicity elicited by multiple vaccinations in immunocompromised groups. The aim of this study was to measure both humoral and cellular vaccine-induced immunity in several immunocompromised cohorts and to compare them to immunocompetent controls. Methods Cytokine release in peptide-stimulated whole blood, and neutralising antibody and baseline SARS-CoV-2 spike-specific IgG levels in plasma were measured in rheumatology patients (n=29), renal transplant recipients (n=46), people living with HIV (PLWH) (n=27) and immunocompetent participants (n=64) post third or fourth vaccination from just one blood sample. Cytokines were measured by ELISA and multiplex array. Neutralising antibody levels in plasma were determined by a 50% neutralising antibody titre assay and SARS-CoV-2 spike specific IgG levels were quantified by ELISA. Results In infection negative donors, IFN-γ, IL-2 and neutralising antibody levels were significantly reduced in rheumatology patients (p=0.0014, p=0.0415, p=0.0319, respectively) and renal transplant recipients (p<0.0001, p=0.0005, p<0.0001, respectively) compared to immunocompetent controls, with IgG antibody responses similarly affected. Conversely, cellular and humoral immune responses were not impaired in PLWH, or between individuals from all groups with previous SARS-CoV-2 infections. Discussion These results suggest that specific subgroups within immunocompromised cohorts could benefit from distinct, personalised immunisation or treatment strategies. Identification of vaccine non-responders could be critical to protect those most at risk.
Collapse
Affiliation(s)
| | | | - Kirsten Bentley
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sayeh Agah
- InBio, Charlottesville, VA, United States
| | - Abigail Aboagye-Odei
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | | | | - Muddassir Shaikh
- Department of Kidney Services, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Neil Hoye
- Department of Rheumatology, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David R. Chadwick
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | |
Collapse
|
13
|
Xu W, Ren W, Wu T, Wang Q, Luo M, Yi Y, Li J. Real-World Safety of COVID-19 mRNA Vaccines: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2023; 11:1118. [PMID: 37376508 DOI: 10.3390/vaccines11061118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
With the mass vaccination program for COVID-19 mRNA vaccines, there has been sufficient real-world study (RWS) on the topic to summarize their safety in the total population and in immunocompromised (IC) patients who were excluded from phase 3 clinical trials. We conducted a systematic review and meta-analysis to evaluate the safety of COVID-19 mRNA vaccines, with a total of 5,132,799 subjects from 122 articles. In the case of the total population vaccinated with first, second, and third doses, the pooled incidence of any adverse events (AEs) was 62.20%, 70.39%, and 58.60%; that of any local AEs was 52.03%, 47.99%, and 65.00%; that of any systemic AEs was 29.07%, 47.86%, and 32.71%. Among the immunocompromised patients, the pooled odds ratio of any AEs, any local AEs, and systemic AEs were slightly lower than or similar to those of the healthy controls at 0.60 (95% CI: 0.33-1.11), 0.19 (95% CI: 0.10-0.37), and 0.36 (95% CI: 0.25-0.54), with pooled incidences of 51.95%, 38.82%, and 31.00%, respectively. The spectrum of AEs associated with the vaccines was broad, but most AEs were transient, self-limiting, and mild to moderate. Moreover, younger adults, women, and people with prior SARS-CoV-2 infection were more likely to experience AEs.
Collapse
Affiliation(s)
- Wanqian Xu
- School of Public Health, The Second Hospital of Nanjing, Nanjing Medical University, Nanjing 211166, China
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
| | - Weigang Ren
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Tongxin Wu
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Qin Wang
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Mi Luo
- School of Public Health, The Second Hospital of Nanjing, Nanjing Medical University, Nanjing 211166, China
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
| | - Yongxiang Yi
- School of Public Health, The Second Hospital of Nanjing, Nanjing Medical University, Nanjing 211166, China
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Junwei Li
- The Clinical Infectious Disease Center of Nanjing, Nanjing 210003, China
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| |
Collapse
|
14
|
Tan TT, Ng HJ, Young B, Khan BA, Shetty V, Azmi N, Clissold S. Effectiveness of vaccination against SARS-CoV-2 and the need for alternative preventative approaches in immunocompromised individuals: a narrative review of systematic reviews. Expert Rev Vaccines 2023; 22:341-365. [PMID: 36920116 DOI: 10.1080/14760584.2023.2191716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including administration of booster doses, continues to be the most effective method for controlling COVID-19-related complications including progression to severe illness and death.However, there is mounting evidence that more needs to be done to protect individuals with compromised immune function. AREAS COVERED Here, we review the effectiveness of COVID-19 vaccination in immunocompromised patients, including those with primary immunodeficiencies, HIV, cancer (including hematological malignancies), solid organ transplant recipients and chronic kidney disease, as reported in systematic reviews/meta-analyses published over a 12-month period in PubMed. Given the varied responses to vaccination patients with compromised immune function, a major goal of this analysis was to try to identify specific risk-factors related to vaccine failure. EXPERT OPINION COVID-19 remains a global problem, with new variants of concern emerging at regular intervals. There is an ongoing need for optimal vaccine strategies to combat the pandemic. In addition, alternative treatment approaches are needed for immunocompromised patients who may not mount an adequate immune response to current COVID-19 vaccines. Identification of high-risk patients, and the introduction of newer antiviral approaches such as monoclonal antibodies, will offer physicians therapeutic options for such vulnerable individuals.
Collapse
Affiliation(s)
- Thuan Tong Tan
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Heng Joo Ng
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Barnaby Young
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Behram Ali Khan
- Medical Services Department, The National Kidney Foundation, Singapore and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | |
Collapse
|
15
|
Gattinger P, Ohradanova-Repic A, Valenta R. Importance, Applications and Features of Assays Measuring SARS-CoV-2 Neutralizing Antibodies. Int J Mol Sci 2023; 24:ijms24065352. [PMID: 36982424 PMCID: PMC10048970 DOI: 10.3390/ijms24065352] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023] Open
Abstract
More than three years ago, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused the unforeseen COVID-19 pandemic with millions of deaths. In the meantime, SARS-CoV-2 has become endemic and is now part of the repertoire of viruses causing seasonal severe respiratory infections. Due to several factors, among them the development of SARS-CoV-2 immunity through natural infection, vaccination and the current dominance of seemingly less pathogenic strains belonging to the omicron lineage, the COVID-19 situation has stabilized. However, several challenges remain and the possible new occurrence of highly pathogenic variants remains a threat. Here we review the development, features and importance of assays measuring SARS-CoV-2 neutralizing antibodies (NAbs). In particular we focus on in vitro infection assays and molecular interaction assays studying the binding of the receptor binding domain (RBD) with its cognate cellular receptor ACE2. These assays, but not the measurement of SARS-CoV-2-specific antibodies per se, can inform us of whether antibodies produced by convalescent or vaccinated subjects may protect against the infection and thus have the potential to predict the risk of becoming newly infected. This information is extremely important given the fact that a considerable number of subjects, in particular vulnerable persons, respond poorly to the vaccination with the production of neutralizing antibodies. Furthermore, these assays allow to determine and evaluate the virus-neutralizing capacity of antibodies induced by vaccines and administration of plasma-, immunoglobulin preparations, monoclonal antibodies, ACE2 variants or synthetic compounds to be used for therapy of COVID-19 and assist in the preclinical evaluation of vaccines. Both types of assays can be relatively quickly adapted to newly emerging virus variants to inform us about the magnitude of cross-neutralization, which may even allow us to estimate the risk of becoming infected by newly appearing virus variants. Given the paramount importance of the infection and interaction assays we discuss their specific features, possible advantages and disadvantages, technical aspects and not yet fully resolved issues, such as cut-off levels predicting the degree of in vivo protection.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University, 3500 Krems an der Donau, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
16
|
Valentini D, Carfi A, Di Paola A, Yarci-Carrión A, Villani A, Real de Asúa D. Factor associated with SARS-CoV-2 vaccination serological efficacy in adolescents and adults with Down syndrome: Data from an international, collaborative initiative of the Trisomy 21 Research Society. J Infect 2023; 86:e91-e93. [PMID: 36813120 PMCID: PMC9940466 DOI: 10.1016/j.jinf.2023.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Affiliation(s)
| | - A Carfi
- Department of Geriatrics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - A Di Paola
- Department of Psychology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - A Yarci-Carrión
- Department of Microbiology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Alberto Villani
- Pediatric Unit, Pediatric Emergency Department (DEA), Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - D Real de Asúa
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Department of Internal Medicine, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain.
