451
|
Liu J, Cang T, Jiang C, Li K, Liu S, Wang H, Wang M, Chen Y, Shao Y, Liu J. CpG 684: an effective adjuvant for the inactivated COVID-19 vaccine in mice. Future Virol 2023; 18:403-410. [PMID: 38051998 PMCID: PMC10241461 DOI: 10.2217/fvl-2022-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 04/05/2023] [Indexed: 12/07/2023]
Abstract
Aim This study used CpG 684 as adjuvant of inactivated COVID-19 vaccine to detect a humoral and cellular immune response in mice. Materials & methods We used 10 and 20 µg CpG 684 as adjuvants of an inactivated COVID-19 vaccine to immunize mice. IgG, IgG1, IgG2a, IgG2b and IgM binding antibodies were detected in serum by ELISA. The IFN-γ cytokine was detected by ELISPOT. Results CpG 684 improved spike-specific IgG and IgM subtype binding antibodies and increased the neutralizing antibody titer against prototype, Delta and Beta strains. CpG 684 also improved cellular immune response. Conclusion CpG 684 is an effective adjuvant for inactivated COVID-19 vaccine.
Collapse
Affiliation(s)
- Jiandong Liu
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Tianle Cang
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Congli Jiang
- Shenzhen Kangtai Biological Products Co., Ltd, Guangzhou, Shenzhen, 518000, China
| | - Kelei Li
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Siyuan Liu
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Haixin Wang
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Meirong Wang
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| | - Yan Chen
- Jiangsu Taipuriu Biotechnology Co., Ltd, Taizhou Pharmaceutical City, Jiangsu, 225300, China
| | - Yan Shao
- Jiangsu Taipuriu Biotechnology Co., Ltd, Taizhou Pharmaceutical City, Jiangsu, 225300, China
| | - Jiankai Liu
- Beijing Minhai Biotechnology Co. Ltd, Beijing, 102600, China
| |
Collapse
|
452
|
Harris CE, Vijenthira A, Ong SY, Baden LR, Hicks LK, Baird JH. COVID-19 and Other Viral Infections in Patients With Hematologic Malignancies. Am Soc Clin Oncol Educ Book 2023; 43:e390778. [PMID: 37163714 DOI: 10.1200/edbk_390778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
COVID-19 and our armamentarium of strategies to combat it have evolved dramatically since the virus first emerged in late 2019. Vaccination remains the primary strategy to prevent severe illness, although the protective effect can vary in patients with hematologic malignancy. Strategies such as additional vaccine doses and now bivalent boosters can contribute to increased immune response, especially in the face of evolving viral variants. Because of these new variants, no approved monoclonal antibodies are available for pre-exposure or postexposure prophylaxis. Patients with symptomatic, mild-to-moderate COVID-19 and risk features for developing severe COVID-19, who present within 5-7 days of symptom onset, should be offered outpatient therapy with nirmatrelvir/ritonavir (NR) or in some cases with intravenous (IV) remdesivir. NR interacts with many blood cancer treatments, and reviewing drug interactions is essential. Patients with severe COVID-19 should be managed with IV remdesivir, tocilizumab (or an alternate interleukin-6 receptor blocker), or baricitinib, as indicated based on the severity of illness. Dexamethasone can be considered on an individual basis, weighing oxygen requirements and patients' underlying disease and their perceived ability to clear infection. Finally, as CD19-targeted and B-cell maturation (BCMA)-targeted chimeric antigen receptor (CAR) T-cell therapies become more heavily used for relapsed/refractory hematologic malignancies, viral infections including COVID-19 are increasingly recognized as common complications, but data on risk factors and prophylaxis in this patient population are scarce. We summarize the available evidence regarding viral infections after CAR T-cell therapy.
Collapse
Affiliation(s)
- Courtney E Harris
- Brigham and Women's Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Abi Vijenthira
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Shin Yeu Ong
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| | - Lindsey Robert Baden
- Brigham and Women's Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Lisa K Hicks
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Hematology/Oncology, St Michael's Hospital, Toronto, Canada
| | - John H Baird
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
453
|
Abstract
PURPOSE OF REVIEW Persons living with HIV (PLWH) may have a moderately increased risk of morbidity and mortality from COVID-19 infection, especially if viral load is not controlled and if they are immunosuppressed. Vaccination against SARS-CoV-2 is the most effective measure to prevent morbidity and mortality. However, individuals with HIV/AIDS may have less protection after vaccination. The purpose of this review is to summarize some of the recent studies focused on examining the safety, immunogenicity and effectiveness of anti-SARS-CoV-2 vaccines. RECENT FINDINGS The safety of all anti-SARS-CoV-2 vaccines among PLWH is not different from the safety of these vaccines among HIV-negative individuals and is acceptable. PLWH with viral suppression and immune reconstitution (CD4 + cell count > 350 cells/μl) may reach almost same immunogenicity such as people without HIV albeit antibody levels and neutralization may decline more rapidly than in people without HIV. PLWH with viremia or immunosuppressed, especially AIDS, have less immunogenicity. SUMMARY Full vaccination against SARS-CoV-2 is a well tolerated and efficient way to prevent mortality and morbidity from COVID-19 among PLWH and AIDS patients. It is very important to follow recommended booster vaccination for a continuous and prompt immunogenicity.
Collapse
Affiliation(s)
- Itzchak Levy
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer
- Sackler Medical School, Tel Aviv University, Tel Aviv, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer
- Sackler Medical School, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
454
|
Hernández-Bernal F, Ricardo-Cobas MC, Martín-Bauta Y, Rodríguez-Martínez E, Urrutia-Pérez K, Urrutia-Pérez K, Quintana-Guerra J, Navarro-Rodríguez Z, Piñera-Martínez M, Rodríguez-Reinoso JL, Chávez-Chong CO, Baladrón-Castrillo I, Melo-Suárez G, Batista-Izquierdo A, Pupo-Micó A, Mora-Betancourt R, Bizet-Almeida J, Martínez-Rodríguez MC, Lobaina-Lambert L, Velázquez-Pérez VM, Soler-Díaz J, Laurencio-Vallina S, Meriño-Hechavarría T, Carmenaty-Campos N, Rodríguez-Montero E, Limonta-Fernández M, Alonso-Valdés M, Hernández-Rodríguez R, Pimentel-Vázquez E, Catasús-Álvarez KM, Cabrera-Núñez MV, Ayala-Ávila M, Muzio-González VL. A phase 3, randomised, double-blind, placebo-controlled clinical trial evaluation of the efficacy and safety of a SARS-CoV-2 recombinant spike RBD protein vaccine in adults (ABDALA-3 study). LANCET REGIONAL HEALTH. AMERICAS 2023; 21:100497. [PMID: 37192953 PMCID: PMC10160525 DOI: 10.1016/j.lana.2023.100497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/28/2023] [Accepted: 04/14/2023] [Indexed: 05/18/2023]
Abstract
Background The pandemic of COVID-19 raised the urgent need for safe and efficacious vaccines against SARS-CoV-2. We evaluated the efficacy and safety of a new SARS-CoV-2 virus receptor-binding domain (RBD) vaccine. Methods A phase 3, multicentre, randomised, double-blind, placebo-controlled trial was carried out at 18 clinical sites in three provinces of the south-eastern region of Cuba. Subjects (healthy or those with controlled chronic diseases) aged between 19 and 80 years, who gave written informed consent were eligible. Subjects were randomly assigned (1:1, in blocks) to two groups: placebo, and 50 μg RBD vaccine (Abdala). The product was administered intramuscularly, 0.5 mL in the deltoid region, in a three-dose immunization schedule at 0-14-28 days. The organoleptic characteristics and presentations of the vaccine and placebo were identical. All participants (subjects, clinical researchers, statisticians, laboratory technicians, and monitors) remained blinded during the study period. The main endpoint was to evaluate the efficacy of the Abdala vaccine in the prevention of symptomatic COVID-19. The trial is registered with the Cuban Public Registry of Clinical Trials, RPCEC00000359. Findings Between March 22 to April 03, 2021, 48,290 subjects were included (24,144 and 24,146 in the placebo and Abdala groups, respectively) in the context of predominant D614G variant circulation. The evaluation of the main efficacy outcomes occurred during May-June 2021, starting at May 3rd, in the context of high circulation of mutant viruses, predominantly VOC Beta. The incidence of adverse reactions for individuals in the placebo and Abdala vaccine groups were 1227/24,144 (5.1%) and 1621/24,146 (6.7%), respectively. Adverse reactions were mostly mild, and from the injection site, which resolved in the first 24-48 h. No severe adverse events with demonstrated cause-effect relationship attributable to the vaccine were reported. Symptomatic COVID-19 disease was confirmed in 142 participants in the placebo group (78.44 incidence per 1000 person-years, 95% confidence interval [CI], 66.07-92.46) and in 11 participants in Abdala vaccine group (6.05 incidence per 1000 person years; 95% CI 3.02-10.82). The Abdala vaccine efficacy against symptomatic COVID-19 was 92.28% (95% CI 85.74-95.82). Moderate/serious forms of COVID-19 occurred in 30 participants (28 in the placebo group and only 2 in the Abdala vaccine group) for a vaccine efficacy of 92.88% (95% CI 70.12-98.31). There were five critical patients (of which four died), all in the placebo group. Interpretation The Abdala vaccine was safe, well tolerated, and highly effective, fulfilling the WHO target product profile for COVID-19 vaccines. Those results, along with its immunization schedule and the advantage of easy storage and handling conditions at 2-8 °C, make this vaccine an option for the use in immunization strategies as a key tool for the control of the pandemic. Funding Centre for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Maria V. Cabrera-Núñez
- Virology Laboratory of the Center for Hygiene, Epidemiology and Microbiology, Santiago de Cuba, Cuba
| | | | | |
Collapse
|
455
|
Crombé A, Bensid L, Seux M, Fadli D, Arnaud F, Benhamed A, Banaste N, Gorincour G. Impact of Vaccination and the Omicron Variant on COVID-19-related Chest CT Findings: A Multicenter Study. Radiology 2023. [PMID: 36880948 DOI: 10.1148/radiol.222730:222730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Background The SARS-CoV-2 Omicron variant has a higher infection rate than previous variants but results in less severe disease. However, the effects of Omicron and vaccination on chest CT findings are difficult to evaluate. Purpose To investigate the effect of vaccination status and predominant variant on chest CT findings, diagnostic scores, and severity scores in a multicenter sample of consecutive patients referred to emergency departments for proven COVID-19. Materials and Methods This retrospective multicenter study included adults referred to 93 emergency departments with SARS-CoV-2 infection according to a reverse-transcriptase polymerase chain reaction test and known vaccination status between July 2021 and March 2022. Clinical data and structured chest CT reports, including semiquantitative diagnostic and severity scores following the French Society of Radiology-Thoracic Imaging Society guidelines, were extracted from a teleradiology database. Observations were divided into Delta-predominant, transition, and Omicron-predominant periods. Associations between scores and variant and vaccination status were investigated with χ2 tests and ordinal regressions. Multivariable analyses evaluated the influence of Omicron variant and vaccination status on the diagnostic and severity scores. Results Overall, 3876 patients were included (median age, 68 years [quartile 1 to quartile 3 range, 54-80]; 1695 women). Diagnostic and severity scores were associated with the predominant variant (Delta vs Omicron, χ2 = 112.4 and 33.7, respectively; both P < .001) and vaccination status (χ2 = 243.6 and 210.1; both P < .001) and their interaction (χ2 = 4.3 [P = .04] and 28.7 [P < .001], respectively). In multivariable analyses, Omicron variant was associated with lower odds of typical CT findings than was Delta variant (odds ratio [OR], 0.46; P < .001). Two and three vaccine doses were associated with lower odds of demonstrating typical CT findings (OR, 0.32 and 0.20, respectively; both P < .001) and of having high severity score (OR, 0.47 and 0.33, respectively; both P < .001), compared with unvaccinated patients. Conclusion Both the Omicron variant and vaccination were associated with less typical chest CT manifestations of COVID-19 and lesser extent of disease. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Yoon and Goo in this issue.
