1601
|
Martins-Filho PR, Ferreira LC, Heimfarth L, Araújo AADS, Quintans-Júnior LJ. Efficacy and safety of hydroxychloroquine as pre-and post-exposure prophylaxis and treatment of COVID-19: A systematic review and meta-analysis of blinded, placebo-controlled, randomized clinical trials. LANCET REGIONAL HEALTH. AMERICAS 2021; 2:100062. [PMID: 34485970 PMCID: PMC8403035 DOI: 10.1016/j.lana.2021.100062] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Hydroxychloroquine (HCQ) is an anti-malarial and immunomodulatory drug considered a potential candidate for drug repurposing in COVID-19 due to their in vitro antiviral activity against SARS-CoV-2. Despite the potential antiviral effects and anti-inflammatory profile, the results based on clinical studies are contradictory. Therefore, the quality of the decision-making process from meta-analyses summarizing the available evidence selecting studies with different designs and unblinded trials is limited. The aim of this study was to synthesize the best evidence on the efficacy and safety of HCQ as pre-and post-exposure prophylaxis and treatment of non-hospitalized and hospitalized patients with COVID-19. METHODS Searches were performed in PubMed, Web of Science, Embase, Lilacs, the website ClinicalTrials.gov and the preprint server medRxiv from January 1, 2020 to May 17, 2021. The following elements were used to define eligibility criteria: (1) Population: individuals at high-risk of exposure to SARS-CoV-2 (pre-exposure), individuals who had close contact with a positive or probable case of COVID-19 (post-exposure), non-hospitalized patients with COVID-19 and hospitalized patients with COVID-19; (2) Intervention: HCQ; (3) Comparison: placebo; (4) Outcomes: incidence of SARS-CoV-2 infection, need for hospitalization, length of hospital stay, need for invasive mechanical ventilation (MV), death, and adverse events; and (5) Study type: blinded, placebo-controlled, randomized clinical trials (RCTs). Risk of bias was judged according to the Cochrane guidelines for RCTs. Treatment effects were reported as relative risk (RR) for dichotomous variables and mean difference (MD) for continuous variables with 95% confidence intervals (CI). We used either a fixed or random-effects model to pool the results of individual studies depending on the presence of heterogeneity. The GRADE system was used to evaluate the strength of evidence between use of HCQ and the outcomes of interest. FINDINGS Fourteen blinded, placebo-controlled RCTs were included in this meta-analysis. Four trials (1942 patients: HCQ = 1271; placebo = 671) used HCQ as a prophylactic medication pre-exposure to COVID-19, two (1650 patients: HCQ = 821; placebo = 829) as a prophylactic medication post-exposure to COVID-19, three (1018 patients: HCQ = 497; placebo = 521) as treatment for non-hospitalized patients, and five (1138 patients: HCQ = 572; placebo = 566) as treatment for hospitalized patients with COVID-19. We found no decreased risk of SARS-CoV-2 infection among individuals receiving HCQ as pre-exposure (RR = 0.90; 95% CI 0.46 to 1.77) or post-exposure (RR = 0.96; 95% CI 0.72 to 1.29) prophylaxis to prevent COVID-19. There was no significant decreased risk of hospitalization for outpatients with SARS-CoV-2 infection (RR = 0.64; 95% CI 0.33 to 1.23) and no decreased risk of MV (RR = 0.81; 95% CI 0.49 to 1.34) and death (RR = 1.05; 95% CI 0.62 to 1.78) among hospitalized patients with COVID-19 receiving HCQ. The certainty of the results on the lack of clinical benefit for HCQ was rated as moderate. Moreover, our results demonstrated an increased risk for any adverse events and gastrointestinal symptoms among those using HCQ. INTERPRETATION Available evidence based on the results of blinded, placebo-controlled RCTs showed no clinical benefits of HCQ as pre-and post-exposure prophylaxis and treatment of non-hospitalized and hospitalized patients with COVID-19. FUNDING There was no funding source.