| |
Collapse
|
17
|
Fowokan A, Samji H, Puyat JH, Janjua NZ, Wilton J, Wong J, Grennan T, Chambers C, Kroch A, Costiniuk CT, Cooper CL, Burchell AN, Anis A. Effectiveness of COVID-19 vaccines in people living with HIV in British Columbia and comparisons with a matched HIV-negative cohort: a test-negative design. Int J Infect Dis 2023; 127:162-170. [PMID: 36462571 PMCID: PMC9711901 DOI: 10.1016/j.ijid.2022.11.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/24/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVES We estimated the effectiveness of COVID-19 vaccines against laboratory-confirmed SARS-CoV-2 infection among people living with HIV (PLWH) and compared the estimates with a matched HIV-negative cohort. METHODS We used the British Columbia COVID-19 Cohort, a population-based data platform, which integrates COVID-19 data on SARS-CoV-2 tests, laboratory-confirmed cases, and immunizations with provincial health services data. The vaccine effectiveness (VE) was estimated with a test-negative design using the multivariable logistic regression. RESULTS The adjusted VE against SARS-CoV-2 infection was 71.1% (39.7, 86.1%) 7-59 days after two doses, rising to 89.3% (72.2, 95.9%) between 60 and 89 days. VE was preserved 4-6 months after the receipt of two doses, after which noticeable waning was observed (51.3% [4.8, 75.0%]). In the matched HIV-negative cohort (n = 375,043), VE peaked at 91.4% (90.9, 91.8%) 7-59 days after two doses and was sustained for up to 4 months, after which evidence of waning was observed, dropping to 84.2% (83.4, 85.0%) between 4 and 6 months. CONCLUSION The receipt of two COVID-19 vaccine doses was effective against SARS-CoV-2 infection among PLWH pre-Omicron. VE estimates appeared to peak later in PLWH than in the matched HIV-negative cohort and the degree of waning was relatively quicker in PLWH; however, peak estimates were comparable in both populations.
Collapse
Affiliation(s)
- Adeleke Fowokan
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Hasina Samji
- British Columbia Centre for Disease Control, Vancouver, Canada,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada,Corresponding author at: Hasina Samji, Senior Scientist British Columbia Centre for Disease Control, Provincial Health Services Authority, Assistant Professor
- Faculty of Health Sciences, Simon Fraser University, 655 West 12th Avenue, Vancouver British Columbia, V5Z 4R4
| | - Joseph H. Puyat
- British Columbia Centre for Disease Control, Vancouver, Canada,School of Population and Public Health, University of British Columbia, Vancouver, Canada,Centre for Health Evaluation and Outcome Sciences, St Paul's Hospital, Vancouver, Canada
| | - Naveed Z. Janjua
- British Columbia Centre for Disease Control, Vancouver, Canada,School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - James Wilton
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Jason Wong
- British Columbia Centre for Disease Control, Vancouver, Canada,School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Troy Grennan
- British Columbia Centre for Disease Control, Vancouver, Canada,School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Catharine Chambers
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | | - Cecilia T. Costiniuk
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Ann N. Burchell
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada,Department of Family and Community Medicine, Faculty of Medicine, University of Toronto, Toronto, Canada,MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health, Toronto, Canada
| | - Aslam Anis
- School of Population and Public Health, University of British Columbia, Vancouver, Canada,Centre for Health Evaluation and Outcome Sciences, St Paul's Hospital, Vancouver, Canada
| | | |
Collapse
|
18
|
Greene SE, Huang Y, Kim W, Liebeskind MJ, Chandrasekaran V, Liu Z, Deepak P, Paley MA, Lew D, Yang M, Matloubian M, Gensler LS, Nakamura MC, O'Hallaran JA, Presti RM, Whelan SPJ, Buchser WJ, Kim AHJ, Weil GJ. A simple point-of-care assay accurately detects anti-spike antibodies after SARS-CoV-2 vaccination. JOURNAL OF CLINICAL VIROLOGY PLUS 2023; 3:100135. [PMID: 36644774 PMCID: PMC9831968 DOI: 10.1016/j.jcvp.2023.100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Objective Lateral flow assays (LFA) are sensitive for detecting antibodies to SARS-CoV-2 proteins within weeks after infection. This study tested samples from immunocompetent adults, and those receiving treatments for chronic inflammatory diseases (CID), before and after mRNA SARS-CoV-2 vaccination. Methods We compared results obtained with the COVIBLOCK Covid-19 LFA to those obtained by anti-spike (S) ELISA. Results The LFA detected anti-S antibodies in 29 of 29 (100%) of the immunocompetent and 110 of 126 (87.3%) of the CID participants after vaccination. Semiquantitative LFA scores were statistically significantly lower in samples from immunosuppressed participants, and were significantly correlated with anti-S antibody levels measured by ELISA. Conclusions This simple LFA test is a practical alternative to laboratory-based assays for detecting anti-S antibodies after infection or vaccination. This type of test may be most useful for testing people in outpatient or resource-limited settings.
Collapse
Affiliation(s)
- Sarah E Greene
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, 4444 Forest Park Avenue, Rm 4184 , St. Louis, MO 63110, United States
| | - Yuefang Huang
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Wooseob Kim
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mariel J Liebeskind
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Vinay Chandrasekaran
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Parakkal Deepak
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael A Paley
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Daphne Lew
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Monica Yang
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Mehrdad Matloubian
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Lianne S Gensler
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States.,San Francisco VA Health Care System, San Francisco, CA, United States
| | - Mary C Nakamura
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States.,San Francisco VA Health Care System, San Francisco, CA, United States
| | - Jane A O'Hallaran
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Rachel M Presti
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - William J Buchser
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Alfred H J Kim
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States.,Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Gary J Weil
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| |
Collapse
|
19
|
Ao G, Li T, Wang Y, Tran C, Gao M, Chen M. The effect of SARS-CoV-2 double vaccination on the outcomes of hemodialysis patients with COVID-19: A meta-analysis. J Infect 2023; 86:e43-e45. [PMID: 36174838 PMCID: PMC9511879 DOI: 10.1016/j.jinf.2022.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 09/17/2022] [Indexed: 02/02/2023]
Affiliation(s)
- Guangyu Ao
- Department of Nephrology, Chengdu First People's Hospital, No.18 Wanxiang North Road, High-tech District, Chengdu, Sichuan 610095, China
| | - Toni Li
- School of Medicine, Queen's University, Kingston, Canada
| | - Yushu Wang
- Chengdu West China Clinical Research Center, Chengdu, Sichuan, China
| | - Carolyn Tran
- Schulich School of Medicine & Dentistry, Western University, London, Canada
| | - Ming Gao
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China.