Collapse
Affiliation(s)
- Amandine Crombé
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - Lounès Bensid
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - Mylène Seux
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - David Fadli
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - François Arnaud
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - Axel Benhamed
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - Nathan Banaste
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| | - Guillaume Gorincour
- From IMADIS, 48 rue Quivogne, Lyon 69002, France (A.C., L.B., M.S., D.F., F.A., N.B., G.G.); Department of Radiology, Pellegrin University Hospital and Bordeaux University, Bordeaux, France (A.C., D.F.); Ramsay Generale de Sante, Hopital Prive Clairval, Marseille, France (F.A.); Service SAMU-Urgences, Centre Hospitalier Universitaire Edouard Herriot, Hospices Civils de Lyon, Lyon, France (A.B.); Ramsay Generale de Sante, Clinique Convert, Bourg-en-Bresse, France (N.B.); and ELSAN, Clinique Bouchard, Marseille, France (G.G.)
| |
Collapse
|
456
|
Raghav PK, Mann Z, Ahluwalia SK, Rajalingam R. Potential treatments of COVID-19: Drug repurposing and therapeutic interventions. J Pharmacol Sci 2023; 152:1-21. [PMID: 37059487 PMCID: PMC9930377 DOI: 10.1016/j.jphs.2023.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The infection is caused when Spike-protein (S-protein) present on the surface of SARS-CoV-2 interacts with human cell surface receptor, Angiotensin-converting enzyme 2 (ACE2). This binding facilitates SARS-CoV-2 genome entry into the human cells, which in turn causes infection. Since the beginning of the pandemic, many different therapies have been developed to combat COVID-19, including treatment and prevention. This review is focused on the currently adapted and certain other potential therapies for COVID-19 treatment, which include drug repurposing, vaccines and drug-free therapies. The efficacy of various treatment options is constantly being tested through clinical trials and in vivo studies before they are made medically available to the public.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
| | | | - Simran Kaur Ahluwalia
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, Uttar Pradesh, India
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
457
|
Rahadi DA, Yusri E, Putra SP, Semiarty R, Pertiwi D, Ilmiawati C. COVID-19 Vaccination and Clinical Outcomes at a Secondary Referral Hospital During the Delta Variant-dominant Period in West Sumatra, Indonesia. J Prev Med Public Health 2023; 56:221-230. [PMID: 37287199 PMCID: PMC10248104 DOI: 10.3961/jpmph.23.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/20/2023] [Indexed: 06/09/2023] Open
Abstract
OBJECTIVES The second wave of coronavirus disease 2019 (COVID-19) cases in Indonesia, during which the Delta variant predominated, took place after a vaccination program had been initiated in the country. This study was conducted to assess the impact of COVID-19 vaccination on unfavorable clinical outcomes including hospitalization, severe COVID-19, intensive care unit (ICU) admission, and death using a real-world model. METHODS This single-center retrospective cohort study involved patients with COVID-19 aged ≥18 years who presented to the COVID-19 emergency room at a secondary referral teaching hospital between June 1, 2021 and August 31, 2021. We used a binary logistic regression model to assess the effect of COVID-19 vaccination on unfavorable clinical outcomes, with age, sex, and comorbidities as confounding variables. RESULTS A total of 716 patients were included, 32.1% of whom were vaccinated. The elderly participants (≥65 years) had the lowest vaccine coverage among age groups. Vaccination had an effectiveness of 50% (95% confidence interval [CI], 25 to 66) for preventing hospitalization, 97% (95% CI, 77 to 99) for preventing severe COVID-19, 95% (95% CI, 56 to 99) for preventing ICU admission, and 90% (95% CI, 22 to 99) for preventing death. Interestingly, patients with type 2 diabetes had a 2-fold to 4-fold elevated risk of unfavorable outcomes. CONCLUSIONS Among adults, COVID-19 vaccination has a moderate preventive impact on hospitalization but a high preventive impact on severe COVID-19, ICU admission, and death. The authors suggest that relevant parties increase COVID-19 vaccination coverage, especially in the elderly population.
Collapse
Affiliation(s)
- Didan Ariadapa Rahadi
- Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Elfira Yusri
- Department of Clinical Pathology, Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Syandrez Prima Putra
- Department of Microbiology, Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
- Center for Infectious Disease Diagnostic and Research (PDRPI), Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Rima Semiarty
- Department of Public Health, Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
- Doctoral Program of Public Health, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Dian Pertiwi
- Department of Clinical Pathology, Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Cimi Ilmiawati
- Doctoral Program of Public Health, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
- Department of Pharmacology, Undergraduate Program of Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| |
Collapse
|
458
|
Chen CY, Su TC. Benefits and Harms of COVID-19 Vaccines in Cardiovascular Disease: A Comprehensive Review. J Lipid Atheroscler 2023; 12:119-131. [PMID: 37265847 PMCID: PMC10232218 DOI: 10.12997/jla.2023.12.2.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 06/03/2023] Open
Abstract
Patients with a history of cardiovascular disease (CVD) who contract coronavirus disease 2019 (COVID-19) tend to have a worse prognosis and more severe cardiovascular side effects. COVID-19 vaccines, which are intended to prevent COVID-19, may also potentially reduce the severity and complications (including cardiovascular sequelae) of COVID-19, especially in patients with a history of CVD. However, there have also been reports of cardiovascular side effects from COVID-19 vaccines of various brands and types. The purpose of this study is to review the benefits and harms of COVID-19 vaccines in relation to CVD. In this thorough review of the most current evidence on the benefits and harms of COVID-19 vaccines, we present information about the characteristics of cardiovascular complications. Most of the evidence focuses on myocarditis or pericarditis, which are most strongly associated with mRNA vaccines and predominantly occur in young males within days of receiving the second dose. Meanwhile, post-vaccination myocardial infarction is more common in older males, and the first dose of adenoviral vector vaccines appears to play a greater role in this complication. This information may guide us in formulating alternative options and implementing targeted surveillance. Gaining more knowledge about the potential benefits and harms of COVID-19 vaccines will improve our ability to make informed decisions and judgments about the balance of these factors.
Collapse
Affiliation(s)
- Chung-Yen Chen
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
459
|
Rando HM, Lordan R, Kolla L, Sell E, Lee AJ, Wellhausen N, Naik A, Kamil JP, COVID-19 Review Consortium, Gitter A, Greene CS. The Coming of Age of Nucleic Acid Vaccines during COVID-19. mSystems 2023; 8:e0092822. [PMID: 36861992 PMCID: PMC10134841 DOI: 10.1128/msystems.00928-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics: the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within 2 weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last 2 decades in particular and suggest a new era in vaccines against emerging pathogens. IMPORTANCE The SARS-CoV-2 pandemic has caused untold damage globally, presenting unusual demands on but also unique opportunities for vaccine development. The development, production, and distribution of vaccines are imperative to saving lives, preventing severe illness, and reducing the economic and social burdens caused by the COVID-19 pandemic. Although vaccine technologies that provide the DNA or RNA sequence of an antigen had never previously been approved for use in humans, they have played a major role in the management of SARS-CoV-2. In this review, we discuss the history of these vaccines and how they have been applied to SARS-CoV-2. Additionally, given that the evolution of new SARS-CoV-2 variants continues to present a significant challenge in 2022, these vaccines remain an important and evolving tool in the biomedical response to the pandemic.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
| | - Ronan Lordan
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Likhitha Kolla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amruta Naik
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - COVID-19 Review Consortium
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
460
|
Barkhordarian M, Behbood A, Ranjbar M, Rahimian Z, Prasad A. Overview of the cardio-metabolic impact of the COVID-19 pandemic. Endocrine 2023; 80:477-490. [PMID: 37103684 PMCID: PMC10133915 DOI: 10.1007/s12020-023-03337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 04/28/2023]
Abstract
Evidence has shown that cardiometabolic disorders (CMDs) are amongst the top contributors to COVID-19 infection morbidity and mortality. The reciprocal impact of COVID-19 infection and the most common CMDs, the risk factors for poor composite outcome among patients with one or several underlying diseases, the effect of common medical management on CMDs and their safety in the context of acute COVID-19 infection are reviewed. Later on, the changes brought by the COVID-19 pandemic quarantine on the general population's lifestyle (diet, exercise patterns) and metabolic health, acute cardiac complications of different COVID-19 vaccines and the effect of CMDs on the vaccine efficacy are discussed. Our review identified that the incidence of COVID-19 infection is higher among patients with underlying CMDs such as hypertension, diabetes, obesity and cardiovascular disease. Also, CMDs increase the risk of COVID-19 infection progression to severe disease phenotypes (e.g. hospital and/or ICU admission, use of mechanical ventilation). Lifestyle modification during COVID-19 era had a great impact on inducing and worsening of CMDs. Finally, the lower efficacy of COVID-19 vaccines was found in patients with metabolic disease.
Collapse
Affiliation(s)
- Maryam Barkhordarian
- Department of Medicine, Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Arezoo Behbood
- MPH department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Maryam Ranjbar
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Zahra Rahimian
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Anand Prasad
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
461
|
Rzymski P. Guillain-Barré syndrome and COVID-19 vaccines: focus on adenoviral vectors. Front Immunol 2023; 14:1183258. [PMID: 37180147 PMCID: PMC10169623 DOI: 10.3389/fimmu.2023.1183258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
COVID-19 vaccination is a life-saving intervention. However, it does not come up without a risk of rare adverse events, which frequency varies between vaccines developed using different technological platforms. The increased risk of Guillain-Barré syndrome (GBS) has been reported for selected adenoviral vector vaccines but not for other vaccine types, including more widely used mRNA preparations. Therefore, it is unlikely that GBS results from the cross-reactivity of antibodies against the SARS-CoV-2 spike protein generated after the COVID-19 vaccination. This paper outlines two hypotheses according to which increased risk of GBS following adenoviral vaccination is due to (1) generation of anti-vector antibodies that may cross-react with proteins involved in biological processes related to myelin and axons, or (2) neuroinvasion of selected adenovirus vectors to the peripheral nervous system, infection of neurons and subsequent inflammation and neuropathies. The rationale behind these hypotheses is outlined, advocating further epidemiological and experimental research to verify them. This is particularly important given the ongoing interest in using adenoviruses in developing vaccines against various infectious diseases and cancer immunotherapeutics.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
462
|
De Vito A, Colpani A, Trunfio M, Fiore V, Moi G, Fois M, Leoni N, Ruiu S, Babudieri S, Calcagno A, Madeddu G. Living with HIV and Getting Vaccinated: A Narrative Review. Vaccines (Basel) 2023; 11:896. [PMID: 37243000 PMCID: PMC10220625 DOI: 10.3390/vaccines11050896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
After 40 years of its appearance, human immunodeficiency virus (HIV) infection remains a leading public health challenge worldwide. Since the introduction of antiretroviral treatment (ART), HIV infection has become a chronic condition, and people living with HIV could have life expectancies close to those of the general population. People with HIV often have an increased risk of infection or experience more severe morbidity following exposure to vaccine-preventable diseases. Nowadays, several vaccines are available against bacteria and viruses. However, national and international vaccination guidelines for people with HIV are heterogeneous, and not every vaccine is included. For these reasons, we aimed to perform a narrative review about the vaccinations available for adults living with HIV, reporting the most updated studies performed for each vaccine among this population. We performed a comprehensive literature search through electronic databases (Pubmed-MEDLINE and Embase) and search engines (Google Scholar). We included English peer-reviewed publications (articles and reviews) on HIV and vaccination. Despite widespread use and guideline recommendations, few vaccine trials have been conducted in people with HIV. In addition, not all vaccines are recommended for people with HIV, especially for those with low CD4 cells count. Clinicians should carefully collect the history of vaccinations and patients' acceptance and preferences and regularly check the presence of antibodies for vaccine-preventable pathogens.
Collapse
Affiliation(s)
- Andrea De Vito
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Agnese Colpani
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Mattia Trunfio
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, 10149 Torino, Italy
| | - Vito Fiore
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Giulia Moi
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Marco Fois
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Nicola Leoni
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Stefano Ruiu
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Sergio Babudieri
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, 10149 Torino, Italy
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medicine, Surgery, and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (G.M.)
| |
Collapse
|
463
|
Boruah AP, Heydari K, Wapniarski AE, Caldwell M, Thakur KT. Neurological Considerations with COVID-19 Vaccinations. Semin Neurol 2023. [PMID: 37094803 DOI: 10.1055/s-0043-1767725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The benefits of coronavirus disease 2019 (COVID-19) vaccination significantly outweigh its risks on a public health scale, and vaccination has been crucial in controlling the spread of SARS-CoV-2. Nonetheless, several reports of adverse events following vaccination have been published.To summarize reports to date and assess the extent and quality of evidence regarding possible serious adverse neurological events following COVID-19 vaccination, focusing on Food and Drug Administration (FDA)-approved vaccines in the United States (BNT162b2, mRNA-1273, and Ad26.COV2.S).A review of literature from five major electronic databases (PubMed, Medline, Embase, Cochrane Library, and Google Scholar) was conducted between December 1, 2020 and June 5, 2022. Articles included in the review were systematic reviews and meta-analysis, cohort studies, retrospective studies, case-control studies, case series, and reports. Editorials, letters, and animal studies were excluded, since these studies did not include quantitative data regarding adverse side effects of vaccination in human subjects.Of 149 total articles and 97 (65%) were case reports or case series. Three phase 3 trials initially conducted for BNT162b2, MRNA-1273, and Ad26.COV2.S were included in the analysis.The amount and quality of evidence for possible neurological adverse events in the context of FDA-approved COVID-19 vaccinations is overall low tier. The current body of evidence continues to suggest that COVID-19 vaccinations have a high neurological safety profile; however, the risks and benefits of vaccination must continue to be closely monitored.