Collapse
Affiliation(s)
- Paulo Ricardo Martins-Filho
- Investigative Pathology Laboratory, Federal University of Sergipe, Aracaju, Sergipe, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Sergipe, Brazil,Corresponding author. Prof. Paulo Ricardo Martins-Filho. Universidade Federal de Sergipe, Hospital Universitário, Laboratório de Patologia Investigativa. Rua Cláudio Batista, s/n. Bairro Sanatório. Aracaju, Sergipe, Brasil. CEP
| | - Lis Campos Ferreira
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Sergipe, Brazil,Department of Medicine, Tiradentes University, Aracaju, Sergipe, Brazil
| | - Luana Heimfarth
- Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristovão, Sergipe, Brazil
| | - Adriano Antunes de Souza Araújo
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Sergipe, Brazil,Laboratory of Pharmaceutical Assays and Toxicity, Department of Pharmacy, Federal University of Sergipe, São Cristovão, Sergipe, Brazil
| | - Lucindo José Quintans-Júnior
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Sergipe, Brazil,Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristovão, Sergipe, Brazil
| |
Collapse
|
1602
|
Different dose regimens of a SARS-CoV-2 recombinant spike protein vaccine (NVX-CoV2373) in younger and older adults: A phase 2 randomized placebo-controlled trial. PLoS Med 2021; 18:e1003769. [PMID: 34597298 PMCID: PMC8486115 DOI: 10.1371/journal.pmed.1003769] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/13/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND NVX-CoV2373 is a recombinant severe acute respiratory coronavirus 2 (rSARS-CoV-2) nanoparticle vaccine composed of trimeric full-length SARS-CoV-2 spike glycoproteins and Matrix-M1 adjuvant. METHODS AND FINDINGS The phase 2 component of our randomized, placebo-controlled, phase 1 to 2 trial was designed to identify which dosing regimen of NVX-CoV2373 should move forward into late-phase studies and was based on immunogenicity and safety data through Day 35 (14 days after the second dose). The trial was conducted at 9 sites in Australia and 8 sites in the United States. Participants in 2 age groups (aged 18 to 59 and 60 to 84 years) were randomly assigned to receive either 1 or 2 intramuscular doses of 5-μg or 25-μg NVX-CoV2373 or placebo, 21 days apart. Primary endpoints were immunoglobulin G (IgG) anti-spike protein response, 7-day solicited reactogenicity, and unsolicited adverse events. A key secondary endpoint was wild-type virus neutralizing antibody response. After enrollment, 1,288 participants were randomly assigned to 1 of 4 vaccine groups or placebo, with 1,283 participants administered at least 1 study treatment. Of these, 45% were older participants 60 to 84 years. Reactogenicity was predominantly mild to moderate in severity and of short duration (median <3 days) after first and second vaccination with NVX-CoV2373, with higher frequencies and intensity after second vaccination and with the higher dose. Reactogenicity occurred less frequently and was of lower intensity in older participants. Both 2-dose regimens of 5-μg and 25-μg NVX-CoV2373 induced robust immune responses in younger and older participants. For the 2-dose regimen of 5 μg, geometric mean titers (GMTs) for IgG anti-spike protein were 65,019 (95% confidence interval (CI) 55,485 to 76,192) and 28,137 (95% CI 21,617 to 36,623) EU/mL and for wild-type virus neutralizing antibody (with an inhibitory concentration of 50%-MN50%) were 2,201 (95% CI 1,343 to 3,608) and 981 (95% CI 560 to 1,717) titers for younger and older participants, respectively, with seroconversion rates of 100% in both age groups. Neutralizing antibody responses exceeded those seen in a panel of convalescent sera for both age groups. Study limitations include the relatively short duration of safety follow-up to date and current lack of immune persistence data beyond the primary vaccination regimen time point assessments, but these data will accumulate over time. CONCLUSIONS The study confirmed the phase 1 findings that the 2-dose regimen of 5-μg NVX-CoV2373 is highly immunogenic and well tolerated in younger adults. In addition, in older adults, the 2-dose regimen of 5 μg was also well tolerated and showed sufficient immunogenicity to support its use in late-phase efficacy studies. TRIAL REGISTRATION ClinicalTrials.gov NCT04368988.
Collapse
|
1603
|
Corbett KS, Werner AP, Connell SO, Gagne M, Lai L, Moliva JI, Flynn B, Choi A, Koch M, Foulds KE, Andrew SF, Flebbe DR, Lamb E, Nurmukhambetova ST, Provost SJ, Bock KW, Minai M, Nagata BM, Ry AV, Flinchbaugh Z, Johnston TS, Mokhtari EB, Mudvari P, Henry AR, Laboune F, Chang B, Porto M, Wear J, Alvarado GS, Boyoglu-Barnum S, Todd JPM, Bart B, Cook A, Dodson A, Pessaint L, Steingrebe K, Elbashir S, Sriparna M, Pekosz A, Andersen H, Wu K, Edwards DK, Kar S, Lewis MG, Boritz E, Moore IN, Carfi A, Suthar MS, McDermott A, Roederer M, Nason MC, Sullivan NJ, Douek DC, Graham BS, Seder RA. mRNA-1273 protects against SARS-CoV-2 beta infection in nonhuman primates. Nat Immunol 2021; 22:1306-1315. [PMID: 34417590 PMCID: PMC8488000 DOI: 10.1038/s41590-021-01021-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
B.1.351 is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant most resistant to antibody neutralization. We demonstrate how the dose and number of immunizations influence protection. Nonhuman primates received two doses of 30 or 100 µg of Moderna's mRNA-1273 vaccine, a single immunization of 30 µg, or no vaccine. Two doses of 100 µg of mRNA-1273 induced 50% inhibitory reciprocal serum dilution neutralizing antibody titers against live SARS-CoV-2 p.Asp614Gly and B.1.351 of 3,300 and 240, respectively. Higher neutralizing responses against B.1.617.2 were also observed after two doses compared to a single dose. After challenge with B.1.351, there was ~4- to 5-log10 reduction of viral subgenomic RNA and low to undetectable replication in bronchoalveolar lavages in the two-dose vaccine groups, with a 1-log10 reduction in nasal swabs in the 100-µg group. These data establish that a two-dose regimen of mRNA-1273 will be critical for providing upper and lower airway protection against major variants of concern.