| | - Min Chen
- Department of Nephrology, Chengdu First People's Hospital, No.18 Wanxiang North Road, High-tech District, Chengdu, Sichuan 610095, China.
| |
Collapse
|
20
|
Salto-Alejandre S, Carretero-Ledesma M, Camacho-Martínez P, Berastegui-Cabrera J, Infante C, Rodríguez-Álvarez R, Alba J, Pérez-Palacios P, García-Díaz E, Roca C, Praena J, Blanco-Vidal MJ, Santibáñez S, Valverde-Ortiz R, Nieto-Arana J, García-García C, Blanco-Vida MJ, Gutiérrez-Campos D, Maldonado N, Bernal G, Gómez-Bravo MÁ, Sobrino JM, Aguilar-Guisado M, Álvarez-Marín R, Goikoetxea-Aguirre J, Oteo JA, Palacios-Baena ZR, Pascual Á, Lepe JA, Rodríguez-Baño J, Cisneros JM, Pachón J, Sánchez-Céspedes J, Cordero E. Serum IFN-γ and RNAemia temporal profiles as biomarkers of severe COVID-19 in solid organ transplant and immunocompetent patients. J Infect 2023; 86:529-533. [PMID: 36690212 PMCID: PMC9859635 DOI: 10.1016/j.jinf.2023.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Affiliation(s)
- Sonsoles Salto-Alejandre
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | - Marta Carretero-Ledesma
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | - Pedro Camacho-Martínez
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | - Judith Berastegui-Cabrera
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | - Carmen Infante
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | | | - Jorge Alba
- Department of Infectious Diseases, San Pedro-CIBIR University Hospital, La Rioja, Logroño, Spain
| | - Patricia Pérez-Palacios
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain
| | - Emilio García-Díaz
- Medical Surgical Respiratory Unit, Virgen del Rocío University Hospital, Seville, Spain
| | - Cristina Roca
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | - Julia Praena
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain
| | | | - Sonia Santibáñez
- Department of Infectious Diseases, San Pedro-CIBIR University Hospital, La Rioja, Logroño, Spain
| | | | - Javier Nieto-Arana
- Department of Infectious Diseases, Hospital Universitario Cruces, Bizkaia, Spain
| | - Concepción García-García
- Department of Infectious Diseases, San Pedro-CIBIR University Hospital, La Rioja, Logroño, Spain
| | | | - David Gutiérrez-Campos
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain
| | - Natalia Maldonado
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gabriel Bernal
- Unit of Urology and Nephrology, Virgen del Rocío University Hospital, Seville, Spain
| | | | - José Manuel Sobrino
- Unit of Cardiology and Cardiovascular Surgery, Virgen del Rocío University Hospital, Seville, Spain
| | - Manuela Aguilar-Guisado
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Rocío Álvarez-Marín
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - José Antonio Oteo
- Department of Infectious Diseases, San Pedro-CIBIR University Hospital, La Rioja, Logroño, Spain
| | - Zaira R. Palacios-Baena
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Álvaro Pascual
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Department of Microbiology, University of Seville, Seville, Spain
| | - José Antonio Lepe
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Department of Microbiology, University of Seville, Seville, Spain
| | - Jesús Rodríguez-Baño
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - José Miguel Cisneros
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - Jerónimo Pachón
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - Javier Sánchez-Céspedes
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Corresponding author at: Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Av. Manuel Siurot s/n, Seville 41013, Spain
| | - Elisa Cordero
- Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain,Institute of Biomedicine of Seville (IBiS), Virgen del Rocío and Virgen Macarena University Hospitals/CSIC/University of Seville, Seville, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain,Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | | |
Collapse
|
21
|
Lupu A, Miron IC, Gavrilovici C, Raileanu AA, Starcea IM, Ioniuc I, Azoicai A, Mocanu A, Butnariu LI, Dragan F, Lupu VV. Pediatric Systemic Lupus Erythematous in COVID-19 Era. Viruses 2023; 15:272. [PMID: 36851487 PMCID: PMC9966057 DOI: 10.3390/v15020272] [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: 12/30/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Pediatric systemic lupus erythematosus is a chronic autoimmune disorder with a highly variable course and prognosis. It results in functional abnormalities in the immune system due to intrinsic factors and the use of immunosuppressive therapies associated with underlying comorbidities seem to increase the risk of severe COVID-19 and poor outcomes of the disease in pediatric systemic lupus erythematosus (SLE) patients. The aim of this review is to obtain a better understanding of the existing link between this new viral infection and pediatric lupus. We have analyzed the characteristics of newly diagnosed cases of pediatric SLE following COVID-19 which have been reported in the literature and which describe the impact that COVID-19 has on patients already suffering with pediatric SLE.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Cristina Gavrilovici
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Anca Adam Raileanu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Ileana Ioniuc
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alice Azoicai
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Adriana Mocanu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Lacramioara Ionela Butnariu
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
22
|
Pessoa IL, Paolilo RB, da Paz JA. COVID-19 in children and adolescents with neuroimmunological disorders. Clinics (Sao Paulo) 2023; 78:100142. [PMID: 36512952 PMCID: PMC9672691 DOI: 10.1016/j.clinsp.2022.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/06/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Ingrid Lacerda Pessoa
- Department of Neurology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Renata Barbosa Paolilo
- Department of Neurology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil.
| | - José Albino da Paz
- Department of Neurology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| |
Collapse
|
23
|
Halma MTJ, Wever MJA, Abeln S, Roche D, Wuite GJL. Therapeutic potential of compounds targeting SARS-CoV-2 helicase. Front Chem 2022; 10:1062352. [PMID: 36561139 PMCID: PMC9763700 DOI: 10.3389/fchem.2022.1062352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The economical and societal impact of COVID-19 has made the development of vaccines and drugs to combat SARS-CoV-2 infection a priority. While the SARS-CoV-2 spike protein has been widely explored as a drug target, the SARS-CoV-2 helicase (nsp13) does not have any approved medication. The helicase shares 99.8% similarity with its SARS-CoV-1 homolog and was shown to be essential for viral replication. This review summarizes and builds on existing research on inhibitors of SARS-CoV-1 and SARS-CoV-2 helicases. Our analysis on the toxicity and specificity of these compounds, set the road going forward for the repurposing of existing drugs and the development of new SARS-CoV-2 helicase inhibitors.