Collapse
Affiliation(s)
- Abhilasha P Boruah
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital (CUIMC/NYP), New York, NY
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kimia Heydari
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital (CUIMC/NYP), New York, NY
| | - Anne E Wapniarski
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital (CUIMC/NYP), New York, NY
| | - Marissa Caldwell
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital (CUIMC/NYP), New York, NY
| | - Kiran T Thakur
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital (CUIMC/NYP), New York, NY
| |
Collapse
|
464
|
Bürzle O, Menges D, Maier JD, Schams D, Puhan MA, Fehr J, Ballouz T, Frei A. Adverse effects, perceptions and attitudes related to BNT162b2, mRNA-1273 or JNJ-78436735 SARS-CoV-2 vaccines: Population-based cohort. NPJ Vaccines 2023; 8:61. [PMID: 37095137 PMCID: PMC10123463 DOI: 10.1038/s41541-023-00657-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Long-term control of SARS-CoV-2 requires effective vaccination strategies. This has been challenged by public mistrust and the spread of misinformation regarding vaccine safety. Better understanding and communication of the longer-term and comparative experiences of individuals in the general population following vaccination are required. In this population-based longitudinal study, we included 575 adults, randomly selected from all individuals presenting to a Swiss reference vaccination center, for receipt of BNT162b2, mRNA1273, or JNJ-78436735. We assessed the prevalence, onset, duration, and severity of self-reported adverse effects over 12 weeks following vaccination. We additionally evaluated participants' perceptions of vaccines, trust in public health authorities and pharmaceutical companies, and compliance with public health measures. Most participants reported at least one adverse effect within 12 weeks following vaccination. Adverse effects were mostly mild or moderate, resolved within three days, and rarely resulted in anaphylaxis or hospitalizations. Female sex, younger age, higher education, and receipt of mRNA-1273 were associated with reporting adverse effects. Compared to JNJ-78436735 recipients, a higher proportion of mRNA vaccine recipients agreed that vaccination is important, and trusted public health authorities. Our findings provide real-world estimates of the prevalence of adverse effects following SARS-CoV-2 vaccination and highlight the importance of transparent communication to ensure the success of current or future vaccination campaigns.
Collapse
Affiliation(s)
- Oliver Bürzle
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Julian D Maier
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Daniel Schams
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Milo A Puhan
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Jan Fehr
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.
| | - Tala Ballouz
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Anja Frei
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
465
|
León UAPD, Pérez AGC, Avila-Vales E. Modeling the SARS-CoV-2 Omicron variant dynamics in the United States with booster dose vaccination and waning immunity. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:10909-10953. [PMID: 37322966 DOI: 10.3934/mbe.2023484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We carried out a theoretical and numerical analysis for an epidemic model to analyze the dynamics of the SARS-CoV-2 Omicron variant and the impact of vaccination campaigns in the United States. The model proposed here includes asymptomatic and hospitalized compartments, vaccination with booster doses, and the waning of natural and vaccine-acquired immunity. We also consider the influence of face mask usage and efficiency. We found that enhancing booster doses and using N95 face masks are associated with a reduction in the number of new infections, hospitalizations and deaths. We highly recommend the use of surgical face masks as well, if usage of N95 is not a possibility due to the price range. Our simulations show that there might be two upcoming Omicron waves (in mid-2022 and late 2022), caused by natural and acquired immunity waning with respect to time. The magnitude of these waves will be 53% and 25% lower than the peak in January 2022, respectively. Hence, we recommend continuing to use face masks to decrease the peak of the upcoming COVID-19 waves.
Collapse
Affiliation(s)
- Ugo Avila-Ponce de León
- Programa de Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Angel G C Pérez
- Facultad de Matemáticas, Universidad Autónoma de Yucatán, Anillo Periférico Norte, Tablaje Catastral 13615, C.P. 97119, Mérida, Yucatán, Mexico
| | - Eric Avila-Vales
- Facultad de Matemáticas, Universidad Autónoma de Yucatán, Anillo Periférico Norte, Tablaje Catastral 13615, C.P. 97119, Mérida, Yucatán, Mexico
| |
Collapse
|
466
|
Zhou Z, Barrett J, He X. Immune Imprinting and Implications for COVID-19. Vaccines (Basel) 2023; 11:vaccines11040875. [PMID: 37112787 PMCID: PMC10142218 DOI: 10.3390/vaccines11040875] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Immunological memory is the key source of protective immunity against pathogens. At the current stage of the COVID-19 pandemic, heterologous combinations of exposure to viral antigens during infection and/or vaccination shape a distinctive immunological memory. Immune imprinting, the downside of memory, might limit the generation of de novo immune response against variant infection or the response to the next-generation vaccines. Here, we review mechanistic basis of immune imprinting by focusing on B cell immunobiology and discuss the extent to which immune imprinting is harmful, as well as its effect on SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Zhiqian Zhou
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Julia Barrett
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Xuan He
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
467
|
Yu J, Thomas PV, Sciacca M, Wu C, Liu J, He X, Miller J, Hachmann NP, Surve N, McMahan K, Jacob-Dolan C, Powers O, Hall K, Barrett J, Hope D, Mazurek CR, Murdza T, Chang WC, Golub E, Rees PA, Peterson CE, Hajduczki A, Chen WH, Martinez EJ, Hussin E, Lange C, Gong H, Matyas GR, Rao M, Suthar M, Boursiquot M, Cook A, Pessaint L, Lewis MG, Andersen H, Bolton DL, Michael NL, Joyce MG, Modjarrad K, Barouch DH. Ad26.COV2.S and SARS-CoV-2 spike protein ferritin nanoparticle vaccine protect against SARS-CoV-2 Omicron BA.5 challenge in macaques. Cell Rep Med 2023; 4:101018. [PMID: 37023746 PMCID: PMC10040355 DOI: 10.1016/j.xcrm.2023.101018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/13/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines demonstrate reduced protection against acquisition of BA.5 subvariant but are still effective against severe disease. However, immune correlates of protection against BA.5 remain unknown. We report the immunogenicity and protective efficacy of vaccine regimens consisting of the vector-based Ad26.COV2.S vaccine and the adjuvanted spike ferritin nanoparticle (SpFN) vaccine against a high-dose, mismatched Omicron BA.5 challenge in macaques. The SpFNx3 and Ad26 + SpFNx2 regimens elicit higher antibody responses than Ad26x3, whereas the Ad26 + SpFNx2 and Ad26x3 regimens induce higher CD8 T cell responses than SpFNx3. The Ad26 + SpFNx2 regimen elicits the highest CD4 T cell responses. All three regimens suppress peak and day 4 viral loads in the respiratory tract, which correlate with both humoral and cellular immune responses. This study demonstrates that both homologous and heterologous regimens involving Ad26.COV2.S and SpFN vaccines provide robust protection against a mismatched BA.5 challenge in macaques.
Collapse
Affiliation(s)
- Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Paul V Thomas
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michaela Sciacca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Cindy Wu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Xuan He
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jessica Miller
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicole P Hachmann
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nehalee Surve
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Olivia Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kevin Hall
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Julia Barrett
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Camille R Mazurek
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tetyana Murdza
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - William C Chang
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA
| | - Emily Golub
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Phyllis A Rees
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Caroline E Peterson
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Agnes Hajduczki
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Wei-Hung Chen
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Elizabeth J Martinez
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Elizabeth Hussin
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Camille Lange
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Hua Gong
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | | | | | | | | | | | - Diane L Bolton
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nelson L Michael
- Center for Infectious Diseases Research, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD 20910, USA
| | - M Gordon Joyce
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
468
|
Pergent M, Haerynck F, Hoste L, Gardulf A. COVID-19 vaccination in patients with primary immunodeficiencies: an international survey on patient vaccine hesitancy and self-reported adverse events. Front Immunol 2023; 14:1166198. [PMID: 37143673 PMCID: PMC10151802 DOI: 10.3389/fimmu.2023.1166198] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The Sars-CoV-2 pandemic caused great concern for this novel virus among patients with primary immunodeficiency (PID) or inborn errors of immunity (IEI) and their families. When COVID-19 vaccination program started, no data existed on adverse events (AEs) in this particular patient population, nor if patients felt hesitancy being vaccinated. Objectives To explore i) reasons for COVID-19 vaccination hesitancy, ii) the number and symptoms of AEs and their severity, durability and management. Method The organisations International Patient Organisation for Primary Immunodeficiencies (IPOPI), European Society for Immunodeficiencies (ESID) and International Nursing Group for Immunodeficiencies (INGID) distributed a global self-administered online survey. Results The survey was completed by 1317 patients (mean 47, range 12-100, years) from 40 countries. 41.7% of the patients denoted some hesitancy to COVID-19 vaccination, mainly having doubts about postvaccination protection related to their underlying PID and concerns about negative long-term effects. More women (22.6%) reported "very" or "pretty much" hesitancy compared to men (16.4%) (P<0.05). The most common systemic AEs were fatigue, muscle/body pain and headache, usually the same day or the day after the vaccination and lasting for 1-2 days. 27.8% of the respondents reported severe systemic AEs after any dose of COVID-19 vaccine. Only a minority (7.8%) of these patients visited a health-care professional and 20 patients (1.5%) were hospitalized or seen at emergency room without specifying subsequent admission at the hospital. Significantly more local and systemic AEs were reported after the second dose. No differences regarding AEs were observed across different PID subgroups or vaccine types. Conclusion At the time of the survey, almost half of the patients reported having felt hesitancy to COVID-19 vaccination highlighting the importance and need of developing joint international guidelines and education programs about COVID-19 vaccination. The types of AEs were comparable to healthy controls, but more frequent AEs were reported. Clinical studies and prospective, detailed registration of AEs related to COVID-19 vaccines in this patient population is of great importance. It is crucial to elucidate whether there is a coincidental or causal association between COVID-19 vaccine and some severe systemic AEs. Our data do not contradict that patients with PID can be advised to be vaccinated against COVID-19, in accordance with applicable national guidelines.
Collapse
Affiliation(s)
- Martine Pergent
- The International Patient Organisation for Primary Immunodeficiencies, Brussels, Belgium
| | - Filomeen Haerynck
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatric Pulmonology, Infectious Diseases and Immune Deficiency, Centre for Primary Immune Deficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
| | - Levi Hoste
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatric Pulmonology, Infectious Diseases and Immune Deficiency, Centre for Primary Immune Deficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
| | - Ann Gardulf
- Department of Clinical Immunology, John Radcliffe Hospital, The International Nursing Group for Immunodeficiencies (INGID), Oxford University Hospitals NHSFT, Oxford, United Kingdom
- Division of Clinical Immunology and Transfusion Medicine, The Unit for Clinical Research, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Faculty of Social and Health Sciences, Department of Health and Nursing Sciences, Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
469
|
Zhao D, Chen X, Wang L, Zhang J, Zhao Z, Yue N, Zhu Y, Fei W, Li X, Tan L, He W. Bidirectional and persistent immunomodulation of Astragalus polysaccharide as an adjuvant of influenza and recombinant SARS-CoV-2 vaccine. Int J Biol Macromol 2023; 234:123635. [PMID: 36801224 PMCID: PMC9932796 DOI: 10.1016/j.ijbiomac.2023.123635] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Respiratory viral infections, such as coronavirus disease of 2019 (COVID-19) and influenza, cause significant morbidity and mortality and have become a worldwide public health concern with tremendous economic and societal burdens. Vaccination is a major strategy for preventing infections. However, some new vaccines have an unmet need for impairing responses in certain individuals, especially COVID-19 vaccines, despite ongoing vaccine and adjuvant research. Here, we evaluated the effectiveness of Astragalus polysaccharide (APS), a bioactive polysaccharide extracted from the traditional Chinese herb Astragalus membranaceus as an immune adjuvant to regulate the efficacy of influenza split vaccine (ISV) and recombinant severe acute respiratory syndrome (SARS)-Cov-2 vaccine in mice. Our data indicated that APS as an adjuvant can facilitate the induction of high levels of hemagglutination inhibition (HAI) titer and specific antibody immunoglobulin G (IgG) and confer protection against the lethal challenge of influenza A viruses, including increased survival and amelioration of weight loss in mice immunized with the ISV. RNA sequencing (RNA-seq) analysis revealed that the NF-κB and Fc gamma R-mediated phagocytosis signaling pathways are essential for the immune response of mice immunized with the recombinant SARS-Cov-2 vaccine (RSV). Another important finding was that bidirectional immunomodulation of APS on cellular and humoral immunity was observed, and APS-adjuvant-induced antibodies persisted at a high level for at least 20 weeks. These findings suggest that APS is a potent adjuvant for influenza and COVID-19 vaccines, and has the advantages of bidirectional immunoregulation and persistent immunity.
Collapse
Affiliation(s)
- Danping Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuhong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhongpeng Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Na Yue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Fei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lingyun Tan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
470
|
Rao R, Musante CJ, Allen R. A quantitative systems pharmacology model of the pathophysiology and treatment of COVID-19 predicts optimal timing of pharmacological interventions. NPJ Syst Biol Appl 2023; 9:13. [PMID: 37059734 PMCID: PMC10102696 DOI: 10.1038/s41540-023-00269-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 02/09/2023] [Indexed: 04/16/2023] Open
Abstract
A quantitative systems pharmacology (QSP) model of the pathogenesis and treatment of SARS-CoV-2 infection can streamline and accelerate the development of novel medicines to treat COVID-19. Simulation of clinical trials allows in silico exploration of the uncertainties of clinical trial design and can rapidly inform their protocols. We previously published a preliminary model of the immune response to SARS-CoV-2 infection. To further our understanding of COVID-19 and treatment, we significantly updated the model by matching a curated dataset spanning viral load and immune responses in plasma and lung. We identified a population of parameter sets to generate heterogeneity in pathophysiology and treatment and tested this model against published reports from interventional SARS-CoV-2 targeting mAb and antiviral trials. Upon generation and selection of a virtual population, we match both the placebo and treated responses in viral load in these trials. We extended the model to predict the rate of hospitalization or death within a population. Via comparison of the in silico predictions with clinical data, we hypothesize that the immune response to virus is log-linear over a wide range of viral load. To validate this approach, we show the model matches a published subgroup analysis, sorted by baseline viral load, of patients treated with neutralizing Abs. By simulating intervention at different time points post infection, the model predicts efficacy is not sensitive to interventions within five days of symptom onset, but efficacy is dramatically reduced if more than five days pass post symptom onset prior to treatment.