Collapse
Affiliation(s)
- Kizzmekia S Corbett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anne P Werner
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah O' Connell
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lilin Lai
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Juan I Moliva
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barbara Flynn
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Kathryn E Foulds
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shayne F Andrew
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dillon R Flebbe
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Evan Lamb
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Saule T Nurmukhambetova
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Samantha J Provost
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Timothy S Johnston
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Elham Bayat Mokhtari
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Prakriti Mudvari
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amy R Henry
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Farida Laboune
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Gabriela S Alvarado
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John-Paul M Todd
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Manjari Sriparna
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Pekosz
- Department of Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Kai Wu
- Moderna, Cambridge, MA, USA
| | | | | | | | - Eli Boritz
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Mehul S Suthar
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Adrian McDermott
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, MD, USA
| | - Nancy J Sullivan
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel C Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Robert A Seder
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
1604
|
Marco JJG, Pasquín MJÁ, Martín SM. Efectividad y seguridad de las vacunas para el SARS-CoV-2 actualmente disponibles. FMC : FORMACION MEDICA CONTINUADA EN ATENCION PRIMARIA 2021; 28:442-451. [PMID: 34611388 PMCID: PMC8483629 DOI: 10.1016/j.fmc.2021.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022]
Abstract
•Actualmente se están administrando cuatro vacunas en España con dos tecnologías diferentes: ARNm (Comirnaty y Moderna) y de vectores (Vaxzevria y Janssen). •Se habían administrado hasta junio de 2021 más de 38 millones de dosis y 15 millones de ciudadanos tenían la pauta vacunal completa. •Las vacunas han demostrado en los ensayos clínicos ser eficaces tanto para prevenir la infección como la hospitalización y el fallecimiento. •Los datos más concluyentes de su efectividad son en población mayor de 65 años ingresados en residencias. •Aún no disponemos de información para conocer la efectividad vacunal en otros segmentos poblacionales. •En general, las vacunas son seguras, aunque aparecen efectos adversos poco frecuentes, pero potencialmente graves, que requieren alta sospecha clínica y seguimiento: síndrome trombosis-trombocitopenia, síndrome de fuga capilar, miocarditis, cuadros anafilácticos, adenopatías. •Los sanitarios tienen un papel fundamental en la aceptabilidad y seguridad vacunal por la sospecha y notificación de posibles efectos adversos y la información dada a la ciudadanía.
Collapse
Affiliation(s)
- José Javier Gómez Marco
- Médico de familia, Miembro del Grupo de Infecciosas PAPPS-semFYC, CSU Las Calesas, SERMAS, Madrid, España
| | | | - Susana Martín Martín
- Médica de familia, Coordinadora del Grupo de Infecciosas PAPPS-semFYC, Centro de Salud Balmaseda, Balmaseda, Vizcaya, España
| |
Collapse
|
1605
|
Chiang TPY, Connolly CM, Ruddy JA, Boyarsky BJ, Alejo JL, Werbel WA, Massie A, Christopher-Stine L, Garonzik-Wang J, Segev DL, Paik JJ. Antibody response to the Janssen/Johnson & Johnson SARS-CoV-2 vaccine in patients with rheumatic and musculoskeletal diseases. Ann Rheum Dis 2021; 80:1365-1366. [PMID: 34429320 PMCID: PMC8440413 DOI: 10.1136/annrheumdis-2021-221145] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/16/2021] [Indexed: 11/04/2022]
Affiliation(s)
| | | | - Jake A Ruddy
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Brian J Boyarsky
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer L Alejo
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - William A Werbel
- Department of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| | - Allan Massie
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - Dorry L Segev
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Julie J Paik
- Division of Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
1606
|
Tregoning JS, Flight KE, Higham SL, Wang Z, Pierce BF. Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat Rev Immunol 2021; 21:626-636. [PMID: 34373623 PMCID: PMC8351583 DOI: 10.1038/s41577-021-00592-1] [Citation(s) in RCA: 746] [Impact Index Per Article: 186.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Where 2020 saw the development and testing of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at an unprecedented pace, the first half of 2021 has seen vaccine rollout in many countries. In this Progress article, we provide a snapshot of ongoing vaccine efficacy studies, as well as real-world data on vaccine effectiveness and the impact of virus variants of concern. Where they have been deployed in a high proportion of the adult population, the currently approved vaccines have been extremely effective in preventing COVID-19, particularly severe disease. Nonetheless, there are still significant challenges in ensuring equitable vaccine access around the globe and lessons that can be learned for controlling this pandemic and for the next pandemic.