Collapse
Affiliation(s)
- Matthew T. J. Halma
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- LUMICKS B. V., Amsterdam, Netherlands
| | - Mark J. A. Wever
- DCM, University of Grenoble Alpes, Grenoble, France
- Edelris, Lyon, France
| | - Sanne Abeln
- Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Gijs J. L. Wuite
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
24
|
Nakashima K, Ishida M, Matsui H, Yoshida C, Nagai T, Shiraga M, Nakaoka H, Otsuka Y, Nakagama Y, Kaku N, Nitahara Y, Kido Y, Hirota Y. Immunogenicity and safety of COVID-19 vaccine in lung cancer patients receiving anticancer treatment: A prospective multicenter cohort study. Hum Vaccin Immunother 2022; 18:2140549. [PMID: 36369871 DOI: 10.1080/21645515.2022.2140549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study assessed the immunogenicity and safety of the BNT162b2 mRNA vaccine in lung cancer patients receiving anticancer treatment. We enrolled lung cancer patients receiving anticancer treatment and non-cancer patients; all participants were fully vaccinated with the BNT162b2 vaccine. Blood samples were collected before the first and second vaccinations and 4 ± 1 weeks after the second vaccination. Anti-severe respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein S1 subunit receptor-binding domain antibody titers were measured using the Architect SARS-CoV-2 IgG II Quant and Elecsys Anti-SARS-CoV-2 S assays. Fifty-five lung cancer patients and 38 non-cancer patients were included in the immunogenicity analysis. Lung cancer patients showed significant increase in the geometric mean antibody concentration, which was significantly lower than that in the non-cancer patients after the first (30 vs. 121 AU/mL, p < .001 on Architect; 4.0 vs 1.2 U/mL, p < .001 on Elecsys) and second vaccinations (1632 vs. 3472 AU/mL, p = .005 on Architect; 213 vs 573 A/mL, p = .002 on Elecsys). The adjusted odds ratio (aOR) for seroprotection was significantly lower (p < .05) in lung cancer patients than that in non-cancer patients. Analysis of the anticancer treatment types showed that the aOR for seroprotection was significantly lower (p < .05) in lung cancer patients receiving cytotoxic agents. They showed no increase in adverse reactions. BNT162b2 vaccination in lung cancer patients undergoing anticancer treatment significantly increased (p < .05) antibody titers and showed acceptable safety. Immunogenicity in these patients could be inadequate compared with that in non-cancer patients.
Collapse
Affiliation(s)
- Kei Nakashima
- Department of Pulmonology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | | | - Hiroki Matsui
- Clinical Research Support Office, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Chihiro Yoshida
- Department of Pulmonology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Tatsuya Nagai
- Department of Pulmonology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Minoru Shiraga
- Department of Pulmonology, Chikamori Hospital, Kochi, Japan
| | | | - Yoshihito Otsuka
- Department of Laboratory medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Yu Nakagama
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Natsuko Kaku
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yuko Nitahara
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yasutoshi Kido
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yoshio Hirota
- Clinical Epidemiology Research Center, SOUSEIKAI Medical Group (Medical Co. LTA), Fukuoka, Japan
| |
Collapse
|
25
|
Chen D, Pan Y, Wang Y, Chen X, Zhang J, Zhang Y, Ding T, Yang Z, Lu T, Zheng Y, Wei J, Kong W, Li H, Yin X, Yang G, Zhang G, Wang X. Serological response to COVID-19 vaccine and its predictors in patients with solid malignancies: A systematic review and meta-analysis of 16 cohort studies. J Infect 2022; 86:154-225. [PMID: 36574520 PMCID: PMC9699821 DOI: 10.1016/j.jinf.2022.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Dong Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Yuancan Pan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yue Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xing Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jiahui Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yutong Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Tongjing Ding
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhengzheng Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Taicheng Lu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Zheng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jiangyan Wei
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weijia Kong
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Haiming Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaohui Yin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Guowang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ganlin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Yarci-Carrión A, Esparcia-Pinedo L, Mateo-Jiménez G, Alfranca A, de Asúa DR, Gutiérrez-Cobos A. Effect of a SARS-CoV-2 booster vaccine dose on the immune response of adults with Down syndrome. J Infect 2022; 86:154-225. [PMID: 36417985 PMCID: PMC9675634 DOI: 10.1016/j.jinf.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Ayla Yarci-Carrión
- Microbiology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Laura Esparcia-Pinedo
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Gloria Mateo-Jiménez
- Fundación de Investigación Biomédica del Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain,Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Diego Real de Asúa
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain,Internal Medicine Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain,Corresponding author: Diego Real de Asúa, Department of Internal Medicine, Hospital Universitario de La Princesa, Diego de León 62, 28006 Madrid, Spain, T: +34 915202222
| | - Ainhoa Gutiérrez-Cobos
- Microbiology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| |
Collapse
|
27
|
Sun H, Bu F, Li L, Zhang X, Yan J, Huang T. COVID-19 vaccine response and safety in patients with cancer: An overview of systematic reviews. Front Public Health 2022; 10:1072137. [PMID: 36457318 PMCID: PMC9707626 DOI: 10.3389/fpubh.2022.1072137] [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: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Background To date, the COVID-19 pandemic does not appear to be overcome with new variants continuously emerging. The vaccination against COVID-19 has been the trend, but there are multiple systematic reviews on COVID-19 vaccines in patients with cancer, resulting in redundant and sub-optimal systematic reviews. There are still some doubts about efficacy and safety of the COVID-19 vaccine in cancer patients. Purpose To identify, summarize and synthesize the available evidence of systematic reviews on response and COVID-19 vaccine safety in patients with cancer. Methods Multiple databases were searched from their inception to May 1, 2022 to fetch the relevant articles. Study quality was assessed by AMSTAR2. The protocol of this study was registered on PROSPERO (CRD42022327931). Results A total of 18 articles were finally included. The seroconversion rates after first dose were ranged from 37.30-54.20% in all cancers, 49.60-62.00% in solid cancers and 33.30-56.00% in hematological malignancies. The seroconversion rates after second dose were ranged from 65.30-87.70% in all cancers, 91.60-96.00% in solid cancers and 58.00-72.60% in hematological malignancies. Cancer types and types of therapy could influence vaccine response. COVID-19 vaccines were safe and well-tolerated. Conclusions This study suggests COVID-19 vaccine response is significantly lower in cancer patients. Number of received doses, cancer types and treatment strategies could influence response of COVID-19 vaccine in cancer patients. COVID-19 vaccines are safe and well-tolerated. Considering the emergence of several new variants of SARS-CoV-2 with potential influence on ongoing vaccination programs, there is a need for booster doses to increase the effectiveness of COVID-19 vaccines. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022327931, identifier CRD42022327931.
Collapse
|
28
|
Lazarus JV, Romero D, Kopka CJ, Karim SA, Abu-Raddad LJ, Almeida G, Baptista-Leite R, Barocas JA, Barreto ML, Bar-Yam Y, Bassat Q, Batista C, Bazilian M, Chiou ST, Del Rio C, Dore GJ, Gao GF, Gostin LO, Hellard M, Jimenez JL, Kang G, Lee N, Matičič M, McKee M, Nsanzimana S, Oliu-Barton M, Pradelski B, Pyzik O, Rabin K, Raina S, Rashid SF, Rathe M, Saenz R, Singh S, Trock-Hempler M, Villapol S, Yap P, Binagwaho A, Kamarulzaman A, El-Mohandes A. A multinational Delphi consensus to end the COVID-19 public health threat. Nature 2022; 611:332-345. [PMID: 36329272 PMCID: PMC9646517 DOI: 10.1038/s41586-022-05398-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Despite notable scientific and medical advances, broader political, socioeconomic and behavioural factors continue to undercut the response to the COVID-19 pandemic1,2. Here we convened, as part of this Delphi study, a diverse, multidisciplinary panel of 386 academic, health, non-governmental organization, government and other experts in COVID-19 response from 112 countries and territories to recommend specific actions to end this persistent global threat to public health. The panel developed a set of 41 consensus statements and 57 recommendations to governments, health systems, industry and other key stakeholders across six domains: communication; health systems; vaccination; prevention; treatment and care; and inequities. In the wake of nearly three years of fragmented global and national responses, it is instructive to note that three of the highest-ranked recommendations call for the adoption of whole-of-society and whole-of-government approaches1, while maintaining proven prevention measures using a vaccines-plus approach2 that employs a range of public health and financial support measures to complement vaccination. Other recommendations with at least 99% combined agreement advise governments and other stakeholders to improve communication, rebuild public trust and engage communities3 in the management of pandemic responses. The findings of the study, which have been further endorsed by 184 organizations globally, include points of unanimous agreement, as well as six recommendations with >5% disagreement, that provide health and social policy actions to address inadequacies in the pandemic response and help to bring this public health threat to an end.