Collapse
Affiliation(s)
- Rohit Rao
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA.
| | - Cynthia J Musante
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Richard Allen
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| |
Collapse
|
471
|
Khullar N, Bhatti JS, Singh S, Thukral B, Reddy PH, Bhatti GK. Insight into the liver dysfunction in COVID-19 patients: Molecular mechanisms and possible therapeutic strategies. World J Gastroenterol 2023; 29:2064-2077. [PMID: 37122601 PMCID: PMC10130970 DOI: 10.3748/wjg.v29.i14.2064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/23/2022] [Accepted: 03/21/2023] [Indexed: 04/13/2023] Open
Abstract
As of June 2022, more than 530 million people worldwide have become ill with coronavirus disease 2019 (COVID-19). Although COVID-19 is most commonly associated with respiratory distress (severe acute respiratory syndrome), meta-analysis have indicated that liver dysfunction also occurs in patients with severe symptoms. Current studies revealed distinctive patterning in the receptors on the hepatic cells that helps in viral invasion through the expression of angiotensin-converting enzyme receptors. It has also been reported that in some patients with COVID-19, therapeutic strategies, including repurposed drugs (mitifovir, lopinavir/ritonavir, tocilizumab, etc.) triggered liver injury and cholestatic toxicity. Several proven indicators support cytokine storm-induced hepatic damage. Because there are 1.5 billion patients with chronic liver disease worldwide, it becomes imperative to critically evaluate the molecular mechanisms concerning hepatotropism of COVID-19 and identify new potential therapeutics. This review also designated a comprehensive outlook of comorbidities and the impact of lifestyle and genetics in managing patients with COVID-19.
Collapse
Affiliation(s)
- Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib 140407, Punjab, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Satwinder Singh
- Department of Computer Science and Technology, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Bhawana Thukral
- Department of Nutrition and Dietetics, University Institute of Applied Health Sciences, Chandigarh University, Mohali 140413, Punjab, India
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali 140413, Punjab, India
| |
Collapse
|
472
|
Tan TJC, Mou Z, Lei R, Ouyang WO, Yuan M, Song G, Andrabi R, Wilson IA, Kieffer C, Dai X, Matreyek KA, Wu NC. High-throughput identification of prefusion-stabilizing mutations in SARS-CoV-2 spike. Nat Commun 2023; 14:2003. [PMID: 37037866 PMCID: PMC10086000 DOI: 10.1038/s41467-023-37786-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
Designing prefusion-stabilized SARS-CoV-2 spike is critical for the effectiveness of COVID-19 vaccines. All COVID-19 vaccines in the US encode spike with K986P/V987P mutations to stabilize its prefusion conformation. However, contemporary methods on engineering prefusion-stabilized spike immunogens involve tedious experimental work and heavily rely on structural information. Here, we establish a systematic and unbiased method of identifying mutations that concomitantly improve expression and stabilize the prefusion conformation of the SARS-CoV-2 spike. Our method integrates a fluorescence-based fusion assay, mammalian cell display technology, and deep mutational scanning. As a proof-of-concept, we apply this method to a region in the S2 domain that includes the first heptad repeat and central helix. Our results reveal that besides K986P and V987P, several mutations simultaneously improve expression and significantly lower the fusogenicity of the spike. As prefusion stabilization is a common challenge for viral immunogen design, this work will help accelerate vaccine development against different viruses.
Collapse
Affiliation(s)
- Timothy J C Tan
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Zongjun Mou
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Wenhao O Ouyang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Collin Kieffer
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xinghong Dai
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Kenneth A Matreyek
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Nicholas C Wu
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
473
|
Hashem M, El-Kassas M. Diagnosis, treatment protocols, and outcomes of liver transplant recipients infected with COVID-19. World J Clin Cases 2023; 11:2140-2159. [PMID: 37122505 PMCID: PMC10131019 DOI: 10.12998/wjcc.v11.i10.2140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/20/2023] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
Several cases of fatal pneumonia during November 2019 were linked initially to severe acute respiratory syndrome coronavirus 2, which the World Health Organization later designated as coronavirus disease 2019 (COVID-19). The World Health Organization declared COVID-19 as a pandemic on March 11, 2020. In the general population, COVID-19 severity can range from asymptomatic/mild symptoms to seriously ill. Its mortality rate could be as high as 49%. The Centers for Disease Control and Prevention have acknowledged that people with specific underlying medical conditions, among those who need immunosuppression after solid organ transplantation (SOT), are at an increased risk of developing severe illness from COVID-19. Liver transplantation is the second most prevalent SOT globally. Due to their immunosuppressed state, liver transplant (LT) recipients are more susceptible to serious infections. Therefore, comorbidities and prolonged immunosuppression among SOT recipients enhance the likelihood of severe COVID-19. It is crucial to comprehend the clinical picture, immunosuppressive management, prognosis, and prophylaxis of COVID-19 infection because it may pose a danger to transplant recipients. This review described the clinical and laboratory findings of COVID-19 in LT recipients and the risk factors for severe disease in this population group. In the following sections, we discussed current COVID-19 therapy choices, reviewed standard practice in modifying immunosuppressant regimens, and outlined the safety and efficacy of currently licensed drugs for inpatient and outpatient management. Additionally, we explored the clinical outcomes of COVID-19 in LT recipients and mentioned the efficacy and safety of vaccination use.
Collapse
Affiliation(s)
- Mai Hashem
- Fellow of Tropical Medicine and Gastroenterology, Assiut University Hospital, Assiut 71515, Egypt
| | - Mohamed El-Kassas
- Department of Endemic Medicine, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
474
|
Volosnikova EA, Merkuleva IA, Esina TI, Shcherbakov DN, Borgoyakova MB, Isaeva AA, Nesmeyanova VS, Volkova NV, Belenkaya SV, Zaykovskaya AV, Pyankov OV, Starostina EV, Zadorozhny AM, Zaitsev BN, Karpenko LI, Ilyichev AA, Danilenko ED. SARS-CoV-2 RBD Conjugated to Polyglucin, Spermidine, and dsRNA Elicits a Strong Immune Response in Mice. Vaccines (Basel) 2023; 11:vaccines11040808. [PMID: 37112720 PMCID: PMC10146165 DOI: 10.3390/vaccines11040808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the rapid development and approval of several COVID vaccines based on the full-length spike protein, there is a need for safe, potent, and high-volume vaccines. Considering the predominance of the production of neutralizing antibodies targeting the receptor-binding domain (RBD) of S-protein after natural infection or vaccination, it makes sense to choose RBD as a vaccine immunogen. However, due to its small size, RBD exhibits relatively poor immunogenicity. Searching for novel adjuvants for RBD-based vaccine formulations is considered a good strategy for enhancing its immunogenicity. Herein, we assess the immunogenicity of severe acute respiratory syndrome coronavirus 2 RBD conjugated to a polyglucin:spermidine complex (PGS) and dsRNA (RBD-PGS + dsRNA) in a mouse model. BALB/c mice were immunized intramuscularly twice, with a 2-week interval, with 50 µg of RBD, RBD with Al(OH)3, or conjugated RBD. A comparative analysis of serum RBD-specific IgG and neutralizing antibody titers showed that PGS, PGS + dsRNA, and Al(OH)3 enhanced the specific humoral response in animals. There was no significant difference between the groups immunized with RBD-PGS + dsRNA and RBD with Al(OH)3. Additionally, the study of the T-cell response in animals showed that, unlike adjuvants, the RBD-PGS + dsRNA conjugate stimulates the production of specific CD4+ and CD8+ T cells in animals.
Collapse
Affiliation(s)
- Ekaterina A Volosnikova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Iuliia A Merkuleva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Tatiana I Esina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Dmitry N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Mariya B Borgoyakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anastasiya A Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Valentina S Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Natalia V Volkova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Svetlana V Belenkaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anna V Zaykovskaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Oleg V Pyankov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Ekaterina V Starostina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexey M Zadorozhny
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Boris N Zaitsev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Larisa I Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexander A Ilyichev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Elena D Danilenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| |
Collapse
|
475
|
Horndler L, Delgado P, Romero-Pinedo S, Quesada M, Balabanov I, Laguna-Goya R, Almendro-Vázquez P, Llamas MA, Fresno M, Paz-Artal E, van Santen HM, Álvarez-Fernández S, Olmo A, Alarcón B. Decreased breadth of the antibody response to the spike protein of SARS-CoV-2 after repeated vaccination. Front Immunol 2023; 14:1157263. [PMID: 37081876 PMCID: PMC10111966 DOI: 10.3389/fimmu.2023.1157263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
Introduction The rapid development of vaccines to prevent COVID-19 has raised the need to compare the capacity of different vaccines in terms of developing a protective humoral response. Previous studies have shown inconsistent results in this area, highlighting the importance of further research to evaluate the efficacy of different vaccines. Methods This study utilized a highly sensitive and reliable flow cytometry method to measure the titers of IgG1 isotype antibodies in the blood of healthy volunteers after receiving one or two doses of various vaccines administered in Spain. The method was also used to simultaneously measure the reactivity of antibodies to the S protein of the original Wuhan strain and variants B.1.1.7 (Alpha), B.1.617.2 (Delta), and B.1.617.1 (Kappa). Results Significant differences were observed in the titer of anti-S antibodies produced after a first dose of the vaccines ChAdOx1 nCov-19/AstraZeneca, mRNA-1273/Moderna, BNT162b2/Pfizer-BioNTech, and Ad26.COV.S/Janssen. Furthermore, a relative reduction in the reactivity of the sera with the Alpha, Delta, and Kappa variants, compared to the Wuhan strain, was observed after the second boosting immunization. Discussion The findings of this study provide a comparison of different vaccines in terms of anti-S antibody generation and cast doubts on the convenience of repeated immunization with the same S protein sequence. The multiplexed capacity of the flow cytometry method utilized in this study allowed for a comprehensive evaluation of the efficacy of various vaccines in generating a protective humoral response. Future research could focus on the implications of these findings for the development of effective COVID-19 vaccination strategies.
Collapse
Affiliation(s)
- Lydia Horndler
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Pilar Delgado
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - Ivaylo Balabanov
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Rocío Laguna-Goya
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | | | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense de Madrid, Madrid, Spain
| | - Hisse M. van Santen
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
476
|
Nanomedicine for drug resistant pathogens and COVID-19 using mushroom nanocomposite inspired with bacteriocin – A Review. INORG CHEM COMMUN 2023; 152:110682. [PMID: 37041990 PMCID: PMC10067464 DOI: 10.1016/j.inoche.2023.110682] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Multidrug resistant (MDR) pathogens have become a major global health challenge and have severely threatened the health of society. Current conditions have gotten worse as a result of the COVID-19 pandemic, and infection rates in the future will rise. It is necessary to design, respond effectively, and take action to address these challenges by investigating new avenues. In this regard, the fabrication of metal NPs utilized by various methods, including green synthesis using mushroom, is highly versatile, cost-effective, eco-compatible, and superior. In contrast, biofabrication of metal NPs can be employed as a powerful weapon against MDR pathogens and have immense biomedical applications. In addition, the advancement in nanotechnology has made possible to modify the nanomaterials and enhance their activities. Metal NPs with biomolecules composite to prevents their microbial adhesion and kills the microbial pathogens through biofilm formation. Bacteriocin is an excellent antimicrobial peptide that works well as an augmentation substance to boost the antimicrobial effects. As a result, we concentrate on the creation of new, eco-compatible mycosynthesized metal NPs with bacteriocin nanocomposite via electrostatic, covalent, or non-covalent bindings. The synergistic benefits of metal NPs with bacteriocin to combat MDR pathogens and COVID-19, as well as other biomedical applications, are discussed in this review. Moreover, the importance of the adverse outcome pathway (AOP) in risk analysis of manufactured metal nanocomposite nanomaterial and their future possibilities also discussed.