Collapse
Affiliation(s)
- John S Tregoning
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK.
| | - Katie E Flight
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Sophie L Higham
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Ziyin Wang
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Benjamin F Pierce
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| |
Collapse
|
1607
|
Fathizadeh H, Afshar S, Masoudi MR, Gholizadeh P, Asgharzadeh M, Ganbarov K, Köse Ş, Yousefi M, Kafil HS. SARS-CoV-2 (Covid-19) vaccines structure, mechanisms and effectiveness: A review. Int J Biol Macromol 2021; 188:740-750. [PMID: 34403674 PMCID: PMC8364403 DOI: 10.1016/j.ijbiomac.2021.08.076] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022]
Abstract
The world has been suffering from COVID-19 disease for more than a year, and it still has a high mortality rate. In addition to the need to minimize transmission of the virus through non-pharmacological measures such as the use of masks and social distance, many efforts are being made to develop a variety of vaccines to prevent the disease worldwide. So far, several vaccines have reached the final stages of safety and efficacy in various phases of clinical trials, and some, such as Moderna/NIAID and BioNTech/Pfizer, have reported very high safety and protection. The important point is that comparing different vaccines is not easy because there is no set standard for measuring neutralization. In this study, we have reviewed the common platforms of COVID-19 vaccines and tried to present the latest reports on the effectiveness of these vaccines.
Collapse
Affiliation(s)
- Hadis Fathizadeh
- Department of laboratory sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Saman Afshar
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Mahmood Reza Masoudi
- Department of Internal Medicine, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Pourya Gholizadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Iran
| | | | | | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Iran.
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
1608
|
Abstract
Mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can enhance the spread and the infectiousness and decrease the protective effect of antibodies present after infection, vaccination or antibody treatment. The alpha variant (B.1.1.7), first seen in Kent/United Kingdom, has increased the R‑value and therefore the infectiousness by 75%; however, the effectiveness of the vaccines against SARS-CoV‑2 available in Germany seems to be only slightly impaired by these mutations. In the case of the beta variant (B.1.351), first described in South Africa, the neutralization ability of antibodies towards SARS-CoV‑2 is decreased. The monoclonal antibodies bamlanivimab and etesivimab, which are used therapeutically, are ineffective. The AstraZeneca vaccine offers almost no protection against mild or moderate disease caused by the beta variant. The gamma variant (P.1 or B.1.1.28.1), which was first found in Brazil, is probably 1.7-2.6 times more transmissible than previous virus strains circulating in Brazil. In addition to the infectiousness, the mortality risk of the gamma variant also seems to be increased between 1.2 and 1.9-fold in adults and between 5 and 8-fold in young persons. The delta variant (B.1.617), first described in India, is now dominant in most countries. It is 50% more infectious than the alpha variant, and the protective effect of vaccinations against symptomatic disease can be decreased (Biontech: delta variant 88%, alpha variant 93.7%; AstraZeneca: delta variant 67%, alpha variant 74.5%). Furthermore, the course of the disease with the delta variant is often more severe than with the wild type. Disease courses with the delta variant are less severe in vaccinated than in nonvaccinated persons, and fatal outcomes are substantially rarer. A high vaccination rate is essential in order to approach herd immunity and to bring the pandemic under control. Even where the protective effect towards mild or moderate disease is decreased, as a rule, vaccination still offers excellent protection against life-threatening and fatal disease courses.
Collapse
Affiliation(s)
- Christoph J Hemmer
- Abteilung für Tropenmedizin und Infektionskrankheiten, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 5, 18057, Rostock, Deutschland.
| | - M Löbermann
- Abteilung für Tropenmedizin und Infektionskrankheiten, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 5, 18057, Rostock, Deutschland
| | - E C Reisinger
- Abteilung für Tropenmedizin und Infektionskrankheiten, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 5, 18057, Rostock, Deutschland
| |
Collapse
|
1609
|
Friedman MA, Curtis JR, Winthrop KL. Impact of disease-modifying antirheumatic drugs on vaccine immunogenicity in patients with inflammatory rheumatic and musculoskeletal diseases. Ann Rheum Dis 2021; 80:1255-1265. [PMID: 34493491 PMCID: PMC8494475 DOI: 10.1136/annrheumdis-2021-221244] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022]
Abstract
Patients with rheumatic diseases are at increased risk of infectious complications; vaccinations are a critical component of their care. Disease-modifying antirheumatic drugs may reduce the immunogenicity of common vaccines. We will review here available data regarding the effect of these medications on influenza, pneumococcal, herpes zoster, SARS-CoV-2, hepatitis B, human papilloma virus and yellow fever vaccines. Rituximab has the most substantial impact on vaccine immunogenicity, which is most profound when vaccinations are given at shorter intervals after rituximab dosing. Methotrexate has less substantial effect but appears to adversely impact most vaccine immunogenicity. Abatacept likely decrease vaccine immunogenicity, although these studies are limited by the lack of adequate control groups. Janus kinase and tumour necrosis factor inhibitors decrease absolute antibody titres for many vaccines, but do not seem to significantly impact the proportions of patients achieving seroprotection. Other biologics (interleukin-6R (IL-6R), IL-12/IL-23 and IL-17 inhibitors) have little observed impact on vaccine immunogenicity. Data regarding the effect of these medications on the SARS-CoV-2 vaccine immunogenicity are just now emerging, and early glimpses appear similar to our experience with other vaccines. In this review, we summarise the most recent data regarding vaccine response and efficacy in this setting, particularly in light of current vaccination recommendations for immunocompromised patients.