Collapse
Affiliation(s)
- Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain.
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York City, NY, USA.
| | - Diana Romero
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York City, NY, USA
| | | | - Salim Abdool Karim
- University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Program of Research in South Africa (CAPRISA), Durban, South Africa
| | - Laith J Abu-Raddad
- Weill Cornell Medicine, Cornell University, Ithaca, NY, USA
- Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation-Education City, Doha, Qatar
| | | | - Ricardo Baptista-Leite
- UNITE Global Parliamentarians Network, Lisbon, Portugal
- Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Institute of Health Sciences (CIIS), Catholic University of Portugal, Lisbon, Portugal
| | | | - Mauricio L Barreto
- Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- University of Bahia, Salvador, Brazil
| | - Yaneer Bar-Yam
- New England Complex Systems Institute, Cambridge, MA, USA
| | - Quique Bassat
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Manhiça Health Research Center (CISM), Maputo, Mozambique
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
- Pediatrics Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Biomedical Research Consortium in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Carolina Batista
- Doctors Without Borders (MSF), Geneva, Switzerland
- Baraka Impact Finance, Geneva, Switzerland
| | | | - Shu-Ti Chiou
- National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Gregory J Dore
- University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia
| | - George F Gao
- Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lawrence O Gostin
- The O'Neill Institute for National and Global Health Law, Georgetown University, Washington, DC, USA
| | | | - Jose L Jimenez
- Department of Chemistry, University of Colorado Boulder, Boulder, CO, USA
- Cooperative Institute for Research in Environmental Sciences (CIRES), University of Colorado Boulder, Boulder, CO, USA
| | | | | | - Mojca Matičič
- Clinic for Infectious Diseases and Febrile Illnesses, University Medical Centre, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Martin McKee
- The London School of Hygiene & Tropical Medicine, London, UK
| | | | | | - Bary Pradelski
- French National Centre for Scientific Research (CNRS), Grenoble, France
| | | | - Kenneth Rabin
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York City, NY, USA
| | - Sunil Raina
- Dr. Rajendra Prasad Government Medical College, Himachal Pradesh, India
| | - Sabina Faiz Rashid
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | | | - Rocio Saenz
- University of Costa Rica, San José, Costa Rica
| | - Sudhvir Singh
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | - Sonia Villapol
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Peiling Yap
- International Digital Health & AI Research Collaborative (I-DAIR), Geneva, Switzerland
| | | | | | - Ayman El-Mohandes
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York City, NY, USA
| |
Collapse
|
29
|
Factors Associated With COVID-19 Vaccine Response in Transplant Recipients: A Systematic Review and Meta-analysis. Transplantation 2022; 106:2068-2075. [PMID: 35761439 PMCID: PMC9521391 DOI: 10.1097/tp.0000000000004256] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The rapid development and universal access to vaccines represent a milestone in combating the coronavirus disease 2019 (COVID-19) pandemic. However, there are major concerns about vaccine response in immunocompromised populations in particular transplant recipients. In the present study, we aim to comprehensively assess the humoral response to COVID-19 vaccination in both orthotopic organ transplant and allogeneic hematopoietic stem cell transplant recipients. METHODS We performed a systematic review and meta-analysis of 96 studies that met inclusion criteria. RESULTS The pooled rates of seroconversion were 49% (95% confidence interval [CI], 43%-55%) in transplant recipients and 99% (95% CI, 99%-99%) in healthy controls after the second dose of vaccine. The pooled rate was 56% (95% CI, 49%-63%) in transplant recipients after the third dose. Immunosuppressive medication is the most prominent risk factor associated with seroconversion failure, but different immunosuppressive regimens are associated with differential outcomes in this respect. Calcineurin inhibitors, steroids, or mycophenolate mofetil/mycophenolic acid are associated with an increased risk of seroconversion failure, whereas azathioprine or mammalian target of rapamycin inhibitors do not. Advanced age, short interval from receiving the vaccine to the time of transplantation, or comorbidities confers a higher risk for seroconversion failure. CONCLUSIONS Transplant recipients compared with the general population have much lower rates of seroconversion upon receiving COVID-19 vaccines. Immunosuppressants are the most prominent factors associated with seroconversion, although different types may have differential effects.
Collapse
|
30
|
Sakuraba A, Luna A, Micic D. A Systematic Review and Meta-Analysis of Serologic Response following Coronavirus Disease 2019 (COVID-19) Vaccination in Solid Organ Transplant Recipients. Viruses 2022; 14:1822. [PMID: 36016444 PMCID: PMC9413038 DOI: 10.3390/v14081822] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/23/2022] Open
Abstract
Solid organ transplant (SOT) recipients are at greater risk of coronavirus disease 2019 (COVID-19) and have attenuated response to vaccinations. In the present meta-analysis, we aimed to evaluate the serologic response to the COVID-19 vaccine in SOT recipients. A search of electronic databases was conducted to identify SOT studies that reported the serologic response to COVID-19 vaccination. We analyzed 44 observational studies including 6158 SOT recipients. Most studies were on mRNA vaccination (mRNA-1273 or BNT162b2). After a single and two doses of vaccine, serologic response rates were 8.6% (95% CI 6.8-11.0) and 34.2% (95% CI 30.1-38.7), respectively. Compared to controls, response rates were lower after a single and two doses of vaccine (OR 0.0049 [95% CI 0.0021-0.012] and 0.0057 [95% CI 0.0030-0.011], respectively). A third dose improved the rate to 65.6% (95% CI 60.4-70.2), but in a subset of patients who had not achieved a response after two doses, it remained low at 35.7% (95% CI 21.2-53.3). In summary, only a small proportion of SOT recipients achieved serologic response to the COVID-19 mRNA vaccine, and that even the third dose had an insufficient response. Alternative strategies for prophylaxis in SOT patients need to be developed. Key Contribution: In this meta-analysis that included 6158 solid organ transplant recipients, the serologic response to the COVID-19 vaccine was extremely low after one (8.6%) and two doses (34.2%). The third dose of the vaccine improved the rate only to 66%, and in the subset of patients who had not achieved a response after two doses, it remained low at 36%. The results of our study suggest that a significant proportion of solid organ transplant recipients are unable to achieve a sufficient serologic response after completing not only the two series of vaccination but also the third booster dose. There is an urgent need to develop strategies for prophylaxis including modified vaccine schedules or the use of monoclonal antibodies in this vulnerable patient population.