Collapse
|
477
|
Takheaw N, Liwsrisakun C, Laopajon W, Pata S, Chaiwong W, Inchai J, Duangjit P, Pothirat C, Bumroongkit C, Deesomchok A, Theerakittikul T, Limsukon A, Tajarernmuang P, Niyatiwatchanchai N, Trongtrakul K, Kasinrerk W. Levels and durability of neutralizing antibodies against SARS-CoV-2 Omicron and other variants after ChAdOx-1 or BNT162b2 booster in CoronaVac-primed elderly individuals. Heliyon 2023; 9:e15653. [PMID: 37095993 PMCID: PMC10116116 DOI: 10.1016/j.heliyon.2023.e15653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023] Open
Abstract
The outbreak of the SARS-CoV-2 Omicron variant raised the need for vaccine boosting. We evaluated the efficiency of the third booster vaccine, ChAdOx-1 or BNT162b2, in causing a neutralizing antibody (NAb) response and its durability against the Omicron and other variants in elderly individuals previously vaccinated with 2-dose CoronaVac inactivated vaccine. After receiving 2-dose CoronaVac, only 2.2% of subjects had NAbs against the Omicron variant above the cut-off value. Four weeks after boosting, the number of subjects who had NAb levels above the cut-off values in the ChAdOx-1 and BNT162b2 vaccine boosting groups increased to 41.7% and 54.5%, respectively. However, after 12 and 24 weeks of boosting with any vaccines, NAb levels against the Omicron variant dramatically waned. Twenty-four weeks after boosting, only 2% had high levels of NAbs against the Omicron variant. Compared to other variants, the Omicron variant was less responsive to boosting vaccines. The waning rate of NAb levels for the Omicron variant was much faster than that observed in the Alpha, Beta and Delta variants. To combat the Omicron variant, the fourth booster dose is, therefore, recommended for elderly individuals.
Collapse
Affiliation(s)
- Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Chalerm Liwsrisakun
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Warawut Chaiwong
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Juthamas Inchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pilaiporn Duangjit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaicharn Pothirat
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiwat Bumroongkit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Athavudh Deesomchok
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Theerakorn Theerakittikul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Atikun Limsukon
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattraporn Tajarernmuang
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nutchanok Niyatiwatchanchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Konlawij Trongtrakul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
478
|
Jin J, Li J. CVSARRP: A framework to predict the risk of adverse to severe adverse reactions for 10855 diseases after COVID-19 vaccination. Heliyon 2023; 9:e14828. [PMID: 37009244 PMCID: PMC10041818 DOI: 10.1016/j.heliyon.2023.e14828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
COVID-19 vaccines greatly reduce the risk of infection with SARS-CoV-2. However, some people have adverse reactions after vaccination, and these can sometimes be severe. Gender, age, vaccines, and especially certain diseases histories are related to severe adverse reactions following COVID-19 vaccination. However, there are thousands of diseases and only some are known to be related to these severe adverse reactions. The risk of severe adverse reactions with other diseases remains unknown. Therefore, there is a need for predictive studies to provide improved medical care and minimize risk. Herein, we analyzed the statistical results of existing COVID-19 vaccine adverse reaction data and proposed a COVID-19 vaccine severe adverse reaction risk prediction method, named CVSARRP. The performance of the CVSARRP method was tested using the leave-one-out cross-validation approach. The correlation coefficient between the predicted and real risk is greater than 0.86. The CVSARRP method predicts the risk from adverse reactions to severe adverse reactions after COVID-19 vaccination for 10855 diseases. People with certain diseases, such as central nervous system diseases, heart diseases, urinary system disease, anemia, cancer, and respiratory tract disease, among others, may potentially have increased of severe adverse reactions following vaccination against COVID-19 and experiencing adverse events.
Collapse
Affiliation(s)
- Jiahuan Jin
- Research Center of Bioinformatics, Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang Province, China
| | - Jie Li
- Research Center of Bioinformatics, Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang Province, China
| |
Collapse
|
479
|
Quinn M, Parra-Rodriguez L, Alsoussi WB, Ayres C, Klebert MK, Liu C, Suessen T, Scheaffer SM, Middleton WD, Teefey SA, Powderly WG, Diamond MS, Presti RM, Ellebedy AH, Turner JS, O’Halloran JA, Mudd PA. Persons with HIV Develop Spike-Specific Lymph Node Germinal Center Responses following SARS-CoV-2 Vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:947-958. [PMID: 36779802 PMCID: PMC10038880 DOI: 10.4049/jimmunol.2200920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/24/2023] [Indexed: 02/14/2023]
Abstract
COVID-19 disproportionately affects persons with HIV (PWH) in worldwide locations with limited access to SARS-CoV-2 vaccines. PWH exhibit impaired immune responses to some, but not all, vaccines. Lymph node (LN) biopsies from PWH demonstrate abnormal LN structure, including dysregulated germinal center (GC) architecture. It is not clear whether LN dysregulation prevents PWH from mounting Ag-specific GC responses in the draining LN following vaccination. To address this issue, we longitudinally collected blood and draining LN fine needle aspiration samples before and after SARS-CoV-2 vaccination from a prospective, observational cohort of 11 PWH on antiretroviral therapy: 2 who received a two-dose mRNA vaccine series and 9 who received a single dose of the Ad26.COV2.S vaccine. Following vaccination, we observed spike-specific Abs, spike-specific B and T cells in the blood, and spike-specific GC B cell and T follicular helper cell responses in the LN of both mRNA vaccine recipients. We detected spike-specific Abs in the blood of all Ad26.COV2.S recipients, and one of six sampled Ad26.COV2.S recipients developed a detectable spike-specific GC B and T follicular helper cell response in the draining LN. Our data show that PWH can mount Ag-specific GC immune responses in the draining LN following SARS-CoV-2 vaccination. Due to the small and diverse nature of this cohort and the limited number of available controls, we are unable to elucidate all potential factors contributing to the infrequent vaccine-induced GC response observed in the Ad26.COV2.S recipients. Our preliminary findings suggest this is a necessary area of future research.
Collapse
Affiliation(s)
- Michael Quinn
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Luis Parra-Rodriguez
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Wafaa B. Alsoussi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Chapelle Ayres
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO
| | - Michael K. Klebert
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO
| | - Chang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Teresa Suessen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Suzanne M. Scheaffer
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - William D. Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Sharlene A. Teefey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - William G. Powderly
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Michael S. Diamond
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Rachel M. Presti
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Jane A. O’Halloran
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Philip A. Mudd
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
480
|
The safety and immunogenicity of inactivated COVID-19 vaccine in old pulmonary tuberculosis patients. Eur J Clin Microbiol Infect Dis 2023; 42:503-512. [PMID: 36849838 PMCID: PMC9970849 DOI: 10.1007/s10096-023-04566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
The immunogenicity and safety of vaccines against coronavirus disease 2019 (COVID-19) remain unknown in patients with a history of pulmonary tuberculosis (OPTB). Therefore, the safety and effectiveness of inactivated vaccines against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) were assessed in patients with a history of PTB. The study cohort included 106 healthy controls and 93 adult patients with OPTB who received a two-dose vaccination. The study period was 21 to 105 days. Concentrations of antibodies (Abs) against receptor-binding domain (RBD) IgG and SARS-CoV-2 neutralizing Abs (NAbs) were measured, in addition to the frequencies of SARS-CoV-2-specific B and a portion T cells. The incidence of adverse events was similar between the OPTB patients and healthy controls. No severe adverse events occurred. Concentrations of Abs against RBD-IgG and CoV-2 neutralizing Abs in addition to the frequencies of RBD-specific memory B cells proportions were lower in OPTB patients than the healthy controls (all, p < 0.05), while the frequencies of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4+) cells were higher (p = 0.023). There was no obvious correlation between age and blood concentrations of Abs against RBD-IgG and CoV-2 neutralizing Abs, while immune responses were similar in the fibrosis and calcification groups. The period of time following full-course vaccination and lymphocyte counts were associated to anti-RBD-IgG responses. Inactivated COVID-19 vaccinations were well tolerated in OPTB patients, although immunogenicity was limited in this population. This study has been registered at ClinicalTrials.gov (NCT05043246).
Collapse
|
481
|
Kolb JP, Hättich A, Strahl A, Rolvien T, Hennigs JK, Barg A, Frosch KH, Hartel MJ, Schlickewei C. Does the COVID-19 personal protective equipment impair the surgeon's performance? Arch Orthop Trauma Surg 2023; 143:1989-1997. [PMID: 35306584 PMCID: PMC8934057 DOI: 10.1007/s00402-022-04416-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Despite increasing vaccination rates, new viral variants of SARS-CoV-2 (severe acute respiratory syndrome coronavirus type 2) are advancing the COVID 19 (coronavirus disease 2019) pandemic and continue to challenge the entire world. Surgical care of SARS-CoV-2 positive patients requires special protective measures. We hypothesized that "COVID-19" personal protective equipment (PPE) during surgery of SARS-CoV-2 positive or potentially positive patients would negatively affect the surgeon and thus the surgical outcome. MATERIALS AND METHODS Ten experienced trauma surgeons participated in the study. Each surgeon performed two simulated surgeries of a distal tibial fracture on a Sawbone® under standardized conditions either wearing regular PPE or special COVID-19 PPE. Baseline values at rest were acquired for heart rate, blood pressure, saturation of peripheral oxygen (SpO2), respiratory rate and capillary blood gas (CBG) analysis including capillary partial pressure of oxygen (pO2) and carbon dioxide (pCO2), followed by four different standardized tests of attentional performance (TAP). Subsequently, the surgeon performed the first surgery according to a randomly determined order, with regular or COVID-19 PPE conditions in an operation theatre. After each surgery vital signs were acquired and CBG and TAP were performed again. RESULTS In our simulated surgical procedure heart rate, respiratory rate, systolic and diastolic blood pressure did not show relevant differences. Percutaneously measured SpO2 decreased with additional layers of PPE, while CBG parameters were not affected. TAP tests showed a significant impairment of attention if PPEs were compared to the baseline, but both PPEs had similar results and no meaningful differences could be measured. CONCLUSIONS According to our results, for surgical procedures additional PPE required during COVID-19 pandemic does not relevant affect the surgeon's mental and physical performance. Surgeries under COVID-19 PPE conditions appear safe and do not increase patient risk. LEVEL OF EVIDENCE Level I.
Collapse
Affiliation(s)
- Jan P Kolb
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Annika Hättich
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - André Strahl
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jan K Hennigs
- Department of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexej Barg
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Trauma Surgery, Orthopaedics and Sports Traumatology, BG Hospital Hamburg, Hamburg, Germany
- Department of Orthopaedics, University of Utah, Salt Lake City, USA
| | - Karl-Heinz Frosch
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Trauma Surgery, Orthopaedics and Sports Traumatology, BG Hospital Hamburg, Hamburg, Germany
| | - Maximilian J Hartel
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Trauma Surgery, Orthopaedics and Sports Traumatology, BG Hospital Hamburg, Hamburg, Germany
| | - Carsten Schlickewei
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
482
|
Yadegari H, Mohammadi M, Maghsood F, Ghorbani A, Bahadori T, Golsaz-Shirazi F, Zarnani AH, Salimi V, Jeddi-Tehrani M, Amiri MM, Shokri F. Diagnostic performance of a novel antigen-capture ELISA for the detection of SARS-CoV-2. Anal Biochem 2023; 666:115079. [PMID: 36754135 PMCID: PMC9902293 DOI: 10.1016/j.ab.2023.115079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/24/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND AND AIMS The coronavirus disease 2019 (COVID-19) pandemic is a serious health problem worldwide. Early virus detection is essential for disease control and management. Viral antigen detection by ELISA is a cost-effective, rapid, and accurate antigen diagnostic assay which could facilitate early viral detection. METHOD An antigen-capture sandwich ELISA was developed using novel nucleocapsid (NP)-specific mouse monoclonal antibodies (MAbs). The clinical performance of the assay was assessed using 403 positive and 150 negative respiratory samples collected during different SARS-CoV-2 variants outbreaks in Iran. RESULTS The limit of detection of our ELISA assay was found to be 43.3 pg/ml for recombinant NP. The overall sensitivity and specificity of this assay were 70.72% (95% CI: 66.01-75.12) and 100% (95% CI: 97.57-100), respectively, regardless of Ct values and SARS-CoV-2 variants. There was no significant difference in our assay sensitivity for the detection of Omicron subvariants compared to Delta variant. Assay sensitivity for the BA.5 Omicron subvariant was calculated as 91.89% (95% CI: 85.17-96.23) for samples with Ct values < 25 and 82.70% (95% CI: 75.19-88.71) for samples with Ct values < 30. CONCLUSION Our newly developed ELISA method is reasonably sensitive and highly specific for detection of SARS-CoV-2 regardless of the variants and subvariants of the virus.