Collapse
Affiliation(s)
| | - Jeffrey R Curtis
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham Department of Medicine, Birmingham, Alabama, USA
| | - Kevin L Winthrop
- Medicine, Oregon Health & Science University, Portland, Oregon, USA
- School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
1610
|
Shenai MB, Rahme R, Noorchashm H. Equivalency of Protection From Natural Immunity in COVID-19 Recovered Versus Fully Vaccinated Persons: A Systematic Review and Pooled Analysis. Cureus 2021; 13:e19102. [PMID: 34868754 PMCID: PMC8627252 DOI: 10.7759/cureus.19102] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
We present a systematic review and pooled analysis of clinical studies to date that (1) specifically compare the protection of natural immunity in the COVID-recovered versus the efficacy of complete vaccination in the COVID-naive, and (2) the added benefit of vaccination in the COVID-recovered, for prevention of subsequent SARS-CoV-2 infection. Using the PRISMA 2020 guidance, we first conducted a systematic review of available literature on PubMed, MedRxIV and FDA briefings to identify clinical studies either comparing COVID vaccination to natural immunity or delineating the benefit of vaccination in recovered individuals. After assessing eligibility, studies were qualitatively appraised and formally graded using the NOS system for observational, case-control and RCTs. Incidence rates were tabulated for the following groups: never infected (NI) and unvaccinated (UV), NI and vaccinated (V), previously infected (PI) and UV, PI and V. Pooling were performed by grouping the RCTs and observational studies separately, and then all studies in total. Risk ratios and differences are reported for individual studies and pooled groups, in 1) NPI/V vs PI/UV and 2) PI/UV vs PI/V analysis. In addition, the number needed to treat (NNT) analysis was performed for vaccination in naïve and previously infected cohorts. Nine clinical studies were identified, including three randomized controlled studies, four retrospective observational cohorts, one prospective observational cohort, and a case-control study. The NOS quality appraisals of these articles ranged from four to nine (out of nine stars). All of the included studies found at least statistical equivalence between the protection of full vaccination and natural immunity; and, three studies found superiority of natural immunity. Four observational studies found a statistically significant incremental benefit to vaccination in the COVID-recovered individuals. In a total pooled analysis, the incidence in NPI/V trended higher than PI/UV groups (RR=1.86 [95%CI 0.77-4.51], P=0.17). Vaccination in COVID-recovered individuals provided modest protection from reinfection (RR=1.82 [95%CI 1.21-2.73], P=0.004), but the absolute risk difference was extremely small (AR= 0.004 person-years [95% CI 0.001-0.007], P=0.02). The NNT to prevent one annual case of infection in COVID-recovered patients was 218, compared to 6.5 in COVID-naïve patients, representing a 33.5-fold difference in benefit between the two populations. COVID-recovered individuals represent a distinctly different benefit-risk calculus. While vaccinations are highly effective at protecting against infection and severe COVID-19 disease, our review demonstrates that natural immunity in COVID-recovered individuals is, at least, equivalent to the protection afforded by complete vaccination of COVID-naïve populations. There is a modest and incremental relative benefit to vaccination in COVID-recovered individuals; however, the net benefit is marginal on an absolute basis. Therefore, vaccination of COVID-recovered individuals should be subject to clinical equipoise and individual preference.
Collapse
Affiliation(s)
| | - Ralph Rahme
- Neurosurgery, St. Barnabas Hospital, New York City, USA
| | - Hooman Noorchashm
- Cardiac/Thoracic/Vascular Surgery • Immunology, American Patient Defense Union, Yardley, USA
| |
Collapse
|
1611
|
Flanagan KL, MacIntyre CR, McIntyre PB, Nelson MR. SARS-CoV-2 Vaccines: Where Are We Now? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2021; 9:3535-3543. [PMID: 34400116 PMCID: PMC8363243 DOI: 10.1016/j.jaip.2021.07.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022]
Abstract
The best and safest way to control the coronavirus disease 2019 (COVID-19) pandemic is by using vaccination to generate widespread immunity. The urgent need to develop safe and effective COVID-19 vaccines was met with unprecedented speed and action from the global community. There are now 289 vaccines in the development pipeline. More remarkably, there are 20 publicly available vaccines, and more than 3.3 billion doses of COVID-19 vaccines have been administered across 180 countries. This is just the beginning of our fight against the pandemic. Even at the current vaccination rate, it could take years to vaccinate the world's population; many high-income countries are focusing on their needs, whereas the poorer nations are waiting for vaccines. There is still much that we do not understand about immunity to this new disease, and we will have to contend with the emerging variants. In this commentary, we describe the current status of COVID-19 vaccine development and provide insights into how the development and approvals happened so quickly. We discuss the clinical trial data that led to rapid emergency use authorization and the many challenges of global rollout. We also comment on some of the key unanswered questions and future directions for COVID-19 vaccine development and deployment.