Collapse
Affiliation(s)
- Atsushi Sakuraba
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The University of Chicago Medicine, 5841 S. Maryland Ave. MC 4076, Chicago, IL 60637, USA
| | | | | |
Collapse
|
31
|
Xia Y, Yao RQ, Zhao PY, Tao ZB, Zheng LY, Zhou HT, Yao YM, Song XM. Publication trends of research on COVID-19 and host immune response: A bibliometric analysis. Front Public Health 2022; 10:939053. [PMID: 36003630 PMCID: PMC9394856 DOI: 10.3389/fpubh.2022.939053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/18/2022] [Indexed: 01/08/2023] Open
Abstract
IntroductionAs the first bibliometric analysis of COVID-19 and immune responses, this study will provide a comprehensive overview of the latest research advances. We attempt to summarize the scientific productivity and cooperation across countries and institutions using the bibliometric methodology. Meanwhile, using clustering analysis of keywords, we revealed the evolution of research hotspots and predicted future research focuses, thereby providing valuable information for the follow-up studies.MethodsWe selected publications on COVID-19 and immune response using our pre-designed search strategy. Web of Science was applied to screen the eligible publications for subsequent bibliometric analyses. GraphPad Prism 8.0, VOSviewer, and CiteSpace were applied to analyze the research trends and compared the contributions of countries, authors, institutions, and journals to the global publications in this field.ResultsWe identified 2,200 publications on COVID-19 and immune response published between December 1, 2019, and April 25, 2022, with a total of 3,154 citations. The United States (611), China (353), and Germany (209) ranked the top three in terms of the number of publications, accounting for 53.3% of the total articles. Among the top 15 institutions publishing articles in this area, four were from France, four were from the United States, and three were from China. The journal Frontiers in Immunology published the most articles (178) related to COVID-19 and immune response. Alessandro Sette (31 publications) from the United States were the most productive and influential scholar in this field, whose publications with the most citation frequency (3,633). Furthermore, the development and evaluation of vaccines might become a hotspot in relevant scope.ConclusionsThe United States makes the most indispensable contribution in this field in terms of publication numbers, total citations, and H-index. Although publications from China also take the lead regarding quality and quantity, their international cooperation and preclinical research need to be further strengthened. Regarding the citation frequency and the total number of published articles, the latest research progress might be tracked in the top-ranking journals in this field. By analyzing the chronological order of the appearance of retrieved keywords, we speculated that vaccine-related research might be the novel focus in this field.
Collapse
Affiliation(s)
- Yun Xia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Ren-qi Yao
| | - Peng-yue Zhao
- Department of General Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zheng-bo Tao
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Li-yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui-ting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
- Yong-ming Yao
| | - Xue-min Song
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xue-min Song
| |
Collapse
|
32
|
Yan Y, Davgadorj C, Lyu C, Zhang S, Qiu Y. Immunogenicity of a third dose of inactivated COVID-19 vaccine in people living with HIV-1, HBV, and tuberculosis during the Omicron variant epidemic: A cross-sectional study. J Infect 2022; 85:e109-e111. [PMID: 35803384 PMCID: PMC9254457 DOI: 10.1016/j.jinf.2022.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yan Yan
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China; The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China; Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi (Wuxi Infectious Disease Hospital), Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chantsalmaa Davgadorj
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China; The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China
| | - Chunyan Lyu
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shiliang Zhang
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Yuanwang Qiu
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China; Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi (Wuxi Infectious Disease Hospital), Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
33
|
Olivier E, Sophie A, Maeva L, Jean-François F, Jocelyn M, Emilia F, Sophie B, Cécile J, Gabrielle C, Audrey F, Simon L, Lena S, Pierre C, Aba M, Guillaume B. Impact of vaccination on the symptoms of hospitalised patients with SARS-CoV-2 delta variant (B.1.617.1) infection. Clin Microbiol Infect 2022; 28:1629-1635. [PMID: 35779764 PMCID: PMC9242694 DOI: 10.1016/j.cmi.2022.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The diffusion of the SARS-CoV-2 delta (B.1.617.2) variant and the waning of immune response after primary Covid-19 vaccination favoured the breakthrough SARS-CoV-2 infections in vaccinated subjects. To assess the impact of vaccination, we determined the severity of infection in hospitalised patients according to vaccine status. METHODS We performed a retrospectiveobservational study on patients hospitalised in 10 centres with a SARS-CoV-2 infection (delta variant) from July to November 2021 by including all patients who had completed their primary vaccination at least 14 days before hospital admission and the same number of completely unvaccinated patients. We assessed the impact of vaccination and other risk factors through logistic regression. RESULTS We included 955 patients (474 vaccinated and 481 unvaccinated). Vaccinated patients were significantly older (75.0 [63.25-84.0] vs. 55.0 [38.0-73.0]; p<0.001), more frequently males (55.1% (261/474) vs. 46.4% (223/481); p=0.009), and had more comorbidities (2.0 [1.0-3.0] vs. 1.0 [0.0-2.0]; p<0.001). Vaccinated patients were less often admitted for Covid-19 (59.3% (281/474) vs. 75.1% (361/481); p<0.001), had less extended lung lesions (≤25%: 64.3% (117/182) vs. 38.4% (88/229); p<0.001), required oxygen less frequently (57.5% (229/398) vs. 73.0% (270/370); p<0.001), at a lower flow (3.0 [0.0-8.7] vs. 6.0 [2.0-50.0] L/min, p<0.001), and for a shorter duration (3 [0.0-8.0] vs. 6 [2.0-12.0] days, p<0.001)., and required less frequently intensive care unit admission (16.2% (60/370) vs. 36.0% (133/369); p<0.001) but had comparable mortality in bivariate analysis (16.7% (74/443) vs. 12.2% (53/433); p=0.075). Multivariate logistic regression showed that vaccination significantly decreased the risk of death (0.38 [0.20-0.70](p=0.002), ICU admission (0.31 [0.21-0.47](p<0.001) and oxygen requirement (0.16 [0.10-0.26](p<0.001), even among older patients or with comorbidities. CONCLUSIONS Among patients hospitalised with a delta variant SARS-CoV-2 infection, vaccination was associated with less severe forms, even in the presence of comorbidities.
Collapse
Affiliation(s)
- Epaulard Olivier
- infectious diseases, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France; Groupe de Recherche en Infectiologie Clinique, CIC-1406, Inserm-CHUGA-UGA, Grenoble, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF).
| | - Abgrall Sophie
- Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF); APHP, Hôpital Antoine Béclère, Service de Médecine Interne, Clamart; Université Paris-Saclay, UVSQ, INSERM U1018, CESP, Le Kremlin-Bicêtre, France
| | - Lefebvre Maeva
- Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF); Infectious Diseases Department, Centre for Prevention of Infectious and Transmissible Diseases, CHU Nantes and INSERM UIC 1413 Nantes University, Nantes, France
| | - Faucher Jean-François
- CHU Limoges, Department of Infectious Diseases and Tropical Medicine, Limoges, France; Inserm U1094, IRD U270, Univ. Limoges, CHU Limoges, EpiMaCT; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Michon Jocelyn
- Department of Infectious diseases, University Hospital of Caen, Caen, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Frentiu Emilia
- Infectious diseases department, Centre Hospitalier Universitaire de Nancy, Nancy, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Blanchi Sophie
- Infectious diseases department, Centre Hospitalier, Le Mans, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Janssen Cécile
- Infectious Disease Unit, Centre Hospitalier Annecy Genevois, Annecy, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Charbonnier Gabrielle
- Infectious diseases department, Centre Hospitalier Universitaire Grenoble-Alpes, Grenoble, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Fresse Audrey
- Pharmacovigilance, Centre Hospitalier Universitaire de Nancy, Nancy, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Laurent Simon
- Department of Infectious diseases, University Hospital of Caen, Caen, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Sandjakian Lena
- CHU Limoges, Department of Infectious Diseases and Tropical Medicine, Limoges, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Casez Pierre
- Infectious Disease Unit, Centre Hospitalier Annecy Genevois, Annecy, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Mahamat Aba
- Regional Centre for Prevention of Infectious Diseases and Healthcare-Associated Infections, General Hospital of Ajaccio; Ajaccio, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| | - Beraud Guillaume
- Department of Internal Medicine and Infectious Diseases, University Hospital of Poitiers; Poitiers, France; Groupe Vaccination-Prévention de la Société de Pathologie Infectieuse de Langue Française (SPILF)
| |
Collapse
|
34
|
Higdon MM, Wahl B, Jones CB, Rosen JG, Truelove SA, Baidya A, Nande AA, ShamaeiZadeh PA, Walter KK, Feikin DR, Patel MK, Deloria Knoll M, Hill AL. A Systematic Review of Coronavirus Disease 2019 Vaccine Efficacy and Effectiveness Against Severe Acute Respiratory Syndrome Coronavirus 2 Infection and Disease. Open Forum Infect Dis 2022; 9:ofac138. [PMID: 35611346 PMCID: PMC9047227 DOI: 10.1093/ofid/ofac138] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 01/13/2023] Open
Abstract
Billions of doses of coronavirus disease 2019 (COVID-19) vaccines have been administered globally, dramatically reducing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) incidence and severity in some settings. Many studies suggest vaccines provide a high degree of protection against infection and disease, but precise estimates vary and studies differ in design, outcomes measured, dosing regime, location, and circulating virus strains. In this study, we conduct a systematic review of COVID-19 vaccines through February 2022. We included efficacy data from Phase 3 clinical trials for 15 vaccines undergoing World Health Organization Emergency Use Listing evaluation and real-world effectiveness for 8 vaccines with observational studies meeting inclusion criteria. Vaccine metrics collected include protection against asymptomatic infection, any infection, symptomatic COVID-19, and severe outcomes including hospitalization and death, for partial or complete vaccination, and against variants of concern Alpha, Beta, Gamma, Delta, and Omicron. We additionally review the epidemiological principles behind the design and interpretation of vaccine efficacy and effectiveness studies, including important sources of heterogeneity.
Collapse
Affiliation(s)
- Melissa M Higdon
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Brian Wahl
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Carli B Jones
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph G Rosen
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Shaun A Truelove
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anurima Baidya
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anjalika A Nande
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Parisa A ShamaeiZadeh
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Karoline K Walter
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Daniel R Feikin
- Department of Immunization, Vaccines, and Biologicals, World Health Organization, Geneva, Switzerland
| | - Minal K Patel
- Department of Immunization, Vaccines, and Biologicals, World Health Organization, Geneva, Switzerland
| | - Maria Deloria Knoll
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Alison L Hill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
35
|
See KC. Vaccination for the Prevention of Infection among Immunocompromised Patients: A Concise Review of Recent Systematic Reviews. Vaccines (Basel) 2022; 10:800. [PMID: 35632555 PMCID: PMC9144891 DOI: 10.3390/vaccines10050800] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Vaccination is crucial for avoiding infection-associated morbidity and mortality among immunocompromised patients. However, immunocompromised patients respond less well to vaccinations compared to healthy people, and little is known about the relative efficacy of various vaccines among different immunocompromised states. A total of 54 systematic reviews (22 COVID-19; 32 non-COVID-19) published within the last 5 years in Pubmed® were reviewed. They demonstrated similar patterns within three seroconversion response categories: good (about >60% when compared to healthy controls), intermediate (~40−60%), and poor (about <40%). Good vaccine responses would be expected for patients with chronic kidney disease, human immunodeficiency virus infection (normal CD4 counts), immune-mediated inflammatory diseases, post-splenectomy states, and solid tumors. Intermediate vaccine responses would be expected for patients with anti-cytotoxic T-lymphocyte antigen-4 therapy, hematologic cancer, and human immunodeficiency virus infection (low CD4 counts). Poor vaccine responses would be expected for patients with B-cell-depleting agents (e.g., anti-CD20 therapy), hematopoietic stem-cell transplant, solid organ transplant, and liver cirrhosis. For all vaccine response categories, vaccination should be timed when patients are least immunosuppressed. For the intermediate and poor vaccine response categories, high-dose vaccine, revaccination when patients are less immunosuppressed, checking for seroconversion, additional booster doses, and long-acting monoclonal antibodies may be considered, supplemented by shielding measures.
Collapse
Affiliation(s)
- Kay Choong See
- Division of Respiratory & Critical Care Medicine, Department of Medicine, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
36
|
Autoimmune post-COVID vaccine syndromes: does the spectrum of autoimmune/inflammatory syndrome expand? Clin Rheumatol 2022. [PMID: 35378658 DOI: 10.1007/s10067-022-06149-4/tables/1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
To date, around 60% of the world population has been protected by vaccines against SARS-CoV-2, significantly reducing the devastating effect of the pandemic and restoring social economic activity through mass vaccination. Multiple studies have demonstrated the effectiveness and safety of vaccines against COVID-19 in healthy populations, in people with risk factors, in people with or without SARS-CoV-2 infection, and in immunocompromised people. According to the criteria for post-vaccine adverse events established by the World Health Organization, a minority of individuals may develop adverse events, including autoimmune syndromes. The exact mechanisms for the development of these autoimmune syndromes are under study, and to date, a cause-effect relationship has not been established. Many of these autoimmune syndromes meet sufficient criteria for the diagnosis of Adjuvant-Induced Autoimmune Syndrome (ASIA syndrome). The descriptions of these autoimmune syndromes open new perspectives to the knowledge of the complex relationship between the host, its immune system, with the new vaccines and the development of new-onset autoimmune syndromes. Fortunately, most of these autoimmune syndromes are easily controlled with steroids and other immunomodulatory medications and are short-lived. Rheumatologists must be alert to the development of these autoimmune syndromes, and investigate the relationship between autoimmune/inflammatory symptoms and vaccination time, and assess their therapeutic response.
Collapse
|
37
|
Prevalence of Anti-SARS-CoV-2 Antibodies and Potential Determinants among the Belgian Adult Population: Baseline Results of a Prospective Cohort Study. Viruses 2022; 14:v14050920. [PMID: 35632663 PMCID: PMC9147735 DOI: 10.3390/v14050920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
The prevalence of anti-SARS-CoV-2 antibodies and potential determinants were assessed in a random sample representative of the Belgian adult population. In total, 14,201 individuals (≥18 years) were invited by mail to provide saliva via an Oracol® swab. Survey weights were applied, and potential determinants were estimated using multivariable logistic regressions. Between March and August 2021, 2767 individuals participated in the first data collection. During this period, which coincided with the onset of the vaccination campaign, the seroprevalence in the population increased from 25.2% in March/April to 78.1% in July. Among the vaccinated there was an increase from 74,2% to 98.8%; among the unvaccinated, the seroprevalence remained stable (around 17%). Among the vaccinated, factors significantly associated with the presence of antibodies were: having at least one chronic disease (ORa 0.22 (95% CI 0.08–0.62)), having received an mRNA-type vaccine (ORa 5.38 (95% CI 1.72–16.80)), and having received an influenza vaccine in 2020–2021 (ORa 3.79 (95% CI 1.30–11.07)). Among the unvaccinated, having a non-O blood type (ORa 2.00 (95% CI 1.09–3.67)) and having one or more positive COVID-19 tests (ORa 11.04 (95% CI 4.69–26.02)) were significantly associated. This study provides a better understanding of vaccine- and/or natural-induced presence of anti-SARS-CoV-2 antibodies and factors that are associated with this presence.
Collapse
|
38
|
Ferrand N, Accorinti M, Agarwal M, Spartalis C, Manni P, Stuebiger N, Zierhut M. COVID-19 Vaccination and Uveitis: Epidemiology, Clinical Features and Visual Prognosis. Ocul Immunol Inflamm 2022; 30:1265-1273. [DOI: 10.1080/09273948.2022.2058964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nawfel Ferrand
- Department of Ophthalmology, University Eye Hospital, Tuebingen, Germany
| | - Massimo Accorinti
- Ocular Immunovirology Service, Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Mamta Agarwal
- Uveitis & Cornea Services, Sankara Nethralaya, Chennai, India
| | - Christoph Spartalis
- Department of Ophthalmology, University Eye Hospital, Hamburg-Eppendorf, Germany
- Department of ophthalmology, Hanusch Hospital, Vienna, Austria
| | - Priscilla Manni
- Ocular Immunovirology Service, Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Nicole Stuebiger
- Department of Ophthalmology, University Eye Hospital, Hamburg-Eppendorf, Germany
| | - Manfred Zierhut
- Department of Ophthalmology, University Eye Hospital, Tuebingen, Germany
| |
Collapse
|
39
|
Autoimmune post-COVID vaccine syndromes: does the spectrum of autoimmune/inflammatory syndrome expand? Clin Rheumatol 2022; 41:1603-1609. [PMID: 35378658 PMCID: PMC8979721 DOI: 10.1007/s10067-022-06149-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 12/14/2022]
Abstract
To date, around 60% of the world population has been protected by vaccines against SARS-CoV-2, significantly reducing the devastating effect of the pandemic and restoring social economic activity through mass vaccination. Multiple studies have demonstrated the effectiveness and safety of vaccines against COVID-19 in healthy populations, in people with risk factors, in people with or without SARS-CoV-2 infection, and in immunocompromised people. According to the criteria for post-vaccine adverse events established by the World Health Organization, a minority of individuals may develop adverse events, including autoimmune syndromes. The exact mechanisms for the development of these autoimmune syndromes are under study, and to date, a cause-effect relationship has not been established. Many of these autoimmune syndromes meet sufficient criteria for the diagnosis of Adjuvant-Induced Autoimmune Syndrome (ASIA syndrome). The descriptions of these autoimmune syndromes open new perspectives to the knowledge of the complex relationship between the host, its immune system, with the new vaccines and the development of new-onset autoimmune syndromes. Fortunately, most of these autoimmune syndromes are easily controlled with steroids and other immunomodulatory medications and are short-lived. Rheumatologists must be alert to the development of these autoimmune syndromes, and investigate the relationship between autoimmune/inflammatory symptoms and vaccination time, and assess their therapeutic response.
Collapse
|
40
|
Schildgen V, Lüsebrink J, Schildgen O. Compulsary SARS-CoV-2 (booster-) vaccination in healthcare facilities can not replace personal protection measures while dealing with vulnarable individuals. J Infect 2022; 84:e111. [PMID: 35339503 PMCID: PMC8942570 DOI: 10.1016/j.jinf.2022.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Verena Schildgen
- Kliniken der Stadt Köln gGmbH, Institut für Pathologie, Klinikum der Privaten Universität Witten/Herdecke, Cologne, Germany
| | - Jessica Lüsebrink
- Kliniken der Stadt Köln gGmbH, Institut für Pathologie, Klinikum der Privaten Universität Witten/Herdecke, Cologne, Germany
| | - Oliver Schildgen
- Kliniken der Stadt Köln gGmbH, Institut für Pathologie, Klinikum der Privaten Universität Witten/Herdecke, Cologne, Germany.
| |
Collapse
|
41
|
Kratzer B, Trapin D, Gattinger P, Oberhofer T, Sehgal ANA, Waidhofer-Söllner P, Rottal A, Körmöczi U, Grabmeier-Pfistershammer K, Kopetzky GH, Tischer F, Valenta R, Pickl WF. Lack of Induction of RBD-Specific Neutralizing Antibodies despite Repeated Heterologous SARS-CoV-2 Vaccination Leading to Seroconversion and Establishment of T Cell-Specific Memory in a Patient in Remission of Multiple Myeloma. Vaccines (Basel) 2022; 10:vaccines10030374. [PMID: 35335006 PMCID: PMC8949333 DOI: 10.3390/vaccines10030374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Prophylactic vaccination against infectious diseases may induce a state of long-term protection in the otherwise healthy host. However, the situation is less predictable in immunocompromised patients and may require adjustment of vaccination schedules and/or basic therapy. Methods: A patient in full remission of multiple myeloma since the last three years and on long-term maintenance therapy with pomalidomide, a drug inhibiting angiogenesis and myeloma cell growth, was vaccinated twice with Comirnaty followed by two vaccinations with Vaxzevria. Seroconversion and SARS-CoV-2-specific cellular responses were monitored. Results: No signs of seroconversion or T cellular memory were observed after the first “full immunization” with Comirnaty. Consequently, long-term-maintenance therapy with Pomalidomide was stopped and two additional shots of Vaxzevria were administered after which the patient seroconverted with Spike(S)-protein specific antibody levels reaching 49 BAU/mL, mild S-peptide pool-specific T cell proliferation, effector cytokine production (IL-2, IL-13), and T cellular activation with increased numbers of CD3+CD4+CD25+ T cells as compared to vaccinated and non-vaccinated control subjects. However, despite suspension of immunosuppression and administration of in total four consecutive heterologous SARS-CoV-2 vaccine shots, the patient did not develop neutralizing RBD-specific antibodies. Conclusions: Despite immunomonitoring-based adjustment of vaccination and/or therapy schedules vaccination success, with clear correlates of protection, the development of RBD-specific antibodies could not be achieved in the immunocompromised patient with current SARS-CoV-2 vaccines. Thus, our report emphasizes the need for improved active and passive immunization strategies for SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
- Correspondence: (B.K.); (W.F.P.)
| | - Doris Trapin
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090Vienna, Austria; (P.G.); (R.V.)
| | - Teresa Oberhofer
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Al Nasar Ahmed Sehgal
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Petra Waidhofer-Söllner
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Arno Rottal
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Ulrike Körmöczi
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Katharina Grabmeier-Pfistershammer
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
| | - Gerhard H. Kopetzky
- 1st Med. Department Hemato-Oncology, Universitätsklinik St. Poelten, 3100 St. Poelten, Austria;
| | - Franz Tischer
- Landesklinikum Lilienfeld, 3180 Lilienfeld, Austria;
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090Vienna, Austria; (P.G.); (R.V.)
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Winfried F. Pickl
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.); (T.O.); (A.N.A.S.); (P.W.-S.); (A.R.); (U.K.); (K.G.-P.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- Correspondence: (B.K.); (W.F.P.)
| |
Collapse
|
42
|
Golshani M, Hrdý J. Multiple Sclerosis Patients and Disease Modifying Therapies: Impact on Immune Responses against COVID-19 and SARS-CoV-2 Vaccination. Vaccines (Basel) 2022; 10:vaccines10020279. [PMID: 35214735 PMCID: PMC8876554 DOI: 10.3390/vaccines10020279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
This article reviews the literature on SARS-CoV-2 pandemic and multiple sclerosis (MS). The first part of the paper focuses on the current data on immunopathology of SARS-CoV-2 and leading vaccines produced against COVID-19 infection. In the second part of the article, we discuss the effect of Disease Modifying Therapies (DMTs) on COVID-19 infection severity or SARS-CoV-2 vaccination in MS patients plus safety profile of different vaccine platforms in MS patients.
Collapse
Affiliation(s)
| | - Jiří Hrdý
- Correspondence: ; Tel.: +420-224968509
| |
Collapse
|