Collapse
Affiliation(s)
- Hamidreza Yadegari
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohammadi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Maghsood
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Ghorbani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Tannaz Bahadori
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
483
|
Lutz CS, Hartman RM, Vigil DE, Britton A, Burrage AB, Campbell AP, Close RM, Desnoyers C, Dobson J, Garcia S, Halasa N, Honie E, Kobayashi M, McMorrow M, Mostafa HH, Parker D, Pohl K, Prill MM, Richards J, Roessler KC, Sutcliffe CG, Taylor K, Swango-Wilson A, Va P, Verani JR, Singleton RJ, Hammitt LL. Effectiveness of COVID-19 mRNA Vaccines in Preventing COVID-19-Associated Outpatient Visits and Hospitalizations Among American Indian and Alaska Native Persons, January-November 2021: A Test-Negative Case-Control Analysis Using Surveillance Data. Open Forum Infect Dis 2023; 10:ofad172. [PMID: 37089780 PMCID: PMC10114530 DOI: 10.1093/ofid/ofad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Background Despite the disproportionate morbidity and mortality experienced by American Indian and Alaska Native (AI/AN) persons during the coronavirus disease 2019 (COVID-19) pandemic, few studies have reported vaccine effectiveness (VE) estimates among these communities. Methods We conducted a test-negative case-control analysis among AI/AN persons aged ≥12 years presenting for care from January 1, 2021, through November 30, 2021, to evaluate the effectiveness of mRNA COVID-19 vaccines against COVID-19-associated outpatient visits and hospitalizations. Cases and controls were patients with ≥1 symptom consistent with COVID-19-like illness; cases were defined as those test-positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and controls were defined as those test-negative for SARS-CoV-2. We used unconditional multivariable logistic regression to estimate VE, defined as 1 minus the adjusted odds ratio for vaccination among cases vs controls. Results The analysis included 207 cases and 267 test-negative controls. Forty-four percent of cases and 78% of controls received 2 doses of either BNT162b2 or mRNA-1273 vaccine. VE point estimates for 2 doses of mRNA vaccine were higher for hospitalized participants (94.6%; 95% CI, 88.0-97.6) than outpatient participants (86.5%; 95% CI, 63.0-95.0), but confidence intervals overlapped. Conclusions Among AI/AN persons, mRNA COVID-19 vaccines were highly effective in preventing COVID-associated outpatient visits and hospitalizations. Maintaining high vaccine coverage, including booster doses, will reduce the burden of disease in this population.
Collapse
Affiliation(s)
- Chelsea S Lutz
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Rachel M Hartman
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Deionna E Vigil
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Amadea Britton
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | - Amanda B Burrage
- Tuba City Regional Health Care Corporation, Tuba
City, Arizona, USA
| | - Angela P Campbell
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | - Ryan M Close
- Whiteriver Service Unit, Phoenix Area, Indian Health Service,
Whiteriver, Arizona, USA
| | | | - Jennifer Dobson
- Alaska Native Tribal Health Consortium, Anchorage,
Alaska, USA
| | - Starla Garcia
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Natasha Halasa
- Vanderbilt University Medical Center, Nashville,
Tennessee, USA
| | - Elvira Honie
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Miwako Kobayashi
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | - Meredith McMorrow
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | - Heba H Mostafa
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Dennie Parker
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Kyle Pohl
- Alaska Native Tribal Health Consortium, Anchorage,
Alaska, USA
| | - Mila M Prill
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | - Jennifer Richards
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Kristen C Roessler
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | - Catherine G Sutcliffe
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public
Health, Baltimore, Maryland, USA
| | - Kim Taylor
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| | | | - Puthiery Va
- Chinle Service Unit, Navajo Area, Indian Health Service,
Chinle, Arizona, USA
| | - Jennifer R Verani
- CDC COVID-19 Response, Centers for Disease Control and
Prevention, Atlanta, Georgia, USA
| | | | - Laura L Hammitt
- Department of International Health, Johns Hopkins Bloomberg School of
Public Health, Baltimore, Maryland, USA
| |
Collapse
|
484
|
Fraser R, Orta-Resendiz A, Mazein A, Dockrell DH. Upper respiratory tract mucosal immunity for SARS-CoV-2 vaccines. Trends Mol Med 2023; 29:255-267. [PMID: 36764906 PMCID: PMC9868365 DOI: 10.1016/j.molmed.2023.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
SARS-CoV-2 vaccination significantly reduces morbidity and mortality, but has less impact on viral transmission rates, thus aiding viral evolution, and the longevity of vaccine-induced immunity rapidly declines. Immune responses in respiratory tract mucosal tissues are crucial for early control of infection, and can generate long-term antigen-specific protection with prompt recall responses. However, currently approved SARS-CoV-2 vaccines are not amenable to adequate respiratory mucosal delivery, particularly in the upper airways, which could account for the high vaccine breakthrough infection rates and limited duration of vaccine-mediated protection. In view of these drawbacks, we outline a strategy that has the potential to enhance both the efficacy and durability of existing SARS-CoV-2 vaccines, by inducing robust memory responses in the upper respiratory tract (URT) mucosa.
Collapse
Affiliation(s)
- Rupsha Fraser
- The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| | - Aurelio Orta-Resendiz
- Institut Pasteur, Université Paris Cité, HIV, Inflammation and Persistence Unit, F-75015 Paris, France
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - David H Dockrell
- The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
485
|
Sim JY, Kim SY, Kim EK. The incidence and clinical characteristics of myocarditis and pericarditis following mRNA-based COVID-19 vaccination in Republic of Korea adolescents from July 2021 to September 2022. Osong Public Health Res Perspect 2023; 14:76-88. [PMID: 37183328 PMCID: PMC10211448 DOI: 10.24171/j.phrp.2023.0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVES Age-specific information regarding myocarditis/pericarditis in adolescents following mRNA-based coronavirus disease 2019 (COVID-19) vaccination in Asia remains insufficient. This study investigated the incidence and clinical characteristics of myocarditis/pericarditis in Republic of Korea adolescents after mRNA-based COVID-19 vaccination. METHODS This retrospective descriptive study utilized patient data from the Korea Immunization Management System. Incidence rates were calculated according to age and sex. Clinical characteristics (symptoms/signs, laboratory values, and imaging results) were compared between mild and severe cases. RESULTS Between July 19, 2021 and September 30, 2022, 3,728,224 individuals aged 12 to 19 years received 6,484,165 mRNA-based COVID-19 vaccines, and 173 cases met the case definition for myocarditis/pericarditis: 151 mild (87.3%) and 22 severe (12.7%). The incidence was 3.8-fold higher in males than in females. Troponin I/ troponin T was elevated in 96% of myocarditis cases, demonstrating higher sensitivity than creatine kinase-myocardial band (67.6%) or C-reactive protein (75.2%). ST-segment or Twave on electrography abnormalities were found in 60.3% (85/141). Paroxysmal/sustained atrial/ventricular arrhythmias were more common in severe than in mild cases (45.5% vs. 16.8%, p=0.008). Edema on T2-weighted magnetic imaging occurred in 21.6% (8/37) and 62.5% (5/8) of mild and severe cases, respectively (p=0.03). Abnormal pericardial fluid collection or pericardial inflammation was found in 75.4% of pericarditis cases (49/65). CONCLUSION Myocarditis/pericarditis occurred in rare cases following mRNA-based COVID-19 vaccination. Most cases were mild, but the incidence was higher in adolescent males and after the second dose. As bivalent severe acute respiratory syndrome coronavirus 2 mRNA vaccination started in Republic of Korea in October 2022, the post-vaccination incidence of myocarditis/pericarditis should be closely monitored, considering clinical characteristics.
Collapse
Affiliation(s)
- Ju-Young Sim
- Division of Healthcare Associated Infection Control, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Seung-Yun Kim
- Adverse Event Investigation Team, COVID-19 Vaccination Task Force, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Eun-Kyoung Kim
- Division of Infectious Disease Control, Bureau of Infectious Disease Policy, Korea Disease Contrㅊol and Prevention Agency, Cheongju, Republic of Korea
| |
Collapse
|
486
|
Thookhamme C, Navinpipat M, Sasakul A, Pattarakosol P, Lertchaisataporn K, Tawinprai K, Praditsuktavorn P. Immunogenicity of the ChAdOx1 nCoV-19 vaccine in patients with hematologic malignancies. Clin Exp Vaccine Res 2023; 12:107-115. [PMID: 37214149 PMCID: PMC10193112 DOI: 10.7774/cevr.2023.12.2.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 01/19/2023] [Accepted: 03/31/2023] [Indexed: 05/24/2023] Open
Abstract
Purpose The present study aimed to study the immunogenicity of the ChAdOx1 nCoV-19 vaccine in patients with hematologic malignancies. Materials and Methods This prospective cohort study of hematology patients aimed to evaluate their antibody levels against the receptor-binding domain of the severe acute respiratory syndrome coronavirus 2 spike protein and seroconversion rates following two doses of the ChAdOx1 nCoV-19 vaccine. Between June and July 2021, we enrolled 61 patients and included 44 patients in our analysis. Antibody levels were assessed 8 and 4 weeks after the first and second injections, respectively, and compared with those of a healthy group. Results Eight weeks after the first dose, the geometric mean antibody level was 1.02 binding antibody units (BAU)/mL in the patient group and 37.91 BAU/mL in the healthy volunteer group (p<0.01). Four weeks after the second dose, the geometric mean antibody level was 9.44 BAU/mL in patients and 641.6 BAU/mL in healthy volunteers (p<0.01). The seroconversion rates 8 weeks after the first dose were 27.27% and 98.86% in the patient and healthy volunteer groups, respectively (p<0.001). The seroconversion rate 4 weeks after the second dose was 47.73% in patients and 100% in healthy volunteers. Factors leading to lower seroconversion rates were rituximab therapy (p=0.002), steroid therapy (p<0.001), and ongoing chemotherapy (p=0.048). Factors that decreased antibody levels were hematologic cancer (p<0.001), ongoing chemotherapy (p=0.004), rituximab (p<0.001), steroid use (p<0.001), and absolute lymphocyte count <1,000/mm3 (p=0.009). Conclusion Immune responses were impaired in individuals with hematologic malignancies, particularly patients undergoing ongoing therapy and B-cell-depleting therapy. Additional vaccinations should be considered for these patients, and further investigated.
Collapse
Affiliation(s)
| | | | - Aimwipa Sasakul
- Department of Hematology, Chulabhorn Hospital, Bangkok, Thailand
| | | | | | - Kriangkrai Tawinprai
- Infectious Disease Unit, Department of Medicine, Chulabhorn Hospital, Bangkok, Thailand
| | - Pannee Praditsuktavorn
- Department of Hematology, Chulabhorn Hospital, Bangkok, Thailand
- Faculty of Medicine, Princess Srisavangvadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
487
|
Firouzabadi N, Ghasemiyeh P, Moradishooli F, Mohammadi-Samani S. Update on the effectiveness of COVID-19 vaccines on different variants of SARS-CoV-2. Int Immunopharmacol 2023; 117:109968. [PMID: 37012880 PMCID: PMC9977625 DOI: 10.1016/j.intimp.2023.109968] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
It has been more than three years since the first emergence of coronavirus disease 2019 (COVID-19) and millions of lives have been taken to date. Like most pandemics caused by viral infections, massive public vaccination is the most promising approach to cease COVID-19 infection. In this regard, several vaccine platforms including inactivated virus, nucleic acid-based (mRNA and DNA vaccines), adenovirus-based, and protein-based vaccines have been designed and developed for COVID-19 prevention and many of them have received FDA or WHO approval. Fortunately, after global vaccination, the transmission rate, disease severity, and mortality rate of COVID-19 infection have diminished significantly. However, a rapid increase in COVID-19 cases due to the omicron variant in vaccinated countries has raised concerns about the effectiveness of these vaccines. In this review, articles published between January 2020 and January 2023 were reviewed using PubMed, Google Scholar, and Web of Science search engines with appropriate related keywords. The related papers were selected and discussed in detail. The current review mainly focuses on the effectiveness and safety of COVID-19 vaccines against SARS-CoV-2 variants. Along with discussing the available and approved vaccines, characteristics of different variants of COVID-19 have also been discussed in brief. Finally, the currently circulating COVID-19 variant i.e Omicron, along with the effectiveness of available COVID-19 vaccines against these new variants are discussed in detail. In conclusion, based on the available data, administration of newly developed bivalent mRNA COVID-19 vaccines, as booster shots, would be crucial to prevent further circulation of the newly developed variants.
Collapse
Affiliation(s)
- Negar Firouzabadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Ghasemiyeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moradishooli
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
488
|
Matson RP, Niesen MJM, Levy ER, Opp DN, Lenehan PJ, Donadio G, O'Horo JC, Venkatakrishnan AJ, Badley AD, Soundararajan V. Paediatric safety assessment of BNT162b2 vaccination in a multistate hospital-based electronic health record system in the USA: a retrospective analysis. Lancet Digit Health 2023; 5:e206-e216. [PMID: 36963910 DOI: 10.1016/s2589-7500(22)00253-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/10/2022] [Accepted: 12/21/2022] [Indexed: 03/26/2023]
Abstract
BACKGROUND The emergency use authorisation of BNT162b2 (tozinameran; Comirnaty, Pfizer-BioNTech) for children aged 5-17 years has resulted in rapid vaccination in the paediatric population. However, there are few studies of adverse events associated with vaccination in children. The aim of this study was to systematically assess the adverse events of two-dose BNT162b2 vaccination in the paediatric population. METHODS We conducted a retrospective analysis of patient electronic health records (EHRs) of children aged 5-17 years who received the primary two-dose series of the BNT162b2 vaccine between Jan 5, 2021, and Aug 5, 2022, at the Mayo Clinic Health System (MN, FL, AZ, IA, and WI), USA. Using natural language processing, we automatically curated adverse events reported by physicians in EHR clinical notes before and after vaccination. To determine significant adverse events after BNT162b2 vaccination, we calculated risk differences, which was defined as the percentage difference between the rate of children with an adverse event after a vaccine dose and the baseline rate of children with an adverse event before vaccination. 95% CIs and p values were calculated using the Miettinen and Nurminen score method. FINDINGS 56 436 individuals aged 5-17 years (20 227 aged 5-11 years and 36 209 aged 12-17 years) with EHRs in the Mayo Clinic Health Systems were included in the study. Overall, the reporting of adverse events remained low in passive surveillance. Serious adverse events were rare after the first and second doses of BNT162b2, with rates of anaphylaxis (six [0·01%] of 56 436), myocarditis (five [0·01%]), and pericarditis (three [0·01%]) consistent with previous studies. Among the 20 227 5-11-year-olds, there were increased risks of fatigue (58 after second dose vs 41 before first dose; risk difference [RD]dose2 0·08% [95% CI -0·01 to 0·18], p=0·044) and fever (104 after second dose vs 77 before first dose; RDdose2 0·13% [0·00 to 0·27], p=0·022) after the second dose. Among the 36 209 12-17-year-olds, there were increased risks of arthralgia (69 after second dose vs 48 before first dose; RDdose2 0·06% [-0·00 to 0·12], p=0·026), chills (58 after second dose vs 40 before first dose; RDdose2 0·05% [-0·00 to 0·11], p=0·034), and myalgia (96 after second dose vs 73 before first dose; RDdose2 0·06% [-0·01 to 0·14], p=0·038) after the second dose. Although the overall incidence was low, there was an increased risk of myocarditis in males aged 12-17 years after the second dose (five after second dose vs zero before first dose; RDdose2 0·03% [0·01 to 0·07], p=0·013), with median age being 15 years (IQR 14 to 16). INTERPRETATION Overall, this data suggests that vaccination with BNT162b2 in the paediatric population is generally safe and well-tolerated. Further research is warranted to investigate the basis for the increased risk of myocarditis in adolescent males. Additionally, further studies are needed to confirm whether the findings in our study population apply to the whole vaccinated paediatric population. FUNDING nference.
Collapse
Affiliation(s)
| | | | - Emily R Levy
- Division of Pediatric Infectious Diseases, Mayo Clinic, Rochester, MN, USA
| | - Derek N Opp
- Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - John C O'Horo
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Andrew D Badley
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
489
|
Hu Y, Wang Y, Shao T, Tang W, Hu K, Zhou Y, Miao L, Liu J, Wang B, Yu W. Safety and immunogenicity of heterologous ChAdOx1-nCoV19 and BNT162b2 vaccination: A meta-analysis of the heterologous COVID-19 vaccination outcomes. Vaccine 2023; 41:3003-3010. [PMID: 37037708 PMCID: PMC10060199 DOI: 10.1016/j.vaccine.2023.03.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 03/10/2023] [Accepted: 03/26/2023] [Indexed: 04/12/2023]
Abstract
INTRODUCTION Here, we systematically assessed the safety and immunogenicity of the heterologous ChAd/BNT vaccination regimens. MATERIALS AND METHODS We evaluated the immunogenicity by the geometric mean titers ratio (GMTR) of the neutralizing antibody and anti-spike IgG. The safety of heterologous ChAd/BNT vaccination was evaluated using the pooled risk ratios (RRs) calculated by the random-effects model about the adverse events. Our study was registered with PROSPERO, CRD42021265165. RESULTS Eleven studies were included in the analyses. Compared to the homologous ChAd/ChAd vaccination, the heterologous ChAd/BNT vaccination showed significantly higher immunogenicity in terms of the neutralizing antibody and GMTR of anti-spike IgG, but at the same time displayed higher incidence of total adverse reactions, especially for the local adverse reactions. Moreover, heterologous ChAd/BNT vaccination showed similar immunogenicity to the homologous BNT/BNT vaccination (GMTR of neutralizing antibody and anti-spike IgG) and similar safety. DISCUSSION Heterologous ChAd/BNT vaccination showed robust immunogenicity and tolerable safety.
Collapse
Affiliation(s)
- Yuxuan Hu
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yanning Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, PR China
| | - Taihang Shao
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing 211198, PR China
| | - Wenxi Tang
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing 211198, PR China
| | - Kerong Hu
- Department of Infectious Diseases, Huangshi Love&Health Hospital, Huangshi 435000, PR China
| | - Yujie Zhou
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing 210008, PR China
| | - Liyun Miao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing 210008, PR China
| | - Jing Liu
- Clinical Laboratory, Nanjing Yuhua Hospital (Yuhua Branch of Nanjing First Hospital), Nanjing 210039, PR China.
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, PR China.
| | - Wenying Yu
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
490
|
Martins M, do Nascimento GM, Conforti A, Noll JCG, Impellizeri JA, Sanchez E, Wagner B, Lione L, Salvatori E, Pinto E, Compagnone M, Viscount B, Hayward J, Shorrock C, Aurisicchio L, Diel DG. A linear SARS-CoV-2 DNA vaccine candidate reduces virus shedding in ferrets. Arch Virol 2023; 168:124. [PMID: 36988739 PMCID: PMC10052258 DOI: 10.1007/s00705-023-05746-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/03/2023] [Indexed: 03/30/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has caused more than 760 million cases and over 6.8 million deaths as of March 2023. Vaccination has been the main strategy used to contain the spread of the virus and to prevent hospitalizations and deaths. Currently, two mRNA-based vaccines and one adenovirus-vectored vaccine have been approved and are available for use in the U.S. population. The versatility, low cost, and rapid production of DNA vaccines provide important advantages over other platforms. Additionally, DNA vaccines efficiently induce both B- and T-cell responses by expressing the antigen within transfected host cells, and the antigen, after being processed into peptides, can associate with MHC class I or II of antigen-presenting cells (APCs) to stimulate different T cell responses. However, the efficiency of DNA vaccination needs to be improved for use in humans. Importantly, in vivo DNA delivery combined with electroporation (EP) has been used successfully in the field of veterinary oncology, resulting in high rates of response after electrochemotherapy. Here, we evaluate the safety, immunogenicity, and protective efficacy of a novel linear SARS-CoV-2 DNA vaccine candidate delivered by intramuscular injection followed by electroporation (Vet-ePorator™) in ferrets. The linear SARS-CoV-2 DNA vaccine candidate did not cause unexpected side effects. Additionally, the vaccine elicited neutralizing antibodies and T cell responses on day 42 post-immunization using a low dose of the linear DNA construct in a prime-boost regimen. Most importantly, vaccination significantly reduced shedding of infectious SARS-CoV-2 through oral and nasal secretions in a ferret model.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gabriela M do Nascimento
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Jessica C G Noll
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | | | | | - Brian Viscount
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - James Hayward
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Clay Shorrock
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Luigi Aurisicchio
- Takis Biotech, Rome, Italy
- Evvivax Biotech, Rome, Italy
- Neomatrix Biotech, Rome, Italy
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
491
|
Luo J, Zhang Z, Zhao S, Gao R. A Comparison of Etiology, Pathogenesis, Vaccinal and Antiviral Drug Development between Influenza and COVID-19. Int J Mol Sci 2023; 24:ijms24076369. [PMID: 37047339 PMCID: PMC10094131 DOI: 10.3390/ijms24076369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virus and coronavirus, two kinds of pathogens that exist widely in nature, are common emerging pathogens that cause respiratory tract infections in humans. In December 2019, a novel coronavirus SARS-CoV-2 emerged, causing a severe respiratory infection named COVID-19 in humans, and raising a global pandemic which has persisted in the world for almost three years. Influenza virus, a seasonally circulating respiratory pathogen, has caused four global pandemics in humans since 1918 by the emergence of novel variants. Studies have shown that there are certain similarities in transmission mode and pathogenesis between influenza and COVID-19, and vaccination and antiviral drugs are considered to have positive roles as well as several limitations in the prevention and control of both diseases. Comparative understandings would be helpful to the prevention and control of these diseases. Here, we review the study progress in the etiology, pathogenesis, vaccine and antiviral drug development for the two diseases.
Collapse
|
492
|
Cotter CA, Americo JL, Earl PL, Moss B. Protection from SARS-CoV-2 Variants by MVAs expressing matched or mismatched S administered intranasally to mice. NPJ Vaccines 2023; 8:47. [PMID: 36973267 PMCID: PMC10040904 DOI: 10.1038/s41541-023-00645-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
SARS-CoV-2 vaccines prevent severe disease but are less efficient in averting infection and transmission of variant strains, making it imperative to explore ways of enhancing protection. Use of inbred mice expressing the human SARS-CoV-2 receptor facilitates such investigations. We employed recombinant MVAs (rMVAs) expressing modified S of several SARS-CoV-2 strains and compared their ability to neutralize variants, bind S proteins and protect K18-hACE2 mice against SARS-CoV-2 challenge when administered intramuscularly or intranasally. The rMVAs expressing Wuhan, Beta and Delta S induced substantial cross neutralizing activities to each other but very low neutralization of Omicron; while rMVA expressing Omicon S induced neutralizing antibody predominanly to Omicron. In mice primed and boosted with rMVA expressing the Wuhan S, neutralizing antibodies to Wuhan increased after one immunization with rMVA expressing Omicron S due to original antigenic sin, but substantial neutralizing antibody to Omicron required a second immunization. Nevertheless, monovalent vaccines with S mismatched to the challenge virus still protected against severe disease and reduced the amounts of virus and subgenomic RNAs in the lungs and nasal turbinates, though not as well as vaccines with matched S. Passive transfer of Wuhan immune serum with Omicron S binding but undetectable neutralizing activity reduced infection of the l-ungs by Omicron suggesting additional effector functions. Notably, there was less infectious virus and viral subgenomic RNAs in the nasal turbinates and lungs when the rMVAs were administered intranasally rather than intramuscularly and this held true for vaccines that were matched or mismatched to the challenge strain of SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
493
|
Kumar R, Srivastava Y, Muthuramalingam P, Singh SK, Verma G, Tiwari S, Tandel N, Beura SK, Panigrahi AR, Maji S, Sharma P, Rai PK, Prajapati DK, Shin H, Tyagi RK. Understanding Mutations in Human SARS-CoV-2 Spike Glycoprotein: A Systematic Review & Meta-Analysis. Viruses 2023; 15:856. [PMID: 37112836 PMCID: PMC10142771 DOI: 10.3390/v15040856] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Genetic variant(s) of concern (VoC) of SARS-CoV-2 have been emerging worldwide due to mutations in the gene encoding spike glycoprotein. We performed comprehensive analyses of spike protein mutations in the significant variant clade of SARS-CoV-2, using the data available on the Nextstrain server. We selected various mutations, namely, A222V, N439K, N501Y, L452R, Y453F, E484K, K417N, T478K, L981F, L212I, N856K, T547K, G496S, and Y369C for this study. These mutations were chosen based on their global entropic score, emergence, spread, transmission, and their location in the spike receptor binding domain (RBD). The relative abundance of these mutations was mapped with global mutation D614G as a reference. Our analyses suggest the rapid emergence of newer global mutations alongside D614G, as reported during the recent waves of COVID-19 in various parts of the world. These mutations could be instrumentally imperative for the transmission, infectivity, virulence, and host immune system's evasion of SARS-CoV-2. The probable impact of these mutations on vaccine effectiveness, antigenic diversity, antibody interactions, protein stability, RBD flexibility, and accessibility to human cell receptor ACE2 was studied in silico. Overall, the present study can help researchers to design the next generation of vaccines and biotherapeutics to combat COVID-19 infection.
Collapse
Affiliation(s)
- Reetesh Kumar
- Faculty of Agricultural Sciences, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, India
- Department of Biotherapeutics, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Yogesh Srivastava
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pandiyan Muthuramalingam
- Division of Horticultural Science, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Sunil Kumar Singh
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Geetika Verma
- Department of Biotherapeutics, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Savitri Tiwari
- Division of Life Sciences, Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Gautam Buddha Nagar, Greater Noida 201310, India
| | - Nikunj Tandel
- Institute of Science, Nirma University, SG Highway, Gujarat 382481, India
| | - Samir Kumar Beura
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda 151401, India
| | | | - Somnath Maji
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Prakriti Sharma
- Biomedical Parasitology and Translational-Immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Pankaj Kumar Rai
- Department of Biotechnology, IIET, Invertis University, Bareilly 243001, India
| | | | - Hyunsuk Shin
- Division of Horticultural Science, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Rajeev K. Tyagi
- Biomedical Parasitology and Translational-Immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| |
Collapse
|
494
|
González-Sánchez ÓA, González-Ortiz LJ, Sánchez-Peña MJ, Gutiérrez-Pulido H, Cervantes O, Márquez-Sandoval F, Hernández-Bello J, Casillas N, Muñoz-Valle JF. A Novel Approach to Obtain Vaccine Effectiveness Continuous Profiles. Example Case: COVID-19 in Elderly Mexicans. Vaccines (Basel) 2023; 11:vaccines11040719. [PMID: 37112631 PMCID: PMC10142991 DOI: 10.3390/vaccines11040719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Population-wide vaccination is the most promising long-term COVID-19 disease management strategy. However, the protection offered by the currently available COVID-19 vaccines wanes over time, requiring boosters to be periodically given, which represents an unattainable challenge, especially if it is necessary to apply several doses per year. Therefore, it is essential to design strategies that contribute to maximizing the control of the pandemic with the available vaccines. Achieving this objective requires knowing, as precisely and accurately as possible, the changes in vaccine effectiveness over time in each population group, considering the eventual dependence on age, sex, etc. Thus, the present work proposes a novel approach to calculating realistic effectiveness profiles against symptomatic disease. In addition, this strategy can be adapted to estimate realistic effectiveness profiles against hospitalizations or deaths. All such time-dependent profiles allow the design of improved vaccination schedules, where each dose can be administrated to the population groups so that the fulfillment of the containment objectives is maximized. As a practical example for this analysis, vaccination against COVID-19 in Mexico was considered. However, this methodology can be applied to other countries' data or to characterize future vaccines with time-dependent effectiveness values. Since this strategy uses aggregated observational data collected from massive databases, assumptions about the data validity and the course of the studied epidemic could eventually be necessary.
Collapse
Affiliation(s)
- Óscar A González-Sánchez
- Division of Technologies for the Cyber-Human Integration, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Luis J González-Ortiz
- Department of Chemistry, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - M Judith Sánchez-Peña
- Department of Chemistry, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Humberto Gutiérrez-Pulido
- Department of Mathematics, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Oscar Cervantes
- Department of Chemistry, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Fabiola Márquez-Sandoval
- Department of Human Reproduction, Child Growth and Development, University Center of Health Sciences (CUCS), University of Guadalajara, Sierra Mojada 950, Col. Independencia, Guadalajara C.P. 44340, Jalisco, Mexico
| | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Sierra Mojada 950, Col. Independencia, Guadalajara C.P. 44340, Jalisco, Mexico
| | - Norberto Casillas
- Department of Chemistry, University Center of Exact Sciences and Engineering (CUCEI), University of Guadalajara, Marcelino García Barragán 1421, Col. Olímpica, Guadalajara C.P. 44430, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Sierra Mojada 950, Col. Independencia, Guadalajara C.P. 44340, Jalisco, Mexico
| |
Collapse
|
495
|
Wang SC, Rai CI, Chen YC. Challenges and Recent Advancements in COVID-19 Vaccines. Microorganisms 2023; 11:microorganisms11030787. [PMID: 36985360 PMCID: PMC10059828 DOI: 10.3390/microorganisms11030787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Vaccination is the most effective method for the prevention of COVID-19 caused by SARS-CoV-2, which is still a global epidemic. However, the evolution of SARS-CoV-2 is so rapid that various variants, including the Alpha, Beta, Gamma, Delta, and Omicron variants, have emerged, lowering the protection rate of vaccines and even resulting in breakthrough infections. Additionally, some rare but severe adverse reactions induced by COVID-19 vaccines may raise safety concerns and hinder vaccine promotion; however, clinical studies have shown that the benefits of vaccination outweigh the risks caused by adverse reactions. Current vaccines approved with emergency use authorization (EUA) were originally adaptive for adults only, and infants, children, and adolescents are not included. New-generation vaccines are needed to overcome the challenges of limited adaptive age population, breakthrough infection (mainly due to virus variant emergencies), and critical adverse reactions. Fortunately, some advances in COVID-19 vaccines have been obtained regarding enlarged adaptive populations for clinical applications, such as the Pfizer/BioNTech vaccine and the Moderna vaccine. In this article, we provide a review on the challenges and recent advancements in COVID-19 vaccines. The development of next-generation COVID-19 vaccines should lay emphasis on the expansion of adaptive age populations in all individuals, the induction of immune responses to viral variants, the avoidance or alleviation of rare but potentially critical adverse reactions, and the discovery of subunit vaccines with adjuvants encapsulated in nanoparticles.
Collapse
Affiliation(s)
- Shao-Cheng Wang
- Department of Psychiatric, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
- Department of Nurse-Midwifery and Women Health, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan
| | - Chung-I Rai
- Health Care Business Group, Foxconn Technology Co., Ltd., New Taipei City 23680, Taiwan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan
| | - Yuan-Chuan Chen
- Department of Nursing, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 35664, Taiwan
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 35664, Taiwan
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
496
|
Wang Y, Wang B, Zhao Z, Xu J, Zhang Z, Zhang J, Chen Y, Song X, Zheng W, Hou L, Wu S, Chen W. Effects of SARS-CoV-2 Omicron BA.1 Spike Mutations on T-Cell Epitopes in Mice. Viruses 2023; 15:763. [PMID: 36992472 PMCID: PMC10056712 DOI: 10.3390/v15030763] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
T-cell immunity plays an important role in the control of SARS-CoV-2 and has a great cross-protective effect on the variants. The Omicron BA.1 variant contains more than 30 mutations in the spike and severely evades humoral immunity. To understand how Omicron BA.1 spike mutations affect cellular immunity, the T-cell epitopes of SARS-CoV-2 wild-type and Omicron BA.1 spike in BALB/c (H-2d) and C57BL/6 mice (H-2b) were mapped through IFNγ ELISpot and intracellular cytokine staining assays. The epitopes were identified and verified in splenocytes from mice vaccinated with the adenovirus type 5 vector encoding the homologous spike, and the positive peptides involved in spike mutations were tested against wide-type and Omicron BA.1 vaccines. A total of eleven T-cell epitopes of wild-type and Omicron BA.1 spike were identified in BALB/c mice, and nine were identified in C57BL/6 mice, only two of which were CD4+ T-cell epitopes and most of which were CD8+ T-cell epitopes. The A67V and Del 69-70 mutations in Omicron BA.1 spike abolished one epitope in wild-type spike, and the T478K, E484A, Q493R, G496S and H655Y mutations resulted in three new epitopes in Omicron BA.1 spike, while the Y505H mutation did not affect the epitope. These data describe the difference of T-cell epitopes in SARS-CoV-2 wild-type and Omicron BA.1 spike in H-2b and H-2d mice, providing a better understanding of the effects of Omicron BA.1 spike mutations on cellular immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Shipo Wu
- Correspondence: (S.W.); (W.C.); Tel.: +86-10-66948692 (S.W.)
| | - Wei Chen
- Correspondence: (S.W.); (W.C.); Tel.: +86-10-66948692 (S.W.)
| |
Collapse
|
497
|
Wussow F, Kha M, Kim T, Ly M, Yll-Pico M, Kar S, Lewis MG, Chiuppesi F, Diamond DJ. Synthetic multiantigen MVA vaccine COH04S1 and variant-specific derivatives protect Syrian hamsters from SARS-CoV-2 Omicron subvariants. NPJ Vaccines 2023; 8:41. [PMID: 36928589 PMCID: PMC10018591 DOI: 10.1038/s41541-023-00640-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Emerging SARS-CoV-2 Omicron subvariants continue to disrupt COVID-19 vaccine efficacy through multiple immune mechanisms including neutralizing antibody evasion. We developed COH04S1, a synthetic modified vaccinia Ankara vector that co-expresses Wuhan-Hu-1-based spike and nucleocapsid antigens. COH04S1 demonstrated efficacy against ancestral virus and Beta and Delta variants in animal models and was safe and immunogenic in a Phase 1 clinical trial. Here, we report efficacy of COH04S1 and analogous Omicron BA.1- and Beta-specific vaccines to protect Syrian hamsters from Omicron subvariants. Despite eliciting strain-specific antibody responses, all three vaccines protect hamsters from weight loss, lower respiratory tract infection, and lung pathology following challenge with Omicron BA.1 or BA.2.12.1. While the BA.1-specifc vaccine affords consistently improved efficacy compared to COH04S1 to protect against homologous challenge with BA.1, all three vaccines confer similar protection against heterologous challenge with BA.2.12.1. These results demonstrate efficacy of COH04S1 and variant-specific derivatives to confer cross-protective immunity against SARS-CoV-2 Omicron subvariants.
Collapse
Affiliation(s)
- Felix Wussow
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| | - Mindy Kha
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Taehyun Kim
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Minh Ly
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Marcal Yll-Pico
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | | | | | - Flavia Chiuppesi
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Don J Diamond
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
498
|
Moyo-Gwete T, Richardson SI, Keeton R, Hermanus T, Spencer H, Manamela NP, Ayres F, Makhado Z, Motlou T, Tincho MB, Benede N, Ngomti A, Baguma R, Chauke MV, Mennen M, Adriaanse M, Skelem S, Goga A, Garrett N, Bekker LG, Gray G, Ntusi NA, Riou C, Burgers WA, Moore PL. Homologous Ad26.COV2.S vaccination results in reduced boosting of humoral responses in hybrid immunity, but elicits antibodies of similar magnitude regardless of prior infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.15.23287288. [PMID: 36993404 PMCID: PMC10055608 DOI: 10.1101/2023.03.15.23287288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The impact of previous SARS-CoV-2 infection on the durability of Ad26.COV2.S vaccine-elicited responses, and the effect of homologous boosting has not been well explored. We followed a cohort of healthcare workers for 6 months after receiving the Ad26.COV2.S vaccine and a further one month after they received an Ad26.COV2.S booster dose. We assessed longitudinal spike-specific antibody and T cell responses in individuals who had never had SARS-CoV-2 infection, compared to those who were infected with either the D614G or Beta variants prior to vaccination. Antibody and T cell responses elicited by the primary dose were durable against several variants of concern over the 6 month follow-up period, regardless of infection history. However, at 6 months after first vaccination, antibody binding, neutralization and ADCC were as much as 33-fold higher in individuals with hybrid immunity compared to those with no prior infection. Antibody cross-reactivity profiles of the previously infected groups were similar at 6 months, unlike at earlier time points suggesting that the effect of immune imprinting diminishes by 6 months. Importantly, an Ad26.COV2.S booster dose increased the magnitude of the antibody response in individuals with no prior infection to similar levels as those with previous infection. The magnitude of spike T cell responses and proportion of T cell responders remained stable after homologous boosting, concomitant with a significant increase in long-lived early differentiated CD4 memory T cells. Thus, these data highlight that multiple antigen exposures, whether through infection and vaccination or vaccination alone, result in similar boosts after Ad26.COV2.S vaccination.
Collapse
Affiliation(s)
- Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Simone I. Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Roanne Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Tandile Hermanus
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Holly Spencer
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Nelia P. Manamela
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Frances Ayres
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Zanele Makhado
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thopisang Motlou
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Marius B. Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Masego V. Chauke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Desmond Tutu HIV Centre, Cape Town, South Africa
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Ntobeko A.B. Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Penny L. Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
499
|
Feghali EJ, Challa A, Mahdi M, Acosta E, Jackson J. New-Onset Amyotrophic Lateral Sclerosis in a Patient who Received the J&J/Janssen COVID-19 Vaccine. Kans J Med 2023; 16:69-70. [PMID: 36970039 PMCID: PMC10035647 DOI: 10.17161/kjm.vol16.18969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/07/2023] [Indexed: 03/18/2023] Open
Affiliation(s)
- Elio Junior Feghali
- Department of Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, KS
| | - Abhiram Challa
- Department of Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, KS
| | - Mahmoud Mahdi
- Department of Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, KS
| | - Eric Acosta
- Department of Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, KS
| | - Jennifer Jackson
- Department of Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, KS
| |
Collapse
|
500
|
Zhou P, Song G, Liu H, Yuan M, He WT, Beutler N, Zhu X, Tse LV, Martinez DR, Schäfer A, Anzanello F, Yong P, Peng L, Dueker K, Musharrafieh R, Callaghan S, Capozzola T, Limbo O, Parren M, Garcia E, Rawlings SA, Smith DM, Nemazee D, Jardine JG, Safonova Y, Briney B, Rogers TF, Wilson IA, Baric RS, Gralinski LE, Burton DR, Andrabi R. Broadly neutralizing anti-S2 antibodies protect against all three human betacoronaviruses that cause deadly disease. Immunity 2023; 56:669-686.e7. [PMID: 36889306 PMCID: PMC9933850 DOI: 10.1016/j.immuni.2023.02.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/10/2022] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Pan-betacoronavirus neutralizing antibodies may hold the key to developing broadly protective vaccines against novel pandemic coronaviruses and to more effectively respond to SARS-CoV-2 variants. The emergence of Omicron and subvariants of SARS-CoV-2 illustrates the limitations of solely targeting the receptor-binding domain (RBD) of the spike (S) protein. Here, we isolated a large panel of broadly neutralizing antibodies (bnAbs) from SARS-CoV-2 recovered-vaccinated donors, which targets a conserved S2 region in the betacoronavirus spike fusion machinery. Select bnAbs showed broad in vivo protection against all three deadly betacoronaviruses, SARS-CoV-1, SARS-CoV-2, and MERS-CoV, which have spilled over into humans in the past two decades. Structural studies of these bnAbs delineated the molecular basis for their broad reactivity and revealed common antibody features targetable by broad vaccination strategies. These bnAbs provide new insights and opportunities for antibody-based interventions and for developing pan-betacoronavirus vaccines.
Collapse
Affiliation(s)
- Panpan Zhou
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wan-Ting He
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Longping V Tse
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R Martinez
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexandra Schäfer
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fabio Anzanello
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter Yong
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Katharina Dueker
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rami Musharrafieh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean Callaghan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tazio Capozzola
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Oliver Limbo
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mara Parren
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stephen A Rawlings
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Davey M Smith
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Joseph G Jardine
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bryan Briney
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas F Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ian A Wilson
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Ralph S Baric
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lisa E Gralinski
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA.
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|