Collapse
Affiliation(s)
- Katie L Flanagan
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, Tas, Australia; School of Medicine, University of Tasmania, Launceston, Tas, Australia; School of Health and Biomedical Science, RMIT University, Bundoora, Vic, Australia; Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia.
| | - C Raina MacIntyre
- Biosecurity Research Program, Kirby Institute, UNSW Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Peter B McIntyre
- Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Michael R Nelson
- Division of Asthma, Allergy and Immunology, University of Virginia, Charlottesville, Va
| |
Collapse
|
1612
|
Saleem A, Akhtar MF, Haris M, Abdel-Daim MM. Recent updates on immunological, pharmacological, and alternative approaches to combat COVID-19. Inflammopharmacology 2021; 29:1331-1346. [PMID: 34331179 PMCID: PMC8324454 DOI: 10.1007/s10787-021-00850-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
The pandemic coronavirus disease 2019 (COVID-19) is instigated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that is mainly transmitted via the inhalation route and characterized by fever, coughing and shortness of breath. COVID-19 affects all age groups with no single cure. The drug discovery, manufacturing, and safety studies require extensive time and sources and, therefore, struggled to match the exponential spread of COVID-19. Yet, various repurposed drugs (antivirals, immune-modulators, nucleotide analogues), and convalescent plasma therapy have been authorized for emergency use against COVID-19 by Food and Drug Administration under certain limits and conditions. The discovery of vaccine is the biggest milestone achieved during the current pandemic era. About nine vaccines were developed for human use with varying claims of efficacy. The rapid emergence of mutations in SARS-CoV-2, suspected adverse drug reactions of current therapies in special population groups and limited availability of drugs in developing countries necessitate the development of more efficacious, safe and cheap drugs/vaccines for treatment and prevention of COVID-19. Keeping in view these limitations, the current review provides an update on the efficacy and safety of the repurposed, and natural drugs to treat COVID-19 as well as the vaccines used for its prophylaxis.
Collapse
Affiliation(s)
- Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan.
| | - Muhammad Haris
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| |
Collapse
|
1613
|
Giannella M, Pierrotti LC, Helanterä I, Manuel O. SARS-CoV-2 vaccination in solid-organ transplant recipients: What the clinician needs to know. Transpl Int 2021; 34:1776-1788. [PMID: 34450686 PMCID: PMC8646251 DOI: 10.1111/tri.14029] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022]
Abstract
In response to the COVID-19 pandemic, SARS-CoV-2 vaccines have been developed at an unparalleled speed, with 14 SARS-CoV-2 vaccines currently authorized. Solid-organ transplant (SOT) recipients are at risk for developing a higher rate of COVID-19-related complications and therefore they are at priority for immunization against SARS-CoV-2. Preliminary data suggest that although SARS-CoV-2 vaccines are safe in SOT recipients (with similar rate of adverse events than in the general population), the antibody responses are decreased in this population. Risk factors for poor vaccine immunogenicity include older age, shorter time from transplantation, use of mycophenolate and belatacept, and worse allograft function. SOT recipients should continue to be advised to maintain hand hygiene, use of facemasks, and social distancing after SARS-CoV-2 vaccine. Vaccination of household contacts should be also prioritized. Although highly encouraged for research purposes, systematic assessment in clinical practice of humoral and cellular immune responses after SARS-CoV-2 vaccination is controversial, since correlation between immunological findings and clinical protection from severe COVID-19, and cutoffs for protection are currently unknown in SOT recipients. Alternative immunization schemes, including a booster dose, higher doses, and modulation of immunosuppression during vaccination, need to be assessed in the context of well-designed clinical trials.
Collapse
Affiliation(s)
- Maddalena Giannella
- Infectious Diseases UnitDepartment of Medical and Surgical SciencesIRCCS Azienda Ospedaliero‐Universitaria di Bologna, Policlinico di Sant’OrsolaAlma Mater Studiorum University of BolognaBolognaItaly
| | - Lígia C. Pierrotti
- Department of Infectious DiseasesUniversity of São Paulo School of Medicine Hospital das ClínicasSão PauloBrazil
| | - Ilkka Helanterä
- Transplantation and Liver SurgeryHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Oriol Manuel
- Infectious Diseases Service and Transplantation CenterLausanne University HospitalLausanneSwitzerland
| |
Collapse
|
1614
|
Watson ME, Inagaki K, Weinberg JB. Severe Acute Respiratory Syndrome Coronavirus 2: Manifestations of Disease and Approaches to Treatment and Prevention in Humans. Comp Med 2021; 71:342-358. [PMID: 34535198 PMCID: PMC8594263 DOI: 10.30802/aalas-cm-21-000011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/08/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic was caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus has challenged civilization and modern science in ways that few infectious diseases and natural disasters have previously, causing globally significant human morbidity and mortality and triggering economic downturns across financial markets that will be dealt with for generations. Despite this, the pandemic has also brought an opportunity for humanity to come together and participate in a shared scientific investigation. Clinically, SARS-CoV-2 is associated with lower mortality rates than other recently emerged coronaviruses, such as SARS-CoV and the Middle East respiratory syndrome coronavirus (MERS-CoV). However, SARS-CoV-2 exhibits efficient human-to-human spread, with transmission often occurring before symptom recognition; this feature averts containment strategies that had worked previ- ously for SARS-CoV and MERS-CoV. Severe COVID-19 disease is characterized by dysregulated inflammatory responses associated with pulmonary congestion and intravascular coagulopathy leading to pneumonia, vascular insults, and multiorgan disease. Approaches to treatment have combined supportive care with antivirals, such as remdesivir, with immunomodulatory medications, including corticosteroids and cytokine-blocking antibody therapies; these treatments have advanced rapidly through clinical trials. Innovative approaches to vaccine development have facilitated rapid advances in design, testing, and distribution. Much remains to be learned about SARS-CoV-2 and COVID-19, and further biomedical research is necessary, including comparative medicine studies in animal models. This overview of COVID-19 in humans will highlight important aspects of disease, relevant pathophysiology, underlying immunology, and therapeutics that have been developed to date.
Collapse
Key Words
- ards, acute respiratory distress syndrome
- ace2, angiotensin-converting enzyme 2
- covid-19, coronavirus disease 2019
- hcov, human coronavirus
- ifn, interferon
- mers, middle east respiratory syndrome
- mis-c, multisystem inflammatory syndrome in children
- rbd, receptor binding domain
- sars, severe acute respiratory syndrome
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- s, spike
- tmprss2, type 2 transmembrane serine protease
Collapse
Affiliation(s)
- Michael E Watson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Kengo Inagaki
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Jason B Weinberg
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
1615
|
Almehdi AM, Khoder G, Alchakee AS, Alsayyid AT, Sarg NH, Soliman SSM. SARS-CoV-2 spike protein: pathogenesis, vaccines, and potential therapies. Infection 2021; 49:855-876. [PMID: 34339040 PMCID: PMC8326314 DOI: 10.1007/s15010-021-01677-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE COVID-19 pandemic has emerged as a result of infection by the deadly pathogenic severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causing enormous threats to humans. Coronaviruses are distinguished by a clove-like spike (S) protein, which plays a key role in viral pathogenesis, evolutions, and transmission. The objectives of this study are to investigate the distinctive structural features of SARS-CoV-2 S protein, its essential role in pathogenesis, and its use in the development of potential therapies and vaccines. METHODOLOGY A literature review was conducted to summarize, analyze, and interpret the available scientific data related to SARS-CoV-2 S protein in terms of characteristics, vaccines development and potential therapies. RESULTS The data indicate that S protein subunits and their variable conformational states significantly affect the virus pathogenesis, infectivity, and evolutionary mutation. A considerable number of potential natural and synthetic therapies were proposed based on S protein. Additionally, neutralizing antibodies were recently approved for emergency use. Furthermore, several vaccines utilizing the S protein were developed. CONCLUSION A better understanding of S protein features, structure and mutations facilitate the recognition of the importance of SARS-CoV-2 S protein in viral infection, as well as the development of therapies and vaccines. The efficacy and safety of these therapeutic compounds and vaccines are still controversial. However, they may potentially reduce or prevent SARS-CoV-2 infection, leading to a significant reduction of the global health burden of this pandemic.
Collapse
Affiliation(s)
- Ahmed M Almehdi
- College of Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Ghalia Khoder
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Aminah S Alchakee
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Azizeh T Alsayyid
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Nadin H Sarg
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Sameh S M Soliman
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| |
Collapse
|
1616
|
Bouland AJ, March JA. Invited Editorial: Characteristics and outcomes of adverse events following COVID-19 vaccination. J Am Coll Emerg Physicians Open 2021; 2:e12566. [PMID: 34611651 PMCID: PMC8478148 DOI: 10.1002/emp2.12566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Andrew J. Bouland
- Department of Emergency MedicineDivision of EMS, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Juan Alberto March
- Department of Emergency MedicineDivision of EMS, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| |
Collapse
|
1617
|
Connolly CM, Koenig D, Ravi SN, Azar A, Kant S, Dalal M, Duchen J, Seo P, Antiochos B, Paik JJ, Geetha D. Correspondence on "SARS-CoV-2 vaccination in rituximab-treated patients: evidence for impaired humoral but inducible cellular immune response" by Bonelli et al. Ann Rheum Dis 2021; 80:e164. [PMID: 34340978 DOI: 10.1136/annrheumdis-2021-220972] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 01/13/2023]
Affiliation(s)
| | - Darya Koenig
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Srekar N Ravi
- Department of Medicine, Florida Atlantic University-Treasure Coast Campus, Boca Raton, Florida, USA
| | - Antoine Azar
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sam Kant
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Monika Dalal
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jessica Duchen
- Biostatistics, Epidemiology and Data Management, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Philip Seo
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Brendan Antiochos
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Julie J Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Duvuru Geetha
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
1618
|
Patel N, Bouchard J, Oliver MB, Badowski ME, Carreno JJ, the Society of Infectious Diseases Pharmacists. Early clinical trial data and real-world assessment of COVID-19 vaccines: Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2021; 41:837-850. [PMID: 34689348 PMCID: PMC8662264 DOI: 10.1002/phar.2622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/19/2023]
Abstract
As of August 2021, there were three COVID-19 vaccines available in the United States for the prevention of coronavirus 2019 (COVID-19). The purpose of this narrative review is to examine the early experience from the Emergency Use Authorization (EUA) of BNT162b2 (Pfizer, Inc./BioNTech), mRNA-1273 (Moderna, Inc.), and Ad26.COV2.S (Johnson and Johnson/Janssen Global Services, LLC) through July 2021. The EUA data from the clinical trials have largely been corroborated by real-world effectiveness investigations post-authorization. These studies indicate that immunity is obtained within 2 weeks post-vaccination and may endure for 6 months. The immunity conferred by the vaccines may also be effective against SARS-CoV-2 variants of concern. Additionally, populations not included in the emergency use authorization studies may also benefit from vaccination. This look back at the initial clinical experience can be used by the global community to inform and develop COVID-19 vaccine programs.
Collapse
Affiliation(s)
- Nimish Patel
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | | | - Meredith B. Oliver
- M Health Fairview University of Minnesota Masonic Children's HospitalMinneapolisMinnesotaUSA
| | | | | | | |
Collapse
|
1619
|
Diotallevi F, Campanati A, Radi G, Martina E, Rizzetto G, Barbadoro P, D'Errico MM, Offidani A. Vaccines Against SARS-CoV-2 in Psoriasis Patients on Immunosuppressive Therapy: Implications of Vaccination Nationwide Campaign on Clinical Practice in Italy. Dermatol Ther (Heidelb) 2021; 11:1889-1903. [PMID: 34586598 PMCID: PMC8480269 DOI: 10.1007/s13555-021-00610-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 01/08/2023] Open
Abstract
More than 12 months have passed since the World Health Organization (WHO) declared Coronavirus Disease 19 (COVID-19), caused by the SARS-CoV2 virus, to be a pandemic on 11 March 2020. The entire world scientific community agrees that at this time vaccine is the most promising weapon to combat the infection and the severity of the disease. According to the document “Draft landscape of COVID-19 candidate vaccines” by WHO, 272 vaccines against SARS-CoV-2 virus are in development, although only four of these, produced by Pfizer-BioNTech (Pfizer, Inc. and BioNTech), Moderna, AstraZeneca, and Janssen companies, respectively, have been approved by European Medicines Agency and Italian Medicines Agency and subsequently distributed nationwide for use. These vaccines are the result of highly innovative procedures and are quite different from each other in terms of composition. Even clinicians in various medical fields may be unfamiliar with the effects of these vaccines. There is the strong emerging need for dermatologists to understand the crucial role of vaccines, with a focus on the need to vaccinate patients suffering from immune-mediated skin diseases, such as psoriasis, while taking the ongoing treatment into consideration regarding the timing of vaccination. Similarly, psoriasis patients aware of having an immune-mediated and inflammatory disease are increasingly asking the dermatologist information about the efficacy and safety of vaccines against SARS-CoV-2 virus. In this narrative review of the literature and critical analysis of the recommendations of the Italian Ministry of Health, we analyze the implications of the vaccination campaign on dermatological patients with psoriasis undergoing immunosuppressive treatment.
Collapse
Affiliation(s)
- Federico Diotallevi
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Anna Campanati
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Giulia Radi
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy.
| | - Emanuela Martina
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Giulio Rizzetto
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Pamela Barbadoro
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, Ancona, Italy
| | - Marcello Mario D'Errico
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, Ancona, Italy
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| |
Collapse
|
1620
|
Ding M, Dong X, Sun YL, Sokolowska M, Akdis M, van de Veen W, Azkur AK, Azkur D, Akdis CA, Gao YD. Recent advances and developments in COVID-19 in the context of allergic diseases. Clin Transl Allergy 2021; 11:e12065. [PMID: 34582102 PMCID: PMC8646609 DOI: 10.1002/clt2.12065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Since the first reports of coronavirus disease 2019 (COVID-19) in Wuhan, China, in December 2019, there have been 198 million confirmed cases worldwide as of August 2021. The scientific community has joined efforts to gain knowledge of the newly emerged virus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the immunopathological mechanisms leading to COVID-19, and its significance for patients with allergies and asthma. METHODS Based on the current literature, recent advances and developments in COVID-19 in the context of allergic diseases were reviewed. RESULTS AND CONCLUSIONS In this review, we discuss the prevalence of COVID-19 in subjects with asthma, attacks of hereditary angioedema, and other allergic diseases during COVID-19. Underlying mechanisms suggest a protective role of allergy in COVID-19, involving eosinophilia, SARS-CoV-2 receptors expression, interferon responses, and other immunological events, but further studies are needed to fully understand those associations. There has been significant progress in disease evaluation and management of COVID-19, and allergy care should continue during the COVID-19 pandemic. The European Academy of Allergy & Clinical Immunology (EAACI) launched a series of statements and position papers providing recommendations on the organization of the allergy clinic, handling of allergen immunotherapy, asthma, drug hypersensitivity, allergic rhinitis, and other allergic diseases. Treatment of allergies using biologics during the COVID-19 pandemic has also been discussed. Allergic reactions to the COVID-19 vaccines, including severe anaphylaxis, have been reported. Vaccination is a prophylactic strategy that can lead to a significant reduction in the mortality and morbidity associated with SARS-CoV-2 infection, and in this review, we discuss the proposed culprit components causing rare adverse reactions and recommendations to mitigate the risk of anaphylactic events during the administration of the vaccines.
Collapse
Affiliation(s