1301
|
Streinu-Cercel A, Săndulescu O, Preotescu LL, Kim JY, Kim YS, Cheon S, Jang YR, Lee SJ, Kim SH, Chang I, Suh JH, Lee SG, Kim MR, Chung DR, Kim HN, Streinu-Cercel A, Eom JS. Efficacy and Safety of Regdanvimab (CT-P59): A Phase 2/3 Randomized, Double-Blind, Placebo-Controlled Trial in Outpatients with Mild-to-Moderate Coronavirus Disease 2019. Open Forum Infect Dis 2022; 9:ofac053. [PMID: 35295819 PMCID: PMC8903348 DOI: 10.1093/ofid/ofac053] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/01/2022] [Indexed: 12/03/2022] Open
Abstract
Background Regdanvimab (CT-P59) is a monoclonal antibody with neutralizing activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We report on part 1 of a 2-part randomized, placebo-controlled, double-blind study for patients with mild-to-moderate coronavirus disease 2019 (COVID-19). Methods Outpatients with mild-to-moderate COVID-19 received a single dose of regdanvimab 40 mg/kg (n = 100), regdanvimab 80 mg/kg (n = 103), or placebo (n = 104). The primary end points were time to negative conversion of SARS-CoV-2 from nasopharyngeal swab based on quantitative reverse transcription polymerase chain reaction (RT-qPCR) up to day 28 and time to clinical recovery up to day 14. Secondary end points included the proportion of patients requiring hospitalization, oxygen therapy, or mortality due to COVID-19. Results Median (95% CI) time to negative conversion of RT-qPCR was 12.8 (9.0–12.9) days with regdanvimab 40 mg/kg, 11.9 (8.9–12.9) days with regdanvimab 80 mg/kg, and 12.9 (12.7–13.9) days with placebo. Median (95% CI) time to clinical recovery was 5.3 (4.0–6.8) days with regdanvimab 40 mg/kg, 6.2 (5.5–7.9) days with regdanvimab 80 mg/kg, and 8.8 (6.8–11.6) days with placebo. The proportion (95% CI) of patients requiring hospitalization or oxygen therapy was lower with regdanvimab 40 mg/kg (4.0% [1.6%–9.8%]) and regdanvimab 80 mg/kg (4.9% [2.1%–10.9%]) vs placebo (8.7% [4.6%–15.6%]). No serious treatment-emergent adverse events or deaths occurred. Conclusions Regdanvimab showed a trend toward a minor decrease in time to negative conversion of RT-qPCR results compared with placebo and reduced the need for hospitalization and oxygen therapy in patients with mild-to-moderate COVID-19. Clinical trial registration. NCT04602000 and EudraCT 2020-003369-20.
Collapse
Affiliation(s)
- Anca Streinu-Cercel
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Oana Săndulescu
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Liliana-Lucia Preotescu
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Jin Yong Kim
- Division of Infectious Diseases, Department of Internal Medicine, Incheon Medical Center, Republic of Korea
| | - Yeon-Sook Kim
- Division of Infectious Diseases, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Shinhye Cheon
- Division of Infectious Diseases, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Young Rock Jang
- Division of Infectious Diseases, Department of Internal Medicine, Incheon Medical Center, Republic of Korea
| | | | | | | | | | | | - Mi Rim Kim
- Celltrion, Inc., Incheon, Republic of Korea
| | | | - Han Na Kim
- Celltrion, Inc., Incheon, Republic of Korea
| | - Adrian Streinu-Cercel
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Joong Sik Eom
- Division of Infectious Diseases, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| |
Collapse
|
1302
|
Jeewandara C, Fernando S, Pushpakumara PD, Ramu ST, Kamaladasa A, Gunasekara B, Aberathna IS, Kuruppu H, Ranasinghe T, Dayarathne S, Dissanayake O, Gamalath N, Ekanayake D, Jayamali J, Wijesinghe A, Dissanayake M, Somathilake G, Harvie M, Danasekara S, Jayathilaka D, Wijayatilake HDK, Weerasooriya N, Kekulandara C, Schimanski L, Rijal P, Tan TK, Dong T, Townsend A, Ogg GS, Malavige GN. Immune responses following the first dose of the Sputnik V (Gam-COVID-Vac). Sci Rep 2022; 12:1727. [PMID: 35110645 PMCID: PMC8810924 DOI: 10.1038/s41598-022-05788-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
As the first dose of Gam-COVID-Vac, is currently used as a single dose vaccine in some countries, we investigated the immunogenicity of this at 4 weeks (327 naïve individuals). 88.7% seroconverted, with significantly lower seroconversion rates in those over 60 years (p = 0.004) and significantly lower than previously seen with AZD1222 (p = 0.018). 82.6% developed ACE2 receptor blocking antibodies, although levels were significantly lower than following natural infection (p = 0.0009) and a single dose of AZD1222 (p < 0.0001). Similar titres of antibodies were observed to the receptor binding domain of WT, B.1.1.7 and B.1.617.2 compared to AZD1222, while the levels for B.1.351 were significantly higher (p = 0.006) for Gam-COVID-Vac. 30% developed ex vivo IFNγ ELISpot responses (significantly lower than AZD1222), and high frequency of CD107a expressing T cells along with memory B cell responses. Although single dose of Gam-COVID-Vac was highly immunogenic, administration of a second dose is likely to be beneficial.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | - Pradeep Darshana Pushpakumara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shyrar Tanussiya Ramu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Banuri Gunasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Inoka Sepali Aberathna
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Heshan Kuruppu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Thushali Ranasinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shashika Dayarathne
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Osanda Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Nayanathara Gamalath
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Dinithi Ekanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Jewantha Jayamali
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Ayesha Wijesinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Madushika Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Gayasha Somathilake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Michael Harvie
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Saubhagya Danasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Deshni Jayathilaka
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | | | | | - Lisa Schimanski
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Pramila Rijal
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tiong K Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Alain Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka.
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
1303
|
Luan N, Wang Y, Cao H, Lin K, Liu C. Comparison of immune responses induced by two or three doses of an alum-adjuvanted inactivated SARS-CoV-2 vaccine in mice. J Med Virol 2022; 94:2250-2258. [PMID: 35112364 PMCID: PMC9015325 DOI: 10.1002/jmv.27637] [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: 01/18/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/27/2022]
Abstract
Waning antibodies and rapidly emerging variants are challenges for severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) vaccine development. Adjusting existing immunization schedules and further boosting strategies are under consideration. Here, the immune responses induced by an alum‐adjuvanted inactivated SARS‐CoV‐2 vaccine in mice were compared among immunization schedules with two or three doses. For the two‐dose schedule, a 0–28‐day schedule induced 5‐fold stronger spike‐specific IgG responses than a 0–14‐day schedule, with only a slight elevation of spike‐specific cellular immunity 14 days after the last immunization. A third homologous boost 2 or 5 months after the second dose for the 0–28‐day schedule slightly strengthened humoral responses (1.3‐fold for the 0–1–3‐month schedule, and 1.8‐fold for the 0–1–6‐month schedule) 14 days after the last immunization. Additionally, a third homologous boost (especially with the 0–1–3‐month schedule) induced significantly stronger cell‐mediated immunity than both two‐dose immunization schedules for all indexes tested, with a response similar to that induced by a one‐dose heterologous boost with BNT162b2 in clinical trials, according to cellular immunity analysis (1.5‐fold). These T cell responses were Th2 oriented, with good CD4+ and CD8+ memory. These results may offer clues for applying a homologous boosting strategy for alum‐adjuvanted inactivated SARS‐CoV‐2 vaccines.
Collapse
Affiliation(s)
- Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| |
Collapse
|
1304
|
Zinatizadeh MR, Zarandi PK, Zinatizadeh M, Yousefi MH, Amani J, Rezaei N. Efficacy of mRNA, adenoviral vector, and perfusion protein COVID-19 vaccines. Biomed Pharmacother 2022; 146:112527. [PMID: 34906769 PMCID: PMC8660177 DOI: 10.1016/j.biopha.2021.112527] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has a devastating impact on global populations triggered by a highly infectious viral sickness, produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The third major cause of mortality in the United States, following heart disease and cancer in 2020, was undoubtedly COVID-19. The centers for disease control and prevention (CDC) and the world health organization (WHO) separately developed a categorization system for differentiating new strains of SARS-CoV-2 into variants of concern (VoCs) and variants of interest (VoIs) with the continuing development of various strains SARS-CoV-2. By December 2021, five of the SARS-CoV-2 VoCs were discovered from the onset of the pandemic depending on the latest epidemiologic report by the WHO: Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529). Mutations in the receptor-binding domain (RBD) and n-terminal domain (NTD) have been found throughout all five identified VoCs. All strains other than the delta mutant are often found with the N501Y mutation situated on the RBD, resulting in higher binding between the spike protein and angiotensin-converting enzyme 2 (ACE2) receptors, enhanced viral adhesion, and following the entrance to host cells. The introduction of these new strains of SRAS-CoV-2 is likely to overcome the remarkable achievements gained in restricting this viral disease to the point where it is presented with remarkable vaccine developments against COVID-19 and strong worldwide mass immunization initiatives. Throughout this literature review, the effectiveness of current COVID-19 vaccines for managing and prohibiting SARS-CoV-2 strains is thoroughly described.
Collapse
Affiliation(s)
- Mohammad Reza Zinatizadeh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran; Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran; Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Zinatizadeh
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Hadi Yousefi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Jaffar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
1305
|
Galmiche S, Luong Nguyen LB, Tartour E, de Lamballerie X, Wittkop L, Loubet P, Launay O. Immunological and clinical efficacy of COVID-19 vaccines in immunocompromised populations: a systematic review. Clin Microbiol Infect 2022; 28:163-177. [PMID: 35020589 PMCID: PMC8595936 DOI: 10.1016/j.cmi.2021.09.036] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Available data show that COVID-19 vaccines may be less effective in immunocompromised populations, who are at increased risk of severe COVID-19. OBJECTIVES We conducted a systematic review of literature to assess immunogenicity, efficacy and effectiveness of COVID-19 vaccines in immunocompromised populations. DATA SOURCES We searched Medline and Embase databases. STUDY ELIGIBILITY CRITERIA, PATIENTS, INTERVENTIONS We included studies of COVID-19 vaccines after complete vaccination in immunocompromised patients until 31 August 2021. Studies with <10 patients, safety data only and case series of breakthrough infections were excluded. METHODS Risk of bias was assessed via the tool developed by the National Institutes of Health on interventional and observational studies. Immunogenicity was assessed through non-response rate defined as no anti-SARS-CoV-2 spike protein antibodies, efficacy and effectiveness by the relative reduction in risk of SARS-CoV-2 infection or COVID-19. We collected factors associated with the risk of non-response. We presented collected data by immunosuppression type. RESULTS We screened 5917 results, included 162 studies. There were 157 on immunogenicity in 25 209 participants, including 7835 cancer or haematological malignancy patients (31.1%), 6302 patients on dialysis (25.0%), 5974 solid organ transplant recipients (23.7%) and 4680 immune-mediated disease patients (18.6%). Proportion of non-responders seemed higher among solid organ transplant recipients (range 18-100%) and patients with haematological malignancy (range 14-61%), and lower in patients with cancer (range 2-36%) and patients on dialysis (range 2-30%). Risk factors for non-response included older age, use of corticosteroids, immunosuppressive or anti-CD20 agent. Ten studies evaluated immunogenicity of an additional dose. Five studies evaluated vaccine efficacy or effectiveness: three on SARS-CoV-2 infection (range 71-81%), one on COVID-19-related hospitalization (62.9%), one had a too small sample size. CONCLUSIONS This systematic review highlights the risk of low immunogenicity of COVID-19 vaccines in immunocompromised populations, especially solid organ transplant recipients and patients with haematological malignancy. Despite lack of vaccine effectiveness data, enhanced vaccine regimens may be necessary.
Collapse
Affiliation(s)
- Simon Galmiche
- Assistance Publique - Hôpitaux de Paris (AP-HP), CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Liem Binh Luong Nguyen
- Assistance Publique - Hôpitaux de Paris (AP-HP), CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- AP-HP, Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Xavier de Lamballerie
- Aix Marseille Université, IRD 190, INSERM 1207, Unité des Virus Emergents, UVE, IHU Méditerranée Infection, Marseille, France
| | - Linda Wittkop
- Institut de Santé Publique d'Epidémiologie et de Développement, INSERM, Bordeaux Population Health Research Center, UMR 1219, Centre d'Investigation Clinique-Epidémiologie Clinique 1401, University of Bordeaux, Service d'Information Médicale, CHU de Bordeaux, Pôle de Santé Publique, Bordeaux, France
| | - Paul Loubet
- INSERM U1047, Department of Infectious and Tropical Diseases, CHU Nîmes, Université Montpellier, Nîmes, France
| | - Odile Launay
- Université de Paris, Faculté de Médecine Paris Descartes, AP-PH, Inserm, CIC Cochin Pasteur, Paris, France.
| |
Collapse
|
1306
|
Reindl-Schwaighofer R, Heinzel A, Mayrdorfer M, Jabbour R, Hofbauer TM, Merrelaar A, Eder M, Regele F, Doberer K, Spechtl P, Aschauer C, Koblischke M, Paschen C, Eskandary F, Hu K, Öhler B, Bhandal A, Kleibenböck S, Jagoditsch RI, Reiskopf B, Heger F, Bond G, Böhmig GA, Strassl R, Weseslindtner L, Indra A, Aberle JH, Binder M, Oberbauer R. Comparison of SARS-CoV-2 Antibody Response 4 Weeks After Homologous vs Heterologous Third Vaccine Dose in Kidney Transplant Recipients: A Randomized Clinical Trial. JAMA Intern Med 2022; 182:165-171. [PMID: 34928302 PMCID: PMC8689434 DOI: 10.1001/jamainternmed.2021.7372] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IMPORTANCE Fewer than 50% of kidney transplant recipients (KTRs) develop antibodies against the SARS-CoV-2 spike protein after 2 doses of an mRNA vaccine. Preliminary data suggest that a heterologous vaccination, combining mRNA and viral vector vaccines, may increase immunogenicity. OBJECTIVE To assess the effectiveness of a third dose of an mRNA vs a vector vaccine in KTRs who did not have antibodies against the SARS-CoV-2 spike protein after 2 doses of an mRNA vaccine. DESIGN, SETTING, AND PARTICIPANTS This was a single center, single-blinded, 1:1 randomized clinical trial of a third dose of vaccine against SARS-CoV-2, conducted from June 15 to August 16, 2021, in 201 KTRs who had not developed SARS-CoV-2 spike protein antibodies after 2 doses of an mRNA vaccine. Data analyses were performed from August 17 to August 31, 2021. INTERVENTIONS mRNA (BNT162b2 or mRNA-1273) or vector (Ad26COVS1) as a third dose of a SARS-CoV-2 vaccine. MAIN OUTCOMES AND MEASURES The primary study end point was seroconversion after 4 weeks (29-42 days) following the third vaccine dose. Secondary end points included neutralizing antibodies and T-cell response assessed by interferon-γ release assays (IGRA). In addition, the association of patient characteristics and vaccine response was assessed using logistic regression, and the reactogenicity of the vaccines was compared. RESULTS Among the study population of 197 kidney transplant recipients (mean [SD] age, 61.2 [12.4] years; 82 [42%] women), 39% developed SARS-CoV-2 antibodies after the third vaccine. There was no statistically significant difference between groups, with an antibody response rate of 35% and 42% for the mRNA and vector vaccines, respectively. Only 22% of seroconverted patients had neutralizing antibodies. Similarly, T-cell response assessed by IGRA was low with only 17 patients showing a positive response after the third vaccination. Receiving nontriple immunosuppression (odds ratio [OR], 3.59; 95% CI, 1.33-10.75), longer time after kidney transplant (OR, 1.44; 95% CI, 1.15-1.83, per doubling of years), and torque teno virus plasma levels (OR, 0.92; 95% CI, 0.88-0.96, per doubling of levels) were associated with vaccine response. The third dose of an mRNA vaccine was associated with a higher frequency of local pain at the injection site compared with the vector vaccine, while systemic symptoms were comparable between groups. CONCLUSIONS AND RELEVANCE This randomized clinical trial found that 39% of KTRs without an immune response against SARS-CoV-2 after 2 doses of an mRNA vaccine developed antibodies against the SARS-CoV-2 spike protein 4 weeks after a third dose of an mRNA or a vector vaccine. The heterologous vaccination strategy with a vector-based vaccine was well tolerated and safe but not significantly better than the homologous mRNA-based strategy. TRIAL REGISTRATION EudraCT Identifier: 2021-002927-39.
Collapse
Affiliation(s)
- Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Manuel Mayrdorfer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Rhea Jabbour
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas M Hofbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Anne Merrelaar
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Eder
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florina Regele
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Paul Spechtl
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Constantin Aschauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Christopher Paschen
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Hu
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Barbara Öhler
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Arshdeep Bhandal
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sabine Kleibenböck
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Rahel I Jagoditsch
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bianca Reiskopf
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florian Heger
- Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Gregor Bond
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Robert Strassl
- Division of Virology, Department of Laboratory Medicine, Medical University Vienna, Vienna, Austria
| | | | - Alexander Indra
- Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria.,Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Judith H Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
1307
|
Zhang S, Chen S, Xiao G, Zhao M, Li J, Dong W, Hu J, Yuan T, Li Y, Liu L. The associations between air pollutant exposure and neutralizing antibody titers of an inactivated SARS-CoV-2 vaccine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:13720-13728. [PMID: 34599446 PMCID: PMC8486374 DOI: 10.1007/s11356-021-16786-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Air pollution is a critical risk factor for the prevalence of COVID-19. However, few studies have focused on whether air pollution affects the efficacy of the SARS-CoV-2 vaccine. To better guide the knowledge surrounding this vaccination, we conducted a cross-section study to identify the relationships between air pollutant exposure and plasma neutralizing antibody (NAb) titers of an inactivated SARS-CoV-2 vaccine (Vero cell, CoronaVac, SINOVΛC, China). We recruited 239 healthcare workers aged 21-50 years who worked at Suining Central Hospital. Of these, 207 were included in this study, depending on vaccination date. The data regarding air pollutants were collected to calculate individual daily exposure dose (DED). The geometric mean of all six pollutant DEDs was applied to estimate the combined toxic effects (DEDcomplex). Then, the participants were divided into two groups based on the mean value of DEDcomplex. The median plasma NAb titer was 12.81 AU/mL, with 85.99% vaccine efficacy in healthcare workers against SARS-CoV-2. In exposure group, observations included lower plasma NAb titers (median: 11.13 AU/mL vs. 14.56 AU/mL), more peripheral counts of white blood cells and monocytes (mean: 6.71 × 109/L vs. 6.29 × 109/L and 0.49 × 109/L vs. 0.40 × 109/L, respectively), and a higher peripheral monocyte ratio (7.38% vs. 6.50%) as compared to the reference group. In addition, elevated air pollutant DEDs were associated with decreased plasma NAb titers. To our knowledge, this study is the first to report the relationship between air pollutant exposure and plasma NAb titers of the SARS-CoV-2 vaccine. This suggests that long-term exposure to air pollutants may inhibit plasma NAb expression by inducing chronic inflammation. Therefore, to achieve early herd immunity and hopefully curb the COVID-19 epidemic, vaccinations should be administered promptly to those eligible, and environmental factors should be considered as well.
Collapse
Affiliation(s)
- Shaocheng Zhang
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China.
| | - Shu Chen
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| | - Guangjun Xiao
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| | - Mingcai Zhao
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| | - Jia Li
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| | - Wenjuan Dong
- Department of Public Health Administration and Health Education, Suining Central Hospital, Suining, 629000, Sichuan, People's Republic of China
| | - Juan Hu
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| | - Tianqi Yuan
- Maccura Biotechnology Co. Ltd., Chengdu, 611731, Sichuan, People's Republic of China
| | - Yong Li
- Maccura Biotechnology Co. Ltd., Chengdu, 611731, Sichuan, People's Republic of China
| | - Lianghua Liu
- Department of Clinical Laboratory Medicine, Suining Central Hospital, 127 Deshengxi Rd., Suining, 629000, Sichuan, People's Republic of China
| |
Collapse
|
1308
|
Jiang L, Driedonks TA, Jong WS, Dhakal S, van den Berg van Saparoea HB, Sitaras I, Zhou R, Caputo C, Littlefield K, Lowman M, Chen M, Lima G, Gololobova O, Smith B, Mahairaki V, Richardson MR, Mulka KR, Lane AP, Klein SL, Pekosz A, Brayton CF, Mankowski JL, Luirink J, Villano JS, Witwer KW. A bacterial extracellular vesicle-based intranasal vaccine against SARS-CoV-2 protects against disease and elicits neutralizing antibodies to wild-type and Delta variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.06.28.450181. [PMID: 35132418 PMCID: PMC8820665 DOI: 10.1101/2021.06.28.450181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Several vaccines have been introduced to combat the coronavirus infectious disease-2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current SARS-CoV-2 vaccines include mRNA-containing lipid nanoparticles or adenoviral vectors that encode the SARS-CoV-2 Spike (S) protein of SARS-CoV-2, inactivated virus, or protein subunits. Despite growing success in worldwide vaccination efforts, additional capabilities may be needed in the future to address issues such as stability and storage requirements, need for vaccine boosters, desirability of different routes of administration, and emergence of SARS-CoV-2 variants such as the Delta variant. Here, we present a novel, well-characterized SARS-CoV-2 vaccine candidate based on extracellular vesicles (EVs) of Salmonella typhimurium that are decorated with the mammalian cell culture-derived Spike receptor-binding domain (RBD). RBD-conjugated outer membrane vesicles (RBD-OMVs) were used to immunize the golden Syrian hamster ( Mesocricetus auratus ) model of COVID-19. Intranasal immunization resulted in high titers of blood anti-RBD IgG as well as detectable mucosal responses. Neutralizing antibody activity against wild-type and Delta variants was evident in all vaccinated subjects. Upon challenge with live virus, hamsters immunized with RBD-OMV, but not animals immunized with unconjugated OMVs or a vehicle control, avoided body mass loss, had lower virus titers in bronchoalveolar lavage fluid, and experienced less severe lung pathology. Our results emphasize the value and versatility of OMV-based vaccine approaches.
Collapse
Affiliation(s)
- Linglei Jiang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christopher Caputo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maggie Lowman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mengfei Chen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gabriela Lima
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Smith
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine and The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M. Riley Richardson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew P. Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Cory F. Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph L. Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joen Luirink
- Abera Bioscience AB, Uppsala, Sweden
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
- These authors are designated as co-corresponding authors. ; ;
| | - Jason S. Villano
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors are designated as co-corresponding authors. ; ;
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine and The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors are designated as co-corresponding authors. ; ;
| |
Collapse
|
1309
|
Périnet S, Cadieux G, Mercure SA, Drouin M, Allard R. Analysis of COVID-19 Risk Following a Ring Vaccination Intervention to Address SARS-CoV-2 Alpha Variant Transmission in Montreal, Canada. JAMA Netw Open 2022; 5:e2147042. [PMID: 35147688 PMCID: PMC8837915 DOI: 10.1001/jamanetworkopen.2021.47042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
IMPORTANCE Given limited COVID-19 vaccine availability early in the pandemic, optimizing immunization strategies was of paramount importance. Ring vaccination has been used successfully to control transmission of other airborne respiratory viruses. OBJECTIVE To assess the association of a ring vaccination intervention on COVID-19 spread in the initial epicenter of SARS-CoV-2 Alpha variant transmission in Montreal, Canada. DESIGN, SETTING, AND PARTICIPANTS This cohort study compared COVID-19 daily disease risk in 3 population-based groups of neighborhoods in Montreal, Canada, defined by their intervention-specific vaccine coverage at the neighborhood level: the primary intervention group (500 or more vaccinated persons per 10 000 persons), secondary intervention group (95 to 499), and control group (0 to 50). The groups were compared within each of 3 time periods: before intervention (December 1, 2020, to March 16, 2021), during and immediately after intervention (March 17 to April 17, 2021), and 3 weeks after the intervention midpoint (April 18 to July 18, 2021). Data were analyzed between June 2021 and November 2021. EXPOSURES Vaccination targeted parents and teachers of children attending the 32 schools and 48 childcare centers in 2 adjacent neighborhoods with highest local transmission (case counts) of Alpha variant shortly after its introduction. Participants were invited to receive 1 dose of mRNA vaccine between March 22 and April 9, 2021 (before vaccine was available to these age groups). MAIN OUTCOMES AND MEASURES COVID-19 risk in 3 groups of neighborhoods based on intervention-specific vaccine coverage. RESULTS A total of 11 794 residents were immunized, with a mean (SD) age of 43 (8) years (range, 16-93 years); 5766 participants (48.9%) lived in a targeted neighborhood, and 9784 (83.0%) were parents. COVID-19 risk in the primary intervention group was significantly higher than in the control group before (unadjusted risk ratio [RR], 1.58; 95% CI 1.52-1.65) and during (RR, 1.63; 95% CI, 1.52-1.76) intervention, and reached a level similar to the other groups in the weeks following the intervention (RR, 1.03; 95% CI, 0.94-1.12). A similar trend was observed when restricting to SARS-CoV-2 variants and persons aged 30 to 59 years (before: RR, 1.72; 95% CI, 1.63-1.83 vs after: RR, 1.01; 95% CI, 0.88-1.17). CONCLUSIONS AND RELEVANCE Our findings show that ring vaccination was associated with a reduction in COVID-19 risk in areas with high local transmission of Alpha variant shortly after its introduction. Ring vaccination may be considered as an adjunct to mass immunization to control transmission in specific areas, based on local epidemiology.
Collapse
Affiliation(s)
- Simone Périnet
- Direction régionale de santé publique de Montréal, Montreal, Quebec, Canada
- Canadian Field Epidemiology Program, Public Health Agency of Canada, Canada
| | - Geneviève Cadieux
- Direction régionale de santé publique de Montréal, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | | | - Mylène Drouin
- Direction régionale de santé publique de Montréal, Montreal, Quebec, Canada
- École de santé publique, Université de Montréal, Montreal, Quebec, Canada
| | - Robert Allard
- Direction régionale de santé publique de Montréal, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
1310
|
Gounant V, Ferré VM, Soussi G, Charpentier C, Flament H, Fidouh N, Collin G, Namour C, Assoun S, Bizot A, Brouk Z, Vicaut E, Teixeira L, Descamps D, Zalcman G. Efficacy of Severe Acute Respiratory Syndrome Coronavirus-2 Vaccine in Patients With Thoracic Cancer: A Prospective Study Supporting a Third Dose in Patients With Minimal Serologic Response After Two Vaccine Doses. J Thorac Oncol 2022; 17:239-251. [PMID: 34798306 PMCID: PMC8593625 DOI: 10.1016/j.jtho.2021.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/10/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Coronavirus disease 2019 resulted in a 30% mortality rate in patients with thoracic cancer. Given that patients with cancer were excluded from serum antisevere acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccine registration trials, it is still unknown whether they would develop a protective antispike antibody response after vaccination. This prospective vaccine monitoring study primarily aimed to assess humoral responses to the SARS-CoV-2 vaccine in patients with thoracic cancer. METHODS SARS-CoV-2-spike antibodies were measured using the Abbot Architect SARS-CoV-2 immunoglobulin G immunoassay before the first injection of BNT162b2 mRNA vaccine, at week 4, and 2 to 16 weeks after the second vaccine dose administration. The factors associated with antibody response were analyzed. RESULTS Overall, 306 patients, with a median age of 67.0 years (interquartile range: 58-74), were vaccinated. Of these, 283 patients received two vaccine doses at 28-day intervals. After a 6.7-month median follow-up, eight patients (2.6%) contracted proven symptomatic SARS-CoV-2 infection, with rapid favorable evolution. Of the 269 serologic results available beyond day 14 after the second vaccine dose administration, 17 patients (6.3%) were still negative (<50 arbitrary units/mL, whereas 34 (11%) were less than 300 arbitrary units/mL (12.5th percentile). In multivariate analysis, only age (p < 0.01) and long-term corticosteroid treatment (p = 0.01) were significantly associated with a lack of immunization. A total of 30 patients received a third vaccine dose, with only three patients showing persistently negative serology thereafter, whereas the others exhibited clear seroconversion. CONCLUSIONS SARS-CoV2 vaccines were found to be efficient in patients with thoracic cancer, most of them being immunized after two doses. A third shot given to 1% of patients with persistent low antibody titers resulted in an 88% immunization rate.
Collapse
Affiliation(s)
- Valérie Gounant
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Valentine Marie Ferré
- Virology Department, Université de Paris, INSERM Unité Mixte de Recherche (UMR) 1137 Infection, Antimicrobials, Modelling, Evolution, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Ghassen Soussi
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Charlotte Charpentier
- Virology Department, Université de Paris, INSERM Unité Mixte de Recherche (UMR) 1137 Infection, Antimicrobials, Modelling, Evolution, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Héloïse Flament
- Hematology, and Immunology Department, Université de Paris, INSERM-U1149, Research Center on Inflammation (CRI) Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Nadhira Fidouh
- Virology Department, Université de Paris, INSERM Unité Mixte de Recherche (UMR) 1137 Infection, Antimicrobials, Modelling, Evolution, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Gilles Collin
- Virology Department, Université de Paris, INSERM Unité Mixte de Recherche (UMR) 1137 Infection, Antimicrobials, Modelling, Evolution, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Céline Namour
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Sandra Assoun
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Alexandra Bizot
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Zohra Brouk
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Eric Vicaut
- Biostatistics and Clinical Research Department, Université de Paris, Lariboisière Hospital (AP-HP.Nord), Paris, France
| | - Luis Teixeira
- Breast Diseases Centre, North-Paris Cancer University Institute, Université de Paris, INSERM U976 Human Immunology, Pathophysiology, Immunotherapy (HIPI) (Pathophysiology of Breast Cancer Team), Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Diane Descamps
- Virology Department, Université de Paris, INSERM Unité Mixte de Recherche (UMR) 1137 Infection, Antimicrobials, Modelling, Evolution, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France
| | - Gérard Zalcman
- Thoracic Oncology Department, Université de Paris, North-Paris Cancer University Institute, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Investigation Clinique 1425, Bichat-Claude Bernard Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP.Nord), Paris, France; INSERM U830 Cancer, Heterogeneity Plasticity, Curie Institute Research Centre, Paris, France.
| |
Collapse
|
1311
|
Yang Y, Iwasaki A. Impact of Chronic HIV Infection on SARS-CoV-2 Infection, COVID-19 Disease and Vaccines. Curr HIV/AIDS Rep 2022; 19:5-16. [PMID: 34843064 PMCID: PMC8628277 DOI: 10.1007/s11904-021-00590-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has developed into a global pandemic that affect the health of hundreds of millions worldwide. In particular, SARS-CoV-2 infection in people with chronic human immune deficiency virus (HIV) infection is of concern, due to their already immunocompromised status. Yet, whether and how the immunological changes brought about by HIV will affect the immune responses against SARS-CoV-2 acute infection and impact the effectiveness of vaccines remain unclear. We discuss the intersection of COVID-19 in HIV-infected individuals. RECENT FINDINGS People living with HIV (PLWH) may be at increased risk of severe SARS-CoV-2 mediated disease complication due to functional impairment of the immune system and persistent inflammation, which can be ameliorated by antiretroviral therapy. Importantly, limited data suggest that current approved vaccines may be safe and efficacious in PLWH. To address remaining questions and supplement limited experimental evidence, more studies examining the interplay between HIV and SARS-CoV-2 through their impact on the host immune system are required.
Collapse
Affiliation(s)
- Yexin Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
1312
|
Peter J, Day C, Takuva S, Takalani A, Engelbrecht I, Garrett N, Goga A, Louw V, Opie J, Jacobson B, Sanne I, Gail-Bekker L, Gray G. Allergic reactions to the Ad26.COV2.S vaccine in South Africa. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2022; 1:2-8. [PMID: 37780073 PMCID: PMC10509997 DOI: 10.1016/j.jacig.2021.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 10/03/2023]
Abstract
Background The Janssen-Ad26.COV2.S vaccine is authorized for use in several countries, with more than 30 million doses administered. Mild and severe allergic adverse events following immunization (AEFI) have been reported. Objective We sought to detail allergic reactions reported during the Sisonke phase 3B study in South Africa. Methods A single dose of the Ad26.COV2.S vaccine was administered to 4,77,234 South African health care workers between February 17 and May 17, 2021. Monitoring of adverse events used a combination of passive reporting and active case finding. Telephonic contact was attempted for all adverse events reported as "allergy." Anaphylaxis adjudication was performed using the Brighton Collaboration and National Institute of Allergy and Infectious Disease case definitions. Results Only 251 (0.052%) patients reported any allergic-type reaction (<1 in 2000), with 4 cases of adjudicated anaphylaxis (Brighton Collaboration level 1, n = 3) (prevalence of 8.4 per million doses). All anaphylaxis cases had a previous history of drug or vaccine-associated anaphylaxis. Cutaneous allergic reactions were the commonest nonanaphylatic reactions and included self-limiting, transient/localized rashes requiring no health care contact (n = 92) or isolated urticaria and/or angioedema (n = 70; median onset, 48 [interquartile range, 11.5-120] hours postvaccination) that necessitated health care contact (81%), antihistamine (63%), and/or systemic/topical corticosteroid (16%). All immediate (including adjudicated anaphylaxis) and most delayed AEFI (65 of 69) cases resolved completely. Conclusions Allergic AEFI are rare following a single dose of Ad26.COV, with complete resolution in all cases of anaphylaxis. Although rare, isolated, delayed-onset urticaria and/or angioedema was the commonest allergic AEFI requiring treatment, with nearly half occurring in participants without known atopic disease.
Collapse
Affiliation(s)
- Jonny Peter
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Groote Schuur Hospital, Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Cascia Day
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Groote Schuur Hospital, Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Simbarashe Takuva
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Wash
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Azwidihwi Takalani
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Wash
- Department of Family Medicine and Primary Care, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Nigel Garrett
- CAPRISA, Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - Ameena Goga
- HIV Prevention Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| | - Vernon Louw
- Groote Schuur Hospital, Cape Town, South Africa
- Division of Clinical Haematology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jessica Opie
- Division of Haematology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Barry Jacobson
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
- the Department of Haematology and Molecular Medicine, University of the Witwatersrand, Johannesburg, South Africa
- Charlotte Maxeke Academic Hospital, Johannesburg, South Africa
| | - Ian Sanne
- Right to Care, Johannesburg, South Africa
| | - Linda Gail-Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
1313
|
Massarweh A, Tschernichovsky R, Stemmer A, Benouaich-Amiel A, Siegal T, Eliakim-Raz N, Stemmer SM, Yust-Katz S. Immunogenicity of the BNT162b2 mRNA COVID-19 vaccine in patients with primary brain tumors: a prospective cohort study. J Neurooncol 2022; 156:483-489. [PMID: 35018613 PMCID: PMC8752179 DOI: 10.1007/s11060-021-03911-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/22/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE Immunogenicity of Covid-19 vaccines may be negatively impacted by anti-cancer treatment. The management of primary brain tumors (PBTs) routinely includes temozolomide and steroids, which are immune-suppressive. We aimed to determine the rate of seropositivity in PBT patients following receipt of two doses of the BNT162b2 vaccine. METHODS We prospectively evaluated IgG levels against SARS-CoV-2 spike protein in 17 PBT patients following two doses of the BNT162b2 vaccine. IgG levels were collected at two time points: T1-after a median of 44 days from the second vaccine dose and T2-after a median of 130 days from the second dose. Titers were compared against a group of healthy controls (HC) comprised of patients' family members. RESULTS At T1, 88.2% (15/17) of PBT patients achieved seroconversion, compared with 100% (12/12) of HCs. Median IgG titer was significantly lower in the PBT group (1908 AU/mL vs 8,198 AU/mL; p = 0.002). At T2, 80% (12/15) of PBT patients seroconverted, compared to 100% (10/10) of HCs. Median IgG titer remained significantly lower in the PBT group (410 AU/mLvs 1687 AU/mL; p = 0.002). During the peri-vaccination period, 15 patients received systemic treatment and 8 patients were treated with corticosteroids. All 3 patients who failed to seroconvert at T2 were treated with corticosteroids. In a univariate analysis, steroid use was negatively associated with antibody titer. CONCLUSION Most PBT patients successfully seroconvert following two doses of the BNT162b2 vaccine, albeit with lower antibody titer compared to HCs. Steroid use during the vaccination period is associated with lower titer.
Collapse
Affiliation(s)
- Amir Massarweh
- Department of Oncology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Roi Tschernichovsky
- Department of Oncology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel.
| | - Amos Stemmer
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Alexandra Benouaich-Amiel
- Neuro-Oncology Unit, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Tali Siegal
- Neuro-Oncology Unit, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Noa Eliakim-Raz
- Department of Medicine E, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Infectious Disease Unit, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Salomon M Stemmer
- Department of Oncology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shlomit Yust-Katz
- Neuro-Oncology Unit, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
1314
|
Izmirly PM, Kim MY, Samanovic M, Fernandez‐Ruiz R, Ohana S, Deonaraine KK, Engel AJ, Masson M, Xie X, Cornelius AR, Herati RS, Haberman RH, Scher JU, Guttmann A, Blank RB, Plotz B, Haj‐Ali M, Banbury B, Stream S, Hasan G, Ho G, Rackoff P, Blazer AD, Tseng C, Belmont HM, Saxena A, Mulligan MJ, Clancy RM, Buyon JP. Evaluation of Immune Response and Disease Status in Systemic Lupus Erythematosus Patients Following SARS-CoV-2 Vaccination. Arthritis Rheumatol 2022; 74:284-294. [PMID: 34347939 PMCID: PMC8426963 DOI: 10.1002/art.41937] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate seroreactivity and disease flares after COVID-19 vaccination in a multiethnic/multiracial cohort of patients with systemic lupus erythematosus (SLE). METHODS Ninety SLE patients and 20 healthy controls receiving a complete COVID-19 vaccine regimen were included. IgG seroreactivity to the SARS-CoV-2 spike receptor-binding domain (RBD) and SARS-CoV-2 microneutralization were used to evaluate B cell responses; interferon-γ (IFNγ) production was measured by enzyme-linked immunospot (ELISpot) assay in order to assess T cell responses. Disease activity was measured by the hybrid SLE Disease Activity Index (SLEDAI), and flares were identified according to the Safety of Estrogens in Lupus Erythematosus National Assessment-SLEDAI flare index. RESULTS Overall, fully vaccinated SLE patients produced significantly lower IgG antibodies against SARS-CoV-2 spike RBD compared to fully vaccinated controls. Twenty-six SLE patients (28.8%) generated an IgG response below that of the lowest control (<100 units/ml). In logistic regression analyses, the use of any immunosuppressant or prednisone and a normal anti-double-stranded DNA antibody level prior to vaccination were associated with decreased vaccine responses. IgG seroreactivity to the SARS-CoV-2 spike RBD strongly correlated with the SARS-CoV-2 microneutralization titers and correlated with antigen-specific IFNγ production determined by ELISpot. In a subset of patients with poor antibody responses, IFNγ production was similarly diminished. Pre- and postvaccination SLEDAI scores were similar in both groups. Postvaccination flares occurred in 11.4% of patients; 1.3% of these were severe. CONCLUSION In a multiethnic/multiracial study of SLE patients, 29% had a low response to the COVID-19 vaccine which was associated with receiving immunosuppressive therapy. Reassuringly, severe disease flares were rare. While minimal protective levels remain unknown, these data suggest that protocol development is needed to assess the efficacy of booster vaccination.
Collapse
Affiliation(s)
| | - Mimi Y. Kim
- Albert Einstein College of MedicineNew YorkNew York
| | - Marie Samanovic
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Sharon Ohana
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Alexis J. Engel
- New York University Grossman School of MedicineNew YorkNew York
| | - Mala Masson
- New York University Grossman School of MedicineNew YorkNew York
| | - Xianhong Xie
- Albert Einstein College of MedicineNew YorkNew York
| | | | - Ramin S. Herati
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Jose U. Scher
- New York University Grossman School of MedicineNew YorkNew York
| | | | | | - Benjamin Plotz
- New York University Grossman School of MedicineNew YorkNew York
| | - Mayce Haj‐Ali
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Sara Stream
- New York University Grossman School of MedicineNew YorkNew York
| | - Ghadeer Hasan
- New York University Grossman School of MedicineNew YorkNew York
| | - Gary Ho
- New York University Grossman School of MedicineNew YorkNew York
| | - Paula Rackoff
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Chung‐E Tseng
- New York University Grossman School of MedicineNew YorkNew York
| | | | - Amit Saxena
- New York University Grossman School of MedicineNew YorkNew York
| | | | | | - Jill P. Buyon
- New York University Grossman School of MedicineNew YorkNew York
| |
Collapse
|
1315
|
Chivu‐Economescu M, Bleotu C, Grancea C, Chiriac D, Botezatu A, Iancu IV, Pitica I, Necula LG, Neagu A, Matei L, Dragu D, Sultana C, Radu EL, Nastasie A, Voicu O, Ataman M, Nedeianu S, Mambet C, Diaconu CC, Ruta SM. Kinetics and persistence of cellular and humoral immune responses to SARS-CoV-2 vaccine in healthcare workers with or without prior COVID-19. J Cell Mol Med 2022; 26:1293-1305. [PMID: 35043552 PMCID: PMC8831971 DOI: 10.1111/jcmm.17186] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 vaccines are highly efficient against severe forms of the disease, hospitalization and death. Nevertheless, insufficient protection against several circulating viral variants might suggest waning immunity and the need for an additional vaccine dose. We conducted a longitudinal study on the kinetics and persistence of immune responses in healthcare workers vaccinated with two doses of BNT162b2 mRNA vaccine with or without prior SARS-CoV-2 infection. No new infections were diagnosed during follow-up. At 6 months, post-vaccination or post-infection, despite a downward trend in the level of anti-S IgG antibodies, the neutralizing activity does not decrease significantly, remaining higher than 75% (85.14% for subjects with natural infection, 88.82% for vaccinated after prior infection and 78.37% for vaccinated only). In a live-virus neutralization assay, the highest neutralization titres were present at baseline and at 6 months follow-up in persons vaccinated after prior infection. Anti-S IgA levels showed a significant descending trend in vaccinated subjects (p < 0.05) after 14 weeks. Cellular immune responses are present even in vaccinated participants with declining antibody levels (index ratio 1.1-3) or low neutralizing activity (30%-40%) at 6 months, although with lower T-cell stimulation index (p = 0.046) and IFN-γ secretion (p = 0.0007) compared to those with preserved humoral responses.
Collapse
Affiliation(s)
| | | | | | | | - Anca Botezatu
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | | | - Ioana Pitica
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | | | - Ana Neagu
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | - Lilia Matei
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | - Denisa Dragu
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | - Camelia Sultana
- Stefan S. Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | - Elena L. Radu
- Stefan S. Nicolau Institute of VirologyBucharestRomania
- Institute for Water Quality and Resource Management TU WienViennaAustria
| | | | - Oana Voicu
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | - Marius Ataman
- Stefan S. Nicolau Institute of VirologyBucharestRomania
| | | | - Cristina Mambet
- Stefan S. Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | | | - Simona Maria Ruta
- Stefan S. Nicolau Institute of VirologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| |
Collapse
|
1316
|
Terpos E, Karalis V, Ntanasis-Stathopoulos I, Evangelakou Z, Gavriatopoulou M, Manola MS, Malandrakis P, Gianniou DD, Kastritis E, Trougakos IP, Dimopoulos MA. Comparison of Neutralizing Antibody Responses at 6 Months Post Vaccination with BNT162b2 and AZD1222. Biomedicines 2022; 10:338. [PMID: 35203547 PMCID: PMC8961789 DOI: 10.3390/biomedicines10020338] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Along with their level of protection against COVID-19, SARS-CoV-2-specific antibodies decline over time following vaccination with BNT162b2. However, relevant data on AZD1222 are scarce. In this context, the aim of this study was to compare SARS-CoV-2 neutralizing antibody (NAb) levels at one, three and six months after second vaccination with the BNT162b2 mRNA vaccine and the ChAdOx1 (AZD1222) viral vector vaccine (NCT04743388). The measurements were performed with the GenScript's cPassTM SARS-CoV-2 NAbs Detection Kit (GenScript, Inc.; Piscataway, NJ, USA). Overall, data from 282 individuals were included (BNT162b2 n = 83, AZD1222 n = 199). Both vaccines induced strong NAbs responses at 1 month following vaccination. Interestingly, NAb activity seemed superior with BNT162b2 compared with AZD1222. A gradual decline in NAbs titers was evident at 3 and 6 months post vaccination with both vaccines. However, the superiority of NAb response with BNT162b2 over AZD1222 remained consistent at all time points examined. Furthermore, the elimination rate of the NAb titer was higher throughout during the study period for those vaccinated with AZD1222 compared with BNT162b2. Age, gender, body mass index or comorbidities did not have a significant impact on NAbs levels over time. Our results may inform public health policies regarding the timing of booster COVID-19 vaccine shots.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| | - Vangelis Karalis
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece;
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| | - Zoi Evangelakou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (Z.E.); (M.S.M.); (D.D.G.); (I.P.T.)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| | - Maria S. Manola
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (Z.E.); (M.S.M.); (D.D.G.); (I.P.T.)
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| | - Despoina D. Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (Z.E.); (M.S.M.); (D.D.G.); (I.P.T.)
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (Z.E.); (M.S.M.); (D.D.G.); (I.P.T.)
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (M.G.); (P.M.); (E.K.); (M.A.D.)
| |
Collapse
|
1317
|
Cook C, Patel NJ, D'Silva KM, Hsu TYT, DiIorio M, Prisco L, Martin LW, Vanni K, Zaccardelli A, Todd D, Sparks JA, Wallace ZS. Clinical characteristics and outcomes of COVID-19 breakthrough infections among vaccinated patients with systemic autoimmune rheumatic diseases. Ann Rheum Dis 2022; 81:289-291. [PMID: 34489304 PMCID: PMC8791428 DOI: 10.1136/annrheumdis-2021-221326] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 11/03/2022]
Affiliation(s)
- Claire Cook
- Division of Rheumatology, Allergy, and Immunology and Clinical Epidemiology Program, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Naomi J Patel
- Division of Rheumatology, Allergy, and Immunology and Clinical Epidemiology Program, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Kristin M D'Silva
- Division of Rheumatology, Allergy, and Immunology and Clinical Epidemiology Program, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tiffany Y-T Hsu
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michael DiIorio
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lauren Prisco
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lily W Martin
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kathleen Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Alessandra Zaccardelli
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Derrick Todd
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jeffrey A Sparks
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Zachary Scott Wallace
- Division of Rheumatology, Allergy, and Immunology and Clinical Epidemiology Program, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
1318
|
Richterman A, Meyerowitz EA, Cevik M. Indirect Protection by Reducing Transmission: Ending the Pandemic With Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination. Open Forum Infect Dis 2022; 9:ofab259. [PMID: 35071679 PMCID: PMC8194790 DOI: 10.1093/ofid/ofab259] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/14/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Aaron Richterman
- Division of Infectious Diseases, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric A Meyerowitz
- Division of Infectious Diseases, Montefiore Medical Center, Bronx, New York, USA
| | - Muge Cevik
- Division of Infection and Global Health Research, School of Medicine, University of St Andrews, Fife, Scotland, United Kingdom
| |
Collapse
|
1319
|
Padmanabhan P, Desikan R, Dixit NM. Modeling how antibody responses may determine the efficacy of COVID-19 vaccines. NATURE COMPUTATIONAL SCIENCE 2022; 2:123-131. [PMID: 38177523 DOI: 10.1038/s43588-022-00198-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2024]
Abstract
Predicting the efficacy of COVID-19 vaccines would aid vaccine development and usage strategies, which is of importance given their limited supplies. Here we develop a multiscale mathematical model that proposes mechanistic links between COVID-19 vaccine efficacies and the neutralizing antibody (NAb) responses they elicit. We hypothesized that the collection of all NAbs would constitute a shape space and that responses of individuals are random samples from this space. We constructed the shape space by analyzing reported in vitro dose-response curves of ~80 NAbs. Sampling NAb subsets from the space, we recapitulated the responses of convalescent patients. We assumed that vaccination would elicit similar NAb responses. We developed a model of within-host SARS-CoV-2 dynamics, applied it to virtual patient populations and, invoking the NAb responses above, predicted vaccine efficacies. Our predictions quantitatively captured the efficacies from clinical trials. Our study thus suggests plausible mechanistic underpinnings of COVID-19 vaccines and generates testable hypotheses for establishing them.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Rajat Desikan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
- Certara QSP, Certara UK Limited, Sheffield, UK
| | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India.
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
1320
|
Huang J, Xia L, Lin J, Liu B, Zhao Y, Xin C, Ai X, Cao W, Zhang X, Tian L, Wu Q. No Effect of Inactivated SARS-CoV-2 Vaccination on in vitro Fertilization Outcomes: A Propensity Score-Matched Study. J Inflamm Res 2022; 15:839-849. [PMID: 35177919 PMCID: PMC8843422 DOI: 10.2147/jir.s347729] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/15/2022] [Indexed: 12/17/2022] Open
Abstract
Purpose Patients and Methods Results Conclusion
Collapse
Affiliation(s)
- Jialyu Huang
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| | - Leizhen Xia
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| | - Jiaying Lin
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai, People’s Republic of China
| | - Bangdong Liu
- Department of Laboratory Medicine, Huanan Hospital of Shenzhen University, Shenzhen, People’s Republic of China
| | - Yan Zhao
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| | - Cailin Xin
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| | - Xiaoyan Ai
- Department of Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| | - Wenting Cao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Xiaocui Zhang
- Center for Reproductive Medicine, Shangrao Maternal and Child Health Hospital, Shangrao, People’s Republic of China
| | - Lifeng Tian
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
- Correspondence: Lifeng Tian; Qiongfang Wu, Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China, Email ;
| | - Qiongfang Wu
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, People’s Republic of China
| |
Collapse
|
1321
|
Bruxvoort KJ, Sy LS, Qian L, Ackerson BK, Luo Y, Lee GS, Tian Y, Florea A, Takhar HS, Tubert JE, Talarico CA, Tseng HF. Real-world effectiveness of the mRNA-1273 vaccine against COVID-19: Interim results from a prospective observational cohort study. LANCET REGIONAL HEALTH. AMERICAS 2022; 6:100134. [PMID: 34849505 PMCID: PMC8614600 DOI: 10.1016/j.lana.2021.100134] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Phase 3 trials found mRNA-1273 was highly effective in preventing COVID-19. We conducted a prospective cohort study at Kaiser Permanente Southern California (KPSC) to determine the real-world vaccine effectiveness (VE) of mRNA-1273 in preventing COVID-19 infection and severe disease. Methods For this planned interim analysis, individuals aged ≥18 years receiving 2 doses of mRNA-1273 ≥24 days apart (18/12/2020-31/03/2021) were 1:1 matched to randomly selected unvaccinated individuals by age, sex, and race/ethnicity, with follow-up through 30/06/2021. Outcomes were COVID-19 infection (SARS-CoV-2 positive molecular test or COVID-19 diagnosis code) or severe disease (COVID-19 hospitalization and COVID-19 hospital death). Adjusted hazard ratios (aHR) and confidence intervals (CI) for COVID-19 outcomes comparing vaccinated and unvaccinated individuals were estimated by Cox proportional hazards models accounting for multiple comparisons. Adjusted VE was calculated as (1-aHR)x100. Whole genome sequencing was performed on SARS-CoV-2 positive specimens from the KPSC population. Findings This analysis included 352,878 recipients of 2 doses of mRNA-1273 matched to 352,878 unvaccinated individuals. VE (99·3% CI) against COVID-19 infection was 87·4% (84·8–89·6%). VE against COVID-19 hospitalization and hospital death was 95·8% (90·7–98·1%) and 97·9% (66·9-99·9%), respectively. VE was higher against symptomatic (88·3% [98·3% CI: 86·1–90·2%]) than asymptomatic COVID-19 (72·7% [53·4–84·0%]), but was generally similar across age, sex, and racial/ethnic subgroups. VE among individuals with history of COVID-19 ranged from 8·2–33·6%. The most prevalent variants were Alpha (41·6%), Epsilon (17·5%), Delta (11·5%), and Gamma (9·1%), with Delta increasing to 54·0% of variants by June 2021. Interpretation These interim results provide reassuring evidence of the VE of 2 doses of mRNA-1273 across age, sex, and racial/ethnic subgroups, and against asymptomatic and symptomatic COVID-19, and severe COVID-19 outcomes. Among individuals with history of COVID-19, mRNA-1273 vaccination may offer added protection beyond immunity acquired from prior infection. Longer follow-up is needed to fully evaluate VE of mRNA-1273 against emerging SARS-CoV-2 variants. Funding Moderna Inc.
Collapse
Affiliation(s)
- Katia J Bruxvoort
- Kaiser Permanente Southern California, Pasadena, CA, USA.,University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lina S Sy
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Lei Qian
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Yi Luo
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Gina S Lee
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Yun Tian
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Ana Florea
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Julia E Tubert
- Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | - Hung Fu Tseng
- Kaiser Permanente Southern California, Pasadena, CA, USA.,Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| |
Collapse
|
1322
|
En quoi la Covid-19 concerne la prise en charge des rhumatismes inflammatoires ? Place de la vaccination selon les cas. REVUE DU RHUMATISME MONOGRAPHIES 2022. [PMCID: PMC8642824 DOI: 10.1016/j.monrhu.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
La pandémie de Covid-19 a clairement impacté la prise en charge des rhumatismes inflammatoires chroniques (RIC) en termes de rupture de suivi et de rupture de traitement en raison du risque d’infection sévère liée à la maladie et aux traitements immuno-modulateurs. Des traitements permettent maintenant de prévenir ces formes sévères incluant des vaccins et des anticorps monoclonaux spécifiques dirigés contre la protéine « Spike » du virus SARS-CoV2. Parmi les traitements utilisés dans les RIC, les corticoïdes et le rituximab apparaissent clairement comme associés à une forme sévère de la Covid. La réponse humorale vaccinale après vaccination avec des vaccins à ARN messager (ARNm) apparaît diminuée sous méthotrexate, abatacept et surtout rituximab. Des schémas vaccinaux spécifiques ont déjà été proposés pour les patients sous rituximab et devraient s’affiner au fur et à mesure des connaissances car ces vaccins devraient s’inscrire maintenant dans le calendrier vaccinal de nos patients. Pour ceux qui ne développent pas d’anticorps après un schéma vaccinal complet, l’association d’anticorps casirivimab et imdévimab peut être administrée mensuellement en préventif ou en post-exposition.
Collapse
|
1323
|
Gralnek IM, Hassan C, Ebigbo A, Fuchs A, Beilenhoff U, Antonelli G, Bisschops R, Arvanitakis M, Bhandari P, Bretthauer M, Kaminski MF, Lorenzo-Zuniga V, Rodriguez de Santiago E, Siersema PD, Tham TC, Triantafyllou K, Tringali A, Voiosu A, Webster G, de Pater M, Fehrke B, Gazic M, Gjergek T, Maasen S, Waagenes W, Dinis-Ribeiro M, Messmann H. ESGE and ESGENA Position Statement on gastrointestinal endoscopy and COVID-19: Updated guidance for the era of vaccines and viral variants. Endoscopy 2022; 54:211-216. [PMID: 34933373 DOI: 10.1055/a-1700-4897] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ian M Gralnek
- Ellen and Pinchas Mamber Institute of Gastroenterology and Hepatology, Emek Medical Center, Afula, Israel and Rappaport Faculty of Medicine Technion Israel Institute of Technology, Haifa, Israel
| | - Cesare Hassan
- Endoscopy Unit, IRCCS Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Alanna Ebigbo
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| | - Andre Fuchs
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| | | | - Giulio Antonelli
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Rome, Italy
- Gastroenterology and Digestive Endoscopy Unit, Ospedale dei Castelli di Ariccia, Rome, Italy
| | - Raf Bisschops
- Department of Gastroenterology and Hepatology, Catholic University of Leuven (KUL), TARGID, University Hospitals Leuven, Leuven, Belgium
| | | | - Pradeep Bhandari
- Gastroenterology, Portsmouth Hospital NHS Trust, Portsmouth, United Kingdom
| | - Michael Bretthauer
- Clinical Effectiveness Research Group, University of Oslo, and Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Michal F Kaminski
- Department of Cancer Prevention and Department of Oncological Gastroenterology, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Vicente Lorenzo-Zuniga
- Department of Gastroenterology, University and Polytechnic La Fe Hospital/IIS La Fe, Valencia. Spain
| | - Enrique Rodriguez de Santiago
- Department of Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Peter D Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tony C Tham
- Division of Gastroenterology, Ulster Hospital, Dundonald, Belfast, Northern Ireland
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Internal Medicine - Propaedeutic, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Alberto Tringali
- Digestive Endoscopy Unit, Fondazione IRCCS-Istituto Nazionale Tumori, Milan, Italy
| | - Andrei Voiosu
- Department of Gastroenterology and Hepatology, Colentina Clinical Hospital, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - George Webster
- Department of Gastroenterology, University College London Hospitals, London, United Kingdom
| | | | - Björn Fehrke
- Department of Pneumonology, Inselspital, University Hospital, Bern, Switzerland
| | - Mario Gazic
- General Hospital Bjelovar, Bjelovar, Croatia
| | | | | | | | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center (Porto.CCC) and RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Helmut Messmann
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| |
Collapse
|
1324
|
Elkrief A, Wu JT, Jani C, Enriquez KT, Glover M, Shah MR, Shaikh HG, Beeghly-Fadiel A, French B, Jhawar SR, Johnson DB, McKay RR, Rivera DR, Reuben DY, Shah S, Tinianov SL, Vinh DC, Mishra S, Warner JL. Learning through a Pandemic: The Current State of Knowledge on COVID-19 and Cancer. Cancer Discov 2022; 12:303-330. [PMID: 34893494 PMCID: PMC8831477 DOI: 10.1158/2159-8290.cd-21-1368] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/26/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has left patients with current or past history of cancer facing disparate consequences at every stage of the cancer trajectory. This comprehensive review offers a landscape analysis of the current state of the literature on COVID-19 and cancer, including the immune response to COVID-19, risk factors for severe disease, and impact of anticancer therapies. We also review the latest data on treatment of COVID-19 and vaccination safety and efficacy in patients with cancer, as well as the impact of the pandemic on cancer care, including the urgent need for rapid evidence generation and real-world study designs. SIGNIFICANCE: Patients with cancer have faced severe consequences at every stage of the cancer journey due to the COVID-19 pandemic. This comprehensive review offers a landscape analysis of the current state of the field regarding COVID-19 and cancer. We cover the immune response, risk factors for severe disease, and implications for vaccination in patients with cancer, as well as the impact of the COVID-19 pandemic on cancer care delivery. Overall, this review provides an in-depth summary of the key issues facing patients with cancer during this unprecedented health crisis.
Collapse
Affiliation(s)
- Arielle Elkrief
- Division of Medical Oncology (Department of Medicine), McGill University Health Centre, Montreal, Quebec, Canada
| | - Julie T Wu
- Stanford University, Palo Alto, California
| | - Chinmay Jani
- Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts
| | - Kyle T Enriquez
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Mansi R Shah
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | | | | | | - Sachin R Jhawar
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Rana R McKay
- University of California San Diego, San Diego, California
| | - Donna R Rivera
- Division of Cancer Control and Population Services, National Cancer Institute, Rockville, Maryland
| | - Daniel Y Reuben
- Medical University of South Carolina, Charleston, South Carolina
| | - Surbhi Shah
- Hematology and Oncology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Stacey L Tinianov
- Advocates for Collaborative Education, UCSF Breast Science Advocacy Core, San Francisco, California
| | - Donald Cuong Vinh
- Division of Infectious Diseases (Department of Medicine), Divisions of Medical Microbiology and of Molecular Diagnostics (OptiLab), McGill University Health Centre, Montreal, Quebec, Canada
| | - Sanjay Mishra
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeremy L Warner
- Vanderbilt University Medical Center, Nashville, Tennessee.
- Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
1325
|
Albalawi OM, Alomran MI, Alsagri GM, Althunian TA, Alshammari TM. Analyzing the U.S. Post-marketing safety surveillance of COVID-19 vaccines. Saudi Pharm J 2022; 30:180-184. [PMID: 35002372 PMCID: PMC8719360 DOI: 10.1016/j.jsps.2021.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/27/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Since December 2020, three COVID-19 vaccines have been authorized in the United States (U.S.) and were proceeded by large immunization programs. The aim of this study was to characterize the U.S. post-marketing safety (PMS) profiles of these vaccines with an in-depth analysis of mortality data. METHODS This was a retrospective database analysis study. Details of the U.S. PMS reports (15 December 2020 to 19 March 2021) of the three vaccines (Pfizer-BioNTech, Moderna, and Janssen Ad26.COV2.S) were retrieved from the U.S. Vaccine Adverse Event Reporting System (VAERS). A descriptive analysis was conducted to characterize the reported adverse events (AEs). A comparative (Pfizer-BioNTech vs. Moderna) analysis of mortality was conducted. The mean count ratio of death between the two vaccines was estimated using a negative binomial regression model adjusting for the measured confounders. RESULTS A total of 44,451 AE reports were retrieved (corresponding to 0.05% of the U.S. population who received at least one dose). The most commonly reported AEs were injection site reactions (30.4% of the reports), pain (reported in 26.7% of the reports), and headache (18.6% of the reports). Serious AEs were reported in only 14.6% of the reports with 4,108 hospitalizations. The total number of deaths was 1,919 with a mean count ratio of Moderna (n = 997) vs. Pfizer-BioNTech (n = 899) of 1.07 (95% confidence interval 0.86 to 1.33). CONCLUSIONS The vast majority of PMS AEs in the U.S. were non-serious, and the number of serious AEs is very low given the total number of vaccinated U.S. population.
Collapse
Affiliation(s)
- Omar M Albalawi
- Research and Studies, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Maha I Alomran
- Research and Studies, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Ghada M Alsagri
- Research and Studies, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Turki A Althunian
- Research and Studies, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Thamir M Alshammari
- Research and Studies, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
- College of Pharmacy, Riyadh Elm University, Riyadh, Saudi Arabia
- Medication Safety Research Chair, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
1326
|
Striking antibody evasion manifested by the Omicron variant of SARS-CoV-2. Nature 2022; 602:676-681. [PMID: 35016198 DOI: 10.1038/s41586-021-04388-0] [Citation(s) in RCA: 918] [Impact Index Per Article: 306.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 01/12/2023]
Abstract
The B.1.1.529/Omicron variant of SARS-CoV-2 was only recently detected in southern Africa, but its subsequent spread has been extensive, both regionally and globally1. It is expected to become dominant in the coming weeks2, probably due to enhanced transmissibility. A striking feature of this variant is the large number of spike mutations3 that pose a threat to the efficacy of current COVID-19 vaccines and antibody therapies4. This concern is amplified by the findings of our study. Here we found that B.1.1.529 is markedly resistant to neutralization by serum not only from patients who recovered from COVID-19, but also from individuals who were vaccinated with one of the four widely used COVID-19 vaccines. Even serum from individuals who were vaccinated and received a booster dose of mRNA-based vaccines exhibited substantially diminished neutralizing activity against B.1.1.529. By evaluating a panel of monoclonal antibodies against all known epitope clusters on the spike protein, we noted that the activity of 17 out of the 19 antibodies tested were either abolished or impaired, including ones that are currently authorized or approved for use in patients. Moreover, we also identified four new spike mutations (S371L, N440K, G446S and Q493R) that confer greater antibody resistance on B.1.1.529. The Omicron variant presents a serious threat to many existing COVID-19 vaccines and therapies, compelling the development of new interventions that anticipate the evolutionary trajectory of SARS-CoV-2.
Collapse
|
1327
|
Ravindra Naik B, Anil Kumar S, Rachegowda N, Yashas Ullas L, Revanth RB, Venkata Sai Aluru NR. Severity of COVID-19 Infection Using Chest Computed Tomography Severity Score Index Among Vaccinated and Unvaccinated COVID-19-Positive Healthcare Workers: An Analytical Cross-Sectional Study. Cureus 2022; 14:e22087. [PMID: 35295366 PMCID: PMC8917791 DOI: 10.7759/cureus.22087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Introduction: Coronavirus disease 2019 (COVID-19) vaccines protect against severe illness. However, data on post-vaccination COVID-19 breakthrough infections are limited. Methods: An analytical cross-sectional study was conducted from May 2021 to July 2021 among 2043 COVID-19-positive healthcare workers who were divided into a vaccinated group (n=1010) and an unvaccinated group (n=1033). A pre-tested questionnaire was circulated among the healthcare workers using Google Forms. Chest computed tomography (CT) severity score was the primary outcome variable analyzed using coGuide. Results: The average age of the study population was less than 45 years in both groups (43.05 ± 13.02 years). Most respondents (62%) were males. Hypertension (39%) and diabetes (33%) were the most common underlying diseases. Significant differences in age and cardiac disease were observed between the two groups (p = 0.07 and p <0.001, respectively). However, the difference was insignificant (p >0.05) for gender, hypertension, and diabetes. Most unvaccinated respondents had an increased CT severity score, and the difference between the studies groups was significant (p <0.001). Of the 1,010 vaccinated individuals, 382 (37.82%) received the only first vaccination dose, and 628 (62.18%) received both doses. The CT severity score decreased after receiving both vaccination doses, and the difference between CT severity score and vaccination status was significant (p <0.001). Conclusion: COVID-19 was mild in the vaccinated group. Chest CT severity score index can be considered an efficient tool in predicting prognosis and monitoring disease in patients with COVID-19 in India.
Collapse
Affiliation(s)
| | | | - N Rachegowda
- Department of Radio-Diagnosis, Sri Devaraj Urs Medical College, Kolar, IND
| | - L Yashas Ullas
- Department of Radio-Diagnosis, Sri Devaraj Urs Medical College, Kolar, IND
| | - R B Revanth
- Department of Radio-Diagnosis, Sri Devaraj Urs Medical College, Kolar, IND
| | | |
Collapse
|
1328
|
Bell TD. COVID-19 in the Critically Ill Patient. Infect Dis Clin North Am 2022; 36:365-377. [PMID: 35636905 PMCID: PMC8847096 DOI: 10.1016/j.idc.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
1329
|
Pelletier AN, Sekaly RP, Tomalka JA. Translating known drivers of COVID-19 disease severity to design better SARS-CoV-2 vaccines. Curr Opin Virol 2022; 52:89-101. [PMID: 34902803 PMCID: PMC8664555 DOI: 10.1016/j.coviro.2021.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/05/2021] [Accepted: 11/19/2021] [Indexed: 01/17/2023]
Abstract
The SARS-CoV-2 pandemic has highlighted how an emergent disease can spread globally and how vaccines are once again the most important public health policy to combat infectious disease. Despite promising initial protection, the rise of new viral variants calls into question how effective current SARS-CoV-2 vaccines will be moving forward. Improving on vaccine platforms represents an opportunity to stay ahead of SARS-CoV-2 and keep the human population protected. Many researchers focus on modifying delivery platforms or altering the antigen(s) presented to improve the efficacy of the vaccines. Identifying mechanisms of natural immunity that result in the control of infection and prevent poor clinical outcomes provides an alternative approach to the development of efficacious vaccines. Early and current evidence shows that SARS-CoV-2 infection is marked by potent lung inflammation and relatively diminished antiviral signaling which leads to impaired immune recognition and viral clearance, essentially making SARS-CoV-2 'too hot to handle'.
Collapse
Affiliation(s)
| | - Rafick P Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey A Tomalka
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
1330
|
Gralnek IM, Hassan C, Ebigbo A, Fuchs A, Beilenhoff U, Antonelli G, Bisschops R, Arvanitakis M, Bhandari P, Bretthauer M, Kaminski MF, Lorenzo-Zuniga V, Rodriguez de Santiago E, Siersema PD, Tham TC, Triantafyllou K, Tringali A, Voiosu A, Webster G, de Pater M, Fehrke B, Gazic M, Gjergek T, Maasen S, Waagenes W, Dinis-Ribeiro M, Messmann H. ESGE and ESGENA Position Statement on gastrointestinal endoscopy and COVID-19: Updated guidance for the era of vaccines and viral variants. Endoscopy 2022; 54:211-216. [DOI: 10.1055/a-1700-4897 pmid: 34933373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Affiliation(s)
- Ian M. Gralnek
- Ellen and Pinchas Mamber Institute of Gastroenterology and Hepatology, Emek Medical Center, Afula, Israel and Rappaport Faculty of Medicine Technion Israel Institute of Technology, Haifa, Israel
| | - Cesare Hassan
- Endoscopy Unit, IRCCS Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Alanna Ebigbo
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| | - Andre Fuchs
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| | | | - Giulio Antonelli
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Rome, Italy
- Gastroenterology and Digestive Endoscopy Unit, Ospedale dei Castelli di Ariccia, Rome, Italy
| | - Raf Bisschops
- Department of Gastroenterology and Hepatology, Catholic University of Leuven (KUL), TARGID, University Hospitals Leuven, Leuven, Belgium
| | | | - Pradeep Bhandari
- Gastroenterology, Portsmouth Hospital NHS Trust, Portsmouth, United Kingdom
| | - Michael Bretthauer
- Clinical Effectiveness Research Group, University of Oslo, and Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Michal F. Kaminski
- Department of Cancer Prevention and Department of Oncological Gastroenterology, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Vicente Lorenzo-Zuniga
- Department of Gastroenterology, University and Polytechnic La Fe Hospital/IIS La Fe, Valencia. Spain
| | - Enrique Rodriguez de Santiago
- Department of Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tony C. Tham
- Division of Gastroenterology, Ulster Hospital, Dundonald, Belfast, Northern Ireland
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Internal Medicine - Propaedeutic, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Alberto Tringali
- Digestive Endoscopy Unit, Fondazione IRCCS-Istituto Nazionale Tumori, Milan, Italy
| | - Andrei Voiosu
- Department of Gastroenterology and Hepatology, Colentina Clinical Hospital, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - George Webster
- Department of Gastroenterology, University College London Hospitals, London, United Kingdom
| | | | - Björn Fehrke
- Department of Pneumonology, Inselspital, University Hospital, Bern, Switzerland
| | - Mario Gazic
- General Hospital Bjelovar, Bjelovar, Croatia
| | | | | | | | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center (Porto.CCC) and RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Helmut Messmann
- III Medizinische Klinik Universitätsklinikum Augsburg, Augsburg, Germany
| |
Collapse
|
1331
|
de Oliveira Resende R, José de Oliveira V, Sousa Correa A, Trica de Araújo P, Akio Taketomi E. COVID-19 scenario and recommendations on the continuity of immunobiological therapy in patients with atopic asthma in Brazil and Latin America. Expert Rev Respir Med 2022; 16:211-220. [PMID: 35034544 DOI: 10.1080/17476348.2022.2027758] [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: 06/29/2021] [Accepted: 01/07/2022] [Indexed: 01/08/2023]
Abstract
INTRODUCTION With the abrupt establishment of the COVID-19 pandemic, treatment for immunological diseases may be influenced by the SARS-CoV-2 infection. Immunobiologics play a pivotal role in the management of severe symptoms of allergy, and an opinion regarding the continuity of this treatment during the COVID-19 pandemic must be issued. AREAS COVERED In Brazil and other countries, patients with severe asthma have been included in the priority groups for COVID-19 vaccination, even those who are undergoing immunobiological therapy. Data are insufficient to support the influence of this therapy on severe COVID-19. Therapeutic strategies for asthma and guidelines/statements of the main societies of Allergy in Latin America on the continuity of treatment with immunobiologics during the COVID-19 pandemic were obtained from the institutional websites and papers published up to September 2021. EXPERT OPINION Although the association between asthma and COVID-19 has been under investigation, immunobiological treatment should follow the consensus-based statements recommending the maintenance of the therapy unless the patient is infected by the SARS-CoV-2. However, it must be closely followed by the medical assistant.
Collapse
Affiliation(s)
- Rafael de Oliveira Resende
- Laboratory of Allergy and Clinical Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Brazil
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Vinícius José de Oliveira
- Laboratory of Allergy and Clinical Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Brazil
| | - Alessandro Sousa Correa
- Laboratory of Allergy and Clinical Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Brazil
| | - Pedro Trica de Araújo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Ernesto Akio Taketomi
- Laboratory of Allergy and Clinical Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Brazil
| |
Collapse
|
1332
|
Sakurai F, Tachibana M, Mizuguchi H. Adenovirus vector-based vaccine for infectious diseases. Drug Metab Pharmacokinet 2022; 42:100432. [PMID: 34974335 PMCID: PMC8585960 DOI: 10.1016/j.dmpk.2021.100432] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 01/10/2023]
Abstract
Replication-incompetent adenovirus (Ad) vectors have been widely used as gene delivery vehicles in both gene therapy studies and basic studies for gene function analysis due to their highly advantageous properties, which include high transduction efficiencies, relatively large capacities for transgenes, and high titer production. In addition, Ad vectors induce moderate levels of innate immunity and have relatively high thermostability, making them very attractive as potential vaccine vectors. Accordingly, it is anticipated that Ad vectors will be used in vaccines for the prevention of infectious diseases, including Ebola virus disease and acquired immune deficiency syndrome (AIDS). Much attention is currently focused on the potential use of an Ad vector vaccine for coronavirus disease 2019 (COVID-19). In this review, we describe the basic properties of an Ad vector, Ad vector-induced innate immunity and immune responses to Ad vector-produced transgene products. Development of novel Ad vectors which can overcome the drawbacks of conventional Ad vector vaccines and clinical application of Ad vector vaccines to several infectious diseases are also discussed.
Collapse
Affiliation(s)
- Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| |
Collapse
|
1333
|
Wang L, Kaelber DC, Xu R, Berger NA. COVID-19 breakthrough infections, hospitalizations and mortality in fully vaccinated patients with hematologic malignancies: A clarion call for maintaining mitigation and ramping-up research. Blood Rev 2022; 54:100931. [PMID: 35120771 PMCID: PMC8802493 DOI: 10.1016/j.blre.2022.100931] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/22/2023]
Abstract
There has been limited data presented to characterize and quantify breakthrough SARS-CoV-2 infections, hospitalizations, and mortality in vaccinated patients with hematologic malignancies (HM). We performed a retrospective cohort study of patient electronic health records of 514,413 fully vaccinated patients from 63 healthcare organizations in the US, including 5956 with HM and 508,457 without malignancies during the period from December 2020 to October 2021. The breakthrough SARS-CoV-2 infections in patients with HM steadily increased and reached 67.7 cases per 1000 persons in October 2021. The cumulative risk of breakthrough infections during the period in patients with HM was 13.4%, ranging from 11.0% for acute lymphocytic leukemia to 17.2% and 17.4% for multiple myeloma and chronic myeloid leukemia respectively, all higher than the risk of 4.5% in patients without malignancies (p < 0.001). No significant racial disparities in breakthrough infections were observed. The overall hospitalization risk was 37.8% for patients with HM who had breakthrough infections, significantly higher than 2.2% for those who had no breakthrough infections (hazard ratio or HR: 34.49, 95% CI: 25.93-45.87). The overall mortality risk was 5.7% for patients with HM who had breakthrough infections, significantly higher than the 0.8% for those who had no breakthrough infections (HR: 10.25, 95% CI: 5.94-17.69). In summary, this study shows that among the fully vaccinated population, patients with HM had significantly higher risk for breakthrough infections compared to patients without cancer and that breakthrough infections in patients with HM were associated with significant clinical outcomes including hospitalizations and mortality.
Collapse
Affiliation(s)
- Lindsey Wang
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Center for Science, Health, and Society, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David C Kaelber
- The Center for Clinical Informatics Research and Education, The MetroHealth System, the Departments of Internal Medicine, Pediatrics, and Population and Quantitative Health Science, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Nathan A Berger
- Center for Science, Health, and Society, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
1334
|
da Silva SJR, de Lima SC, da Silva RC, Kohl A, Pena L. Viral Load in COVID-19 Patients: Implications for Prognosis and Vaccine Efficacy in the Context of Emerging SARS-CoV-2 Variants. Front Med (Lausanne) 2022; 8:836826. [PMID: 35174189 PMCID: PMC8841511 DOI: 10.3389/fmed.2021.836826] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022] Open
Abstract
The worldwide spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused an unprecedented public health crisis in the 21st century. As the pandemic evolves, the emergence of SARS-CoV-2 has been characterized by the emergence of new variants of concern (VOCs), which resulted in a catastrophic impact on SARS-CoV-2 infection. In light of this, research groups around the world are unraveling key aspects of the associated illness, coronavirus disease 2019 (COVID-19). A cumulative body of data has indicated that the SARS-CoV-2 viral load may be a determinant of the COVID-19 severity. Here we summarize the main characteristics of the emerging variants of SARS-CoV-2, discussing their impact on viral transmissibility, viral load, disease severity, vaccine breakthrough, and lethality among COVID-19 patients. We also provide a rundown of the rapidly expanding scientific evidence from clinical studies and animal models that indicate how viral load could be linked to COVID-19 prognosis and vaccine efficacy among vaccinated individuals, highlighting the differences compared to unvaccinated individuals.
Collapse
Affiliation(s)
- Severino Jefferson Ribeiro da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Suelen Cristina de Lima
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Ronaldo Celerino da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Lindomar Pena
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| |
Collapse
|
1335
|
Fazio ND, Delogu G, Bertozzi G, Fineschi V, Frati P. SARS-CoV2 Vaccination Adverse Events Trend in Italy: A Retrospective Interpretation of the Last Year (December 2020-September 2021). Vaccines (Basel) 2022; 10:216. [PMID: 35214674 PMCID: PMC8880467 DOI: 10.3390/vaccines10020216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
At the end of 2020, a vaccination campaign against COVID-19 was launched. In 2021, legal obligations for health workers, as well as specific regulations for all workers, were introduced. The global SARS-CoV-2 pandemic was followed by epochal changes in life, school, and work habits in Italy. Therefore, the pharmacovigilance work currently being conducted in Italy by the AIFA concerning the recording and analysing of adverse reactions related to the use of vaccines has proved to be very important. The latest report, including a period of 10 months from December 2020 to September 2021, has allowed us to combine the results received so far, and to compare the safety of all vaccines currently available in Italy. The results of this analysis are highly encouraging and reveal the statistical reliability of the safety of the COVID-19 vaccines currently used in Italy. The dissemination of these findings could increase the public's awareness of vaccines and their ability to make free and informed choices concerning vaccination. The potential increase in the Italian population's adherence to the vaccination campaign could ultimately be a decisive factor in achieving herd immunity and the final resolution of the pandemic.
Collapse
Affiliation(s)
- Nicola Di Fazio
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (G.D.); (P.F.)
| | - Giuseppe Delogu
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (G.D.); (P.F.)
| | - Giuseppe Bertozzi
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy;
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (G.D.); (P.F.)
- IRCSS Neuromed Mediterranean Neurological Institute, Via Atinense 18, 86077 Pozzilli, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (G.D.); (P.F.)
- IRCSS Neuromed Mediterranean Neurological Institute, Via Atinense 18, 86077 Pozzilli, Italy
| |
Collapse
|
1336
|
Oh W, Jayaraman P, Sawant AS, Chan L, Levin MA, Charney AW, Kovatch P, Glicksberg BS, Nadkarni GN. Using sequence clustering to identify clinically relevant subphenotypes in patients with COVID-19 admitted to the intensive care unit. J Am Med Inform Assoc 2022; 29:489-499. [PMID: 35092685 PMCID: PMC8800515 DOI: 10.1093/jamia/ocab252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The novel coronavirus disease 2019 (COVID-19) has heterogenous clinical courses, indicating that there might be distinct subphenotypes in critically ill patients. Although prior research has identified these subphenotypes, the temporal pattern of multiple clinical features has not been considered in cluster models. We aimed to identify temporal subphenotypes in critically ill patients with COVID-19 using a novel sequence cluster analysis and associate them with clinically relevant outcomes. MATERIALS AND METHODS We analyzed 1036 confirmed critically ill patients with laboratory-confirmed SARS-COV-2 infection admitted to the Mount Sinai Health System in New York city. The agglomerative hierarchical clustering method was used with Levenshtein distance and Ward's minimum variance linkage. RESULTS We identified four subphenotypes. Subphenotype I (N = 233 [22.5%]) included patients with rapid respirations and a rapid heartbeat but less need for invasive interventions within the first 24 hours, along with a relatively good prognosis. Subphenotype II (N = 418 [40.3%]) represented patients with the least degree of ailments, relatively low mortality, and the highest probability of discharge from the hospital. Subphenotype III (N = 259 [25.0%]) represented patients who experienced clinical deterioration during the first 24 hours of intensive care unit admission, leading to poor outcomes. Subphenotype IV (N = 126 [12.2%]) represented an acute respiratory distress syndrome trajectory with an almost universal need for mechanical ventilation. CONCLUSION We utilized the sequence cluster analysis to identify clinical subphenotypes in critically ill COVID-19 patients who had distinct temporal patterns and different clinical outcomes. This study points toward the utility of including temporal information in subphenotyping approaches.
Collapse
Affiliation(s)
- Wonsuk Oh
- Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Data Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pushkala Jayaraman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Data Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashwin S Sawant
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lili Chan
- Division of Data Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Matthew A Levin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patricia Kovatch
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pharmacological Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin S Glicksberg
- Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Girish N Nadkarni
- Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Data Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
1337
|
Watkins LKF, Mitruka K, Dorough L, Bressler SS, Kugeler KJ, Sadigh KS, Birhane MG, Nolen LD, Fischer M. Characteristics of Reported Deaths Among Fully Vaccinated Persons with COVID-19 -United States, January-April 2021. Clin Infect Dis 2022; 75:e645-e652. [PMID: 35092677 PMCID: PMC8807315 DOI: 10.1093/cid/ciac066] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Background COVID-19 vaccines are highly efficacious, but SARS-CoV-2 infections post-vaccination occur. We characterized COVID-19 cases among fully vaccinated persons with an outcome of death. Methods We analyzed COVID-19 cases voluntarily reported to CDC by US health departments during January 1, 2021–April 30, 2021. We included cases among U.S. residents with a positive SARS-CoV-2 test ≥14 days after completion of an authorized primary vaccine series and who had a known outcome (alive or death) as of May 31, 2021. When available, specimens were sequenced for viral lineage and death certificates were reviewed for cause(s) of death. Results Of 8,084 reported COVID-19 cases among fully vaccinated persons during the surveillance period, 245 (3.0%) died. Compared with patients who remained alive, those who died were older (median age 82 years vs. 57 years, P <0.01), more likely to reside in a long-term care facility (51% vs. 18%, P <0.01), and more likely to have at least one underlying health condition associated with risk for severe disease (64% vs. 24%, P <0.01). Among 245 deaths, 191 (78%) were classified as COVID-19-related. Of 106 deaths with available death certificates, COVID-19 was listed on 81 (77%). There were no differences in the type of vaccine administered or the most common viral lineage (B.1.1.7). Conclusions COVID-19 deaths are rare in fully vaccinated persons, occurring most commonly in those with risk factors for severe disease, including older age and underlying health conditions. All eligible persons should be fully vaccinated against COVID-19 and follow other prevention measures to mitigate exposure risk.
Collapse
Affiliation(s)
| | - Kiren Mitruka
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| | - Layne Dorough
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA; ORISE fellow, Oak Ridge Institute for Science and Education, Oak Ridge, TN
| | - Sara S Bressler
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| | - Kiersten J Kugeler
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| | - Katrin S Sadigh
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA; Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, GA.,COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA; ORISE fellow, Oak Ridge Institute for Science and Education, Oak Ridge, TN
| | - Meseret G Birhane
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| | - Leisha D Nolen
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| | - Marc Fischer
- COVID-19 Emergency Response, Centers for Disease Control and Prevention, Atlanta, GA
| |
Collapse
|
1338
|
Bravo L, Smolenov I, Han HH, Li P, Hosain R, Rockhold F, Clemens SAC, Roa C, Borja-Tabora C, Quinsaat A, Lopez P, López-Medina E, Brochado L, Hernández EA, Reynales H, Medina T, Velasquez H, Toloza LB, Rodriguez EJ, de Salazar DIM, Rodríguez CA, Sprinz E, Cerbino-Neto J, Luz KG, Schwarzbold AV, Paiva MS, Carlos J, Montellano MEB, de Los Reyes MRA, Yu CY, Alberto ER, Panaligan MM, Salvani-Bautista M, Buntinx E, Hites M, Martinot JB, Bhorat QE, Badat A, Baccarini C, Hu B, Jurgens J, Engelbrecht J, Ambrosino D, Richmond P, Siber G, Liang J, Clemens R. Efficacy of the adjuvanted subunit protein COVID-19 vaccine, SCB-2019: a phase 2 and 3 multicentre, double-blind, randomised, placebo-controlled trial. Lancet 2022; 399:461-472. [PMID: 35065705 PMCID: PMC8776284 DOI: 10.1016/s0140-6736(22)00055-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/05/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND A range of safe and effective vaccines against SARS CoV 2 are needed to address the COVID 19 pandemic. We aimed to assess the safety and efficacy of the COVID-19 vaccine SCB-2019. METHODS This ongoing phase 2 and 3 double-blind, placebo-controlled trial was done in adults aged 18 years and older who were in good health or with a stable chronic health condition, at 31 sites in five countries (Belgium, Brazil, Colombia, Philippines, and South Africa). The participants were randomly assigned 1:1 using a centralised internet randomisation system to receive two 0·5 mL intramuscular doses of SCB-2019 (30 μg, adjuvanted with 1·50 mg CpG-1018 and 0·75 mg alum) or placebo (0·9% sodium chloride for injection supplied in 10 mL ampoules) 21 days apart. All study staff and participants were masked, but vaccine administrators were not. Primary endpoints were vaccine efficacy, measured by RT-PCR-confirmed COVID-19 of any severity with onset from 14 days after the second dose in baseline SARS-CoV-2 seronegative participants (the per-protocol population), and the safety and solicited local and systemic adverse events in the phase 2 subset. This study is registered on EudraCT (2020-004272-17) and ClinicalTrials.gov (NCT04672395). FINDINGS 30 174 participants were enrolled from March 24, 2021, until the cutoff date of Aug 10, 2021, of whom 30 128 received their first assigned vaccine (n=15 064) or a placebo injection (n=15 064). The per-protocol population consisted of 12 355 baseline SARS-CoV-2-naive participants (6251 vaccinees and 6104 placebo recipients). Most exclusions (13 389 [44·4%]) were because of seropositivity at baseline. There were 207 confirmed per-protocol cases of COVID-19 at 14 days after the second dose, 52 vaccinees versus 155 placebo recipients, and an overall vaccine efficacy against any severity COVID-19 of 67·2% (95·72% CI 54·3-76·8), 83·7% (97·86% CI 55·9-95·4) against moderate-to-severe COVID-19, and 100% (97·86% CI 25·3-100·0) against severe COVID-19. All COVID-19 cases were due to virus variants; vaccine efficacy against any severity COVID-19 due to the three predominant variants was 78·7% (95% CI 57·3-90·4) for delta, 91·8% (44·9-99·8) for gamma, and 58·6% (13·3-81·5) for mu. No safety issues emerged in the follow-up period for the efficacy analysis (median of 82 days [IQR 63-103]). The vaccine elicited higher rates of mainly mild-to-moderate injection site pain than the placebo after the first (35·7% [287 of 803] vs 10·3% [81 of 786]) and second (26·9% [189 of 702] vs 7·4% [52 of 699]) doses, but the rates of other solicited local and systemic adverse events were similar between the groups. INTERPRETATION Two doses of SCB-2019 vaccine plus CpG and alum provides notable protection against the entire severity spectrum of COVID-19 caused by circulating SAR-CoV-2 viruses, including the predominating delta variant. FUNDING Clover Biopharmaceuticals and the Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
- Lulu Bravo
- University of the Philippines Manila, Ermita, Manila, Philippines
| | | | | | - Ping Li
- Clover Biopharmaceuticals, Cambridge, MA, USA
| | | | - Frank Rockhold
- Duke University Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Camilo Roa
- Manila Doctors Hospital, Manila, Philippines
| | | | | | - Pio Lopez
- Centro de Estudios en Infectología Pediátrica, Universidad Del Valle Clínica Imbanaco, Cali, Colombia
| | - Eduardo López-Medina
- Centro de Estudios en Infectología Pediátrica, Universidad Del Valle Clínica Imbanaco, Cali, Colombia
| | | | | | | | - Tatiana Medina
- Center of Attention in Medical Research, Bogotá, Colombia
| | | | | | | | | | | | - Eduardo Sprinz
- Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | - Maria Sanali Paiva
- Atena Institute of Clinical Research, Rio Grande do Norte, Natal, Brazil
| | - Josefina Carlos
- University of the East Ramon Magsaysay Memorial Medical Center, Quezon City, Philippines
| | | | | | - Charles Y Yu
- De La Salle Medical and Health Sciences Institute, Cavite City, Philippines
| | | | - Mario M Panaligan
- Infection Control Service, St Luke's Medical Center, Taguig, Philippines
| | | | | | - Maya Hites
- Clinic of Infectious Diseases, CUB-Hôpital Erasme, Bruxelles, Belgium
| | - Jean-Benoit Martinot
- Pulmonology Department, CHU Universite Catholique de Louvain Namur Site Sainte-Elisabeth, Namur, Belgium
| | - Qasim E Bhorat
- Soweto Clinical Trials Centre, Johannesburg, South Africa
| | - Aysha Badat
- Wits Clinical Research, Soweto, Johannesburg, South Africa
| | | | - Branda Hu
- Clover Biopharmaceuticals, Cambridge, MA, USA
| | - Jaco Jurgens
- DJW Research, Noordheuwel, Krugersdorp, Gauteng, South Africa
| | - Jan Engelbrecht
- Dr JM Engelbrecht Trial Site, Vergelegen Mediclinic, Western Cape, South Africa
| | | | - Peter Richmond
- Division of Paediatrics, University of Western Australia, Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute and Perth Children's Hospital, Perth, WA, Australia
| | | | | | - Ralf Clemens
- Global Research in Infectious Diseases, Rio de Janeiro, Brazil.
| |
Collapse
|
1339
|
Huang X, Yan Y, Su B, Xiao D, Yu M, Jin X, Duan J, Zhang X, Zheng S, Fang Y, Zhang T, Tang W, Wang L, Wang Z, Xu J. Comparing Immune Responses to Inactivated Vaccines against SARS-CoV-2 between People Living with HIV and HIV-Negative Individuals: A Cross-Sectional Study in China. Viruses 2022; 14:277. [PMID: 35215870 PMCID: PMC8875274 DOI: 10.3390/v14020277] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
This study compared the immunogenicity of inactivated SARS-CoV-2 vaccines between people living with HIV (PLWH) and HIV-negative individuals. We recruited 120 PLWH and 53 HIV-negative individuals aged 18-59 years who had received an inactivated SARS-CoV-2 vaccine in two Chinese cities between April and June 2021. Blood samples were tested for immunogenicity of the inactivated SARS-CoV-2 vaccines. The prevalence and severity of adverse events associated with SARS-CoV-2 vaccines were similar between PLWH and HIV-negative individuals. The seropositivity of neutralizing activity against authentic SARS-CoV-2, of the total amount of antibody (total antibody) and of S-IgG were 71.3%, 81.9%, and 92.6%, respectively, among fully vaccinated PLWH. Among all participants, PLWH had lower neutralizing activity, total antibody, S-IgG, and T-cell-specific immune response levels, compared to HIV-negative individuals, after controlling for types of vaccine, time interval between first and second dose, time after receiving the second dose, and sociodemographic factors. PLWH with a longer interval since HIV diagnosis, who received their second dose 15-28 days prior to study commencement, and who had an interval of ≥21 days between first and second dose had higher neutralizing activity levels. The immunogenicity of the inactivated SARS-CoV-2 vaccines was lower among PLWH as compared to HIV-negative individuals. Vaccination guideline specific for PLWH should be developed.
Collapse
Affiliation(s)
- Xiaojie Huang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (X.H.); (B.S.); (J.D.); (T.Z.)
| | - Ying Yan
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Bin Su
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (X.H.); (B.S.); (J.D.); (T.Z.)
| | - Dong Xiao
- Rainbow Clinic of Beijing Jingcheng Skin Hospital, Beijing 100101, China;
| | - Maohe Yu
- Department of AIDS/STD Control and Prevention, Tianjin Center for Disease Control and Prevention, Tianjin 300011, China;
| | - Xia Jin
- AIDS Healthcare Foundation (AHF), Beijing 100088, China;
| | - Junyi Duan
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (X.H.); (B.S.); (J.D.); (T.Z.)
| | - Xiangjun Zhang
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA;
| | - Shimin Zheng
- Department of Biostatistics and Epidemiology, East Tennessee State University, Johnson City, TN 70300, USA;
| | - Yuan Fang
- Department of Health and Physical Education, The Education University of Hong Kong, Hong Kong 200092, China;
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (X.H.); (B.S.); (J.D.); (T.Z.)
| | - Weiming Tang
- University of North Carolina Project-China, Guangzhou 510095, China
| | - Lunan Wang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Zixin Wang
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 666888, China
| | - Junjie Xu
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen 518036, China
| |
Collapse
|
1340
|
Grodzki M, Bluhm AP, Schaefer M, Tagmount A, Russo M, Sobh A, Rafiee R, Vulpe CD, Karst SM, Norris MH. Genome-scale CRISPR screens identify host factors that promote human coronavirus infection. Genome Med 2022; 14:10. [PMID: 35086559 PMCID: PMC8792531 DOI: 10.1186/s13073-022-01013-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has resulted in 275 million infections and 5.4 million deaths as of December 2021. While effective vaccines are being administered globally, there is still a great need for antiviral therapies as antigenically novel SARS-CoV-2 variants continue to emerge across the globe. Viruses require host factors at every step in their life cycle, representing a rich pool of candidate targets for antiviral drug design. METHODS To identify host factors that promote SARS-CoV-2 infection with potential for broad-spectrum activity across the coronavirus family, we performed genome-scale CRISPR knockout screens in two cell lines (Vero E6 and HEK293T ectopically expressing ACE2) with SARS-CoV-2 and the common cold-causing human coronavirus OC43. Gene knockdown, CRISPR knockout, and small molecule testing in Vero, HEK293, and human small airway epithelial cells were used to verify our findings. RESULTS While we identified multiple genes and functional pathways that have been previously reported to promote human coronavirus replication, we also identified a substantial number of novel genes and pathways. The website https://sarscrisprscreens.epi.ufl.edu/ was created to allow visualization and comparison of SARS-CoV2 CRISPR screens in a uniformly analyzed way. Of note, host factors involved in cell cycle regulation were enriched in our screens as were several key components of the programmed mRNA decay pathway. The role of EDC4 and XRN1 in coronavirus replication in human small airway epithelial cells was verified. Finally, we identified novel candidate antiviral compounds targeting a number of factors revealed by our screens. CONCLUSIONS Overall, our studies substantiate and expand the growing body of literature focused on understanding key human coronavirus-host cell interactions and exploit that knowledge for rational antiviral drug development.
Collapse
Affiliation(s)
- Marco Grodzki
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Andrew P Bluhm
- Department of Geography, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Moritz Schaefer
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Max Russo
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
- Present address: Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Amin Sobh
- Univeristy of Florida Heath Cancer Center, University of Florida, Gainesville, FL, USA
| | - Roya Rafiee
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Chris D Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Stephanie M Karst
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Michael H Norris
- Department of Geography, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
1341
|
Orfanoudaki E, Zacharopoulou E, Kitsou V, Karmiris K, Theodoropoulou A, Mantzaris GJ, Tzouvala M, Michopoulos S, Zampeli E, Michalopoulos G, Karatzas P, Viazis N, Liatsos C, Bamias G, Koutroubakis IE, on behalf of the Hellenic Group for the Study of IBD. Real-World Use and Adverse Events of SARS-CoV-2 Vaccination in Greek Patients with Inflammatory Bowel Disease. J Clin Med 2022; 11:641. [PMID: 35160092 PMCID: PMC8836981 DOI: 10.3390/jcm11030641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/12/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Since inflammatory bowel disease (IBD) patients were excluded from vaccine authorization studies, limited knowledge exists regarding perceptions and unfavorable effects of COVID-19 vaccination in this group. We aimed to investigate the real-world use and adverse events (AEs) of COVID-19 vaccines in Greek IBD patients. Fully vaccinated IBD patients followed in Greek centers were invited to participate. All patients filled out an anonymous online survey concerning the vaccination program, which included information regarding demographics, clinical characteristics, treatment, vaccination perceptions and potential AEs. Overall, 1007 IBD patients were included. Vaccine hesitancy was reported by 49%. Total AEs to vaccination were reported by 81% after dose 1 (D1) and 76% after dose 2 (D2), including isolated injection site reactions (36% and 24% respectively). Systemic AEs were more common after D2 (51%, D2 vs. 44%, D1, p < 0.0001). Very few patients reported new onset abdominal symptoms (abdominal pain 4% (D1), 6% (D2) and diarrhea 5% (D1), 7% (D2)). There were no serious AEs leading to emergency room visit or hospitalization. In multivariate analysis, AEs occurrence was positively associated with young age and female gender (p < 0.0005 for both doses), whereas inactive disease was negatively associated with AE in D1 (p = 0.044). SARS-CoV-2 vaccination in Greek IBD patients demonstrated a favorable and reassuring safety profile.
Collapse
Affiliation(s)
- Eleni Orfanoudaki
- Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, 71110 Heraklion, Greece;
| | - Eirini Zacharopoulou
- Department of Gastroenterology, General Hospital of Nikaia Piraeus “Ag. Panteleimon”-General Hospital Dytikis Attikis “Agia Varvara”, 12351 Athens, Greece; (E.Z.); (M.T.)
| | - Vassiliki Kitsou
- Gastroenterology Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian Univeristy of Athens, “Sotiria” General Hospital, 11527 Athens, Greece; (V.K.); (G.B.)
| | - Konstantinos Karmiris
- Department of Gastroenterology, Venizelio General Hospital, 71409 Heraklion, Greece; (K.K.); (A.T.)
| | - Angeliki Theodoropoulou
- Department of Gastroenterology, Venizelio General Hospital, 71409 Heraklion, Greece; (K.K.); (A.T.)
| | - Gerassimos J. Mantzaris
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos-Polykliniki, 10676 Athens, Greece; (G.J.M.); (N.V.)
| | - Maria Tzouvala
- Department of Gastroenterology, General Hospital of Nikaia Piraeus “Ag. Panteleimon”-General Hospital Dytikis Attikis “Agia Varvara”, 12351 Athens, Greece; (E.Z.); (M.T.)
| | - Spyridon Michopoulos
- Department of Gastroenterology, General Hospital of Athens “Alexandra”, 11528 Athens, Greece; (S.M.); (E.Z.)
| | - Evanthia Zampeli
- Department of Gastroenterology, General Hospital of Athens “Alexandra”, 11528 Athens, Greece; (S.M.); (E.Z.)
| | - Georgios Michalopoulos
- Department of Gastroenterology, General Hospital of Piraeus “Tzaneio”, Piraeus, 18536 Athens, Greece;
| | - Pantelis Karatzas
- Department of Gastroenterology, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece;
| | - Nikos Viazis
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos-Polykliniki, 10676 Athens, Greece; (G.J.M.); (N.V.)
| | - Christos Liatsos
- Gastroenterology Department, 401 General Army Hospital of Athens, 11525 Athens, Greece;
| | - Giorgos Bamias
- Gastroenterology Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian Univeristy of Athens, “Sotiria” General Hospital, 11527 Athens, Greece; (V.K.); (G.B.)
| | - Ioannis E. Koutroubakis
- Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, 71110 Heraklion, Greece;
| | | |
Collapse
|
1342
|
Liu H, Wei P, Kappler JW, Marrack P, Zhang G. SARS-CoV-2 Variants of Concern and Variants of Interest Receptor Binding Domain Mutations and Virus Infectivity. Front Immunol 2022; 13:825256. [PMID: 35154144 PMCID: PMC8828474 DOI: 10.3389/fimmu.2022.825256] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 12/24/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has lasted more than 2 years with over 260 million infections and 5 million deaths worldwide as of November 2021. To combat the virus, monoclonal antibodies blocking the virus binding to human receptor, the angiotensin converting enzyme 2 (ACE2), have been approved to treat the infected patients. Inactivated whole virus or the full-length virus spike encoding adenovirus or mRNA vaccines are being used to immunize the public. However, SARS-CoV-2 variants are emerging. These, to some extent, escape neutralization by the therapeutic antibodies and vaccine-induced immunity. Thus, breakthrough infections by SARS-CoV-2 variants have been reported in previously virus-infected or fully vaccinated individuals. The receptor binding domain (RBD) of the virus spike protein reacts with host ACE2, leading to the entry of the virus into the cell. It is also the major antigenic site of the virus, with more than 90% of broadly neutralizing antibodies from either infected patients or vaccinated individuals targeting the spike RBD. Therefore, mutations in the RBD region are effective ways for SARS-CoV-2 variants to gain infectivity and escape the immunity built up by the original vaccines or infections. In this review, we focus on the impact of RBD mutations in SARS-CoV-2 variants of concern (VOC) and variants of interest (VOI) on ACE2 binding affinity and escape of serum antibody neutralization. We also provide protein structure models to show how the VOC and VOI RBD mutations affect ACE2 binding and allow escape of the virus from the therapeutic antibody, bamlanivimab.
Collapse
Affiliation(s)
- Haolin Liu
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Center, University of Colorado, Aurora, CO, United States
| | - Pengcheng Wei
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Center, University of Colorado, Aurora, CO, United States
| | - John W. Kappler
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Center, University of Colorado, Aurora, CO, United States
| | - Philippa Marrack
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Center, University of Colorado, Aurora, CO, United States
| | - Gongyi Zhang
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Center, University of Colorado, Aurora, CO, United States
| |
Collapse
|
1343
|
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the unprecedented pace of development of multiple vaccines. This review evaluates how adenovirus (Ad) vector platforms have been leveraged in response to this pandemic. Ad vectors have been used in the past for vaccines against other viruses, most notably HIV and Ebola, but they never have been produced, distributed, or administered to humans at such a large scale. Several different serotypes of Ads encoding SARS-CoV-2 Spike have been tested and found to be efficacious against COVID-19. As vaccine rollouts continue and the number of people receiving these vaccines increases, we will continue to learn about this vaccine platform for COVID-19 prevention and control.
Collapse
Affiliation(s)
- Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA;
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA;
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
1344
|
Borah P, Xiao X, Lai Lee DK. Narrative messages, information seeking and COVID-19 vaccine intention: The moderating role of perceived behavioral control. Am J Health Promot 2022; 36:923-933. [PMID: 35081757 PMCID: PMC8960749 DOI: 10.1177/08901171221075019] [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] [Indexed: 11/21/2022]
Abstract
Purpose The main purposes of the current study are to examine 1) the influence of narrative vs statistics messages on COVID-19 related information seeking and COVID-19 vaccine intention and 2) the moderating role of perceived behavioral control (PBC). Design Data for a between-subject randomized experiment were collected online. The manipulation messages were presented as screenshots from the CDC’s Facebook page. Setting The participants were recruited from Amazon MTurk. Subjects A total of 300 subjects participated in the study, who were 18 years and above (M = 38.40). Measures Intention to seek information, COVID-19 vaccine intention, and PBC. Analysis To test the hypotheses, we utilized Hayes’s (2014) PROCESS for SPSS (Model 1). For intention to seek information, the main effect of the message manipulation (narrative vs statistics) [b = −2.10, t (300) = −4.14, P < .001] and the interaction [b = .41, t (300) = 3.88, P < .001] were significant. For vaccine intention, the main effects of message manipulation [b = 1.64, t (300) = −2.61, P < .005] and the interaction [b = .34, t (300) = 2.64, P < .005] were significant. Results Our research found that narrative messages were more persuasive for both information seeking and vaccine intention. But this was true only in the case of individuals whose PBC was low. Conclusions Our findings have critical implications for vaccine promotion research.
Collapse
Affiliation(s)
| | - Xizhu Xiao
- School of Journalism and Communication12593Qingdao University
| | | |
Collapse
|
1345
|
Alberca GGF, Alberca RW. Role of vitamin D deficiency and comorbidities in COVID-19. World J Virol 2022; 11:85-89. [PMID: 35117974 PMCID: PMC8788214 DOI: 10.5501/wjv.v11.i1.85] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/01/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Recent manuscripts described the incidence of vitamin D hypovitaminosis in coronavirus disease 2019 (COVID-19) patients. Vitamin D deficiency is also common in patients with comorbidities that are associated with a poor COVID-19 prognosis. In this letter, we review the literature regarding the association of comorbidities, vitamin D deficiency, and COVID-19.
Collapse
Affiliation(s)
- Gabriela Gama Freire Alberca
- Department of Microbiology, Institute of Biomedical Sciences-University of São Paulo, São Paulo 04307-100, Brazil
| | - Ricardo Wesley Alberca
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 04307-100, Brazil
| |
Collapse
|
1346
|
Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primates. NPJ Vaccines 2022; 7:7. [PMID: 35064109 PMCID: PMC8782996 DOI: 10.1038/s41541-022-00436-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022] Open
Abstract
Second-generation COVID-19 vaccines could contribute to establish protective immunity against SARS-CoV-2 and its emerging variants. We developed COH04S1, a synthetic multiantigen modified vaccinia Ankara-based SARS-CoV-2 vaccine that co-expresses spike and nucleocapsid antigens. Here, we report COH04S1 vaccine efficacy in animal models. We demonstrate that intramuscular or intranasal vaccination of Syrian hamsters with COH04S1 induces robust Th1-biased antigen-specific humoral immunity and cross-neutralizing antibodies (NAb) and protects against weight loss, lower respiratory tract infection, and lung injury following intranasal SARS-CoV-2 challenge. Moreover, we demonstrate that single-dose or two-dose vaccination of non-human primates with COH04S1 induces robust antigen-specific binding antibodies, NAb, and Th1-biased T cells, protects against both upper and lower respiratory tract infection following intranasal/intratracheal SARS-CoV-2 challenge, and triggers potent post-challenge anamnestic antiviral responses. These results demonstrate COH04S1-mediated vaccine protection in animal models through different vaccination routes and dose regimens, complementing ongoing investigation of this multiantigen SARS-CoV-2 vaccine in clinical trials.
Collapse
|
1347
|
Effectiveness of Vaccination against SARS-CoV-2 Infection in the Pre-Delta Era: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2022; 10:vaccines10020157. [PMID: 35214615 PMCID: PMC8879968 DOI: 10.3390/vaccines10020157] [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: 11/17/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022] Open
Abstract
(1) Background: The objective of this study was to assess the effectiveness of SARS-CoV-2 vaccines in terms of prevention of disease and transmission in the pre-Delta era. The evaluation was narrowed to two mRNA vaccines and two modified adenovirus-vectored vaccines. (2) Methods: The overall risk of any SARS-CoV-2 infection confirmed by positive real-time Polymerase Chain Reaction (PCR) test was estimated in partially and fully vaccinated individuals. The evidence synthesis was pursued through a random-effects meta-analysis. The effect size was expressed as relative risk (RR) and RRR (RR reduction) of SARS-CoV-2 infection following vaccination. Heterogeneity was investigated through a between-study heterogeneity analysis and a subgroup meta-analysis. (3) Results: The systematic review identified 27 studies eligible for the quantitative synthesis. Partially vaccinated individuals presented a RRR = 73% (95%CI = 59–83%) for positive SARS-CoV-2 PCR (RR = 0.27) and a RRR=79% (95%CI = 30–93%) for symptomatic SARS-CoV-2 PCR (RR = 0.21). Fully vaccinated individuals showed a RRR = 94% (95%CI = 88–98%) for SARS-CoV-2 positive PCR (RR = 0.06) compared to unvaccinated individuals. The full BNT162b2 vaccination protocol achieved a RRR = 84–94% against any SARS-CoV-2-positive PCR and a RRR = 68–84% against symptomatic positive PCR. (4) Conclusions: The meta-analysis results suggest that full vaccination might block transmission. In particular, the risk of SARS-CoV-2 infection appeared higher for non-B.1.1.7 variants and individuals aged ≥69 years. Considering the high level of heterogeneity, these findings must be taken with caution. Further research on SARS-CoV-2 vaccine effectiveness against emerging SARS-CoV-2 variants is encouraged.
Collapse
|
1348
|
Korang SK, von Rohden E, Veroniki AA, Ong G, Ngalamika O, Siddiqui F, Juul S, Nielsen EE, Feinberg JB, Petersen JJ, Legart C, Kokogho A, Maagaard M, Klingenberg S, Thabane L, Bardach A, Ciapponi A, Thomsen AR, Jakobsen JC, Gluud C. Vaccines to prevent COVID-19: A living systematic review with Trial Sequential Analysis and network meta-analysis of randomized clinical trials. PLoS One 2022; 17:e0260733. [PMID: 35061702 PMCID: PMC8782520 DOI: 10.1371/journal.pone.0260733] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/11/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 is rapidly spreading causing extensive burdens across the world. Effective vaccines to prevent COVID-19 are urgently needed. METHODS AND FINDINGS Our objective was to assess the effectiveness and safety of COVID-19 vaccines through analyses of all currently available randomized clinical trials. We searched the databases CENTRAL, MEDLINE, Embase, and other sources from inception to June 17, 2021 for randomized clinical trials assessing vaccines for COVID-19. At least two independent reviewers screened studies, extracted data, and assessed risks of bias. We conducted meta-analyses, network meta-analyses, and Trial Sequential Analyses (TSA). Our primary outcomes included all-cause mortality, vaccine efficacy, and serious adverse events. We assessed the certainty of evidence with GRADE. We identified 46 trials; 35 trials randomizing 219 864 participants could be included in our analyses. Our meta-analyses showed that mRNA vaccines (efficacy, 95% [95% confidence interval (CI), 92% to 97%]; 71 514 participants; 3 trials; moderate certainty); inactivated vaccines (efficacy, 61% [95% CI, 52% to 68%]; 48 029 participants; 3 trials; moderate certainty); protein subunit vaccines (efficacy, 77% [95% CI, -5% to 95%]; 17 737 participants; 2 trials; low certainty); and viral vector vaccines (efficacy 68% [95% CI, 61% to 74%]; 71 401 participants; 5 trials; low certainty) prevented COVID-19. Viral vector vaccines decreased mortality (risk ratio, 0.25 [95% CI 0.09 to 0.67]; 67 563 participants; 3 trials, low certainty), but comparable data on inactivated, mRNA, and protein subunit vaccines were imprecise. None of the vaccines showed evidence of a difference on serious adverse events, but observational evidence suggested rare serious adverse events. All the vaccines increased the risk of non-serious adverse events. CONCLUSIONS The evidence suggests that all the included vaccines are effective in preventing COVID-19. The mRNA vaccines seem most effective in preventing COVID-19, but viral vector vaccines seem most effective in reducing mortality. Further trials and longer follow-up are necessary to provide better insight into the safety profile of these vaccines.
Collapse
Affiliation(s)
- Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Elena von Rohden
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Areti Angeliki Veroniki
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Metabolism, Digestion and Reproduction & Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, United Kingdom
| | - Giok Ong
- Systematic Review Initiative, NHS Blood and Transplant, John Radcliffe Hospital, Headley Way, Oxford, United Kingdom
| | - Owen Ngalamika
- Dermatology & Venereology Division, University Teaching Hospital, University of Zambia School of Medicine, Lusaka, Zambia
| | - Faiza Siddiqui
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sophie Juul
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Emil Eik Nielsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Joshua Buron Feinberg
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Johanne Juul Petersen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Christian Legart
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Afoke Kokogho
- United States Army Medical Research Directorate West Africa, Henry M. Jackson Foundation Medical Research International (HJFMRI), Walter Reed Army Institute of Research, Abuja, Nigeria
| | - Mathias Maagaard
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Centre for Anaesthesiological Research, Department of Anaesthesiology, Zealand University Hospital, The Zealand Region of Denmark, Køge, Denmark
| | - Sarah Klingenberg
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Lehana Thabane
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Ariel Bardach
- Argentine Cochrane Center. Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
| | - Agustín Ciapponi
- Argentine Cochrane Center. Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
| | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Janus C Jakobsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Heath Sciences, University of Southern Denmark, Odense, Denmark
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Heath Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
1349
|
Montero-Menárguez J, Falkenhain-López D, Guzmán-Pérez LM, Rodríguez-Peralto JL, Tous-Romero F, Gallego-Álvarez S. Massive bullous local reaction following administration of Ad26.COV2.S COVID-19 vaccine. Int J Dermatol 2022; 61:636-638. [PMID: 35049059 DOI: 10.1111/ijd.16037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/19/2021] [Accepted: 12/08/2021] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | - Fátima Tous-Romero
- Department of Dermatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | |
Collapse
|
1350
|
Corrao G, Bertolaso G, Pavesi G, Moratti L. Eight Good Reasons for Careful Monitoring and Evaluation of the Vaccine Campaign against COVID-19: Lessons Learned through the Lombardy Experience for Dealing with Next Challenges. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031073. [PMID: 35162097 PMCID: PMC8834613 DOI: 10.3390/ijerph19031073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
Background: Using the knowledge gained during the first eleven months of the vaccine campaign in Lombardy, Italy, we provide an overview of the benefits of using reliable, complete, and rapidly available observational data to monitor the progress of the vaccine strategy. Methods: A population-based platform was implemented by linking four registries reporting individual data on: (i) date, type, and dose of vaccine dispensed; (ii) SARS-CoV-2 infections and hospital admissions and deaths due to COVID-19; (iii) inpatient diagnoses and outpatient services supplied by the Regional Health Services (RHS); and the (iv) health registry reporting and updating data on patient status. Background, methods, findings, and implications of eight COVID-19 relevant questions are reported. Results: Before starting the vaccine campaign, we identified high-risk individuals who need to be prioritized. During the vaccine campaign, we: (i) monitored the trend in the speed of the vaccine campaign progression and the number of prevented clinical outcomes; (ii) verified that available vaccines work in real-life, assessed their effectiveness-harm profile, and measured their reduced effectiveness against the delta variant. Finally, we studied the reduced effectiveness of the vaccine over time and identified risk factors of post-vaccine infection and severe illness. Conclusions: The correct use of rapidly available observational data of good quality and completeness generates reliable evidence to promptly inform patients and policymakers.
Collapse
Affiliation(s)
- Giovanni Corrao
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
- Correspondence: ; Tel.: +39-02-64485854
| | - Guido Bertolaso
- Vaccination Campaign Management, Lombardy Region, 20124 Milan, Italy;
| | - Giovanni Pavesi
- General Directorate of Welfare Department, Lombardy Region, 20124 Milan, Italy;
| | | |
Collapse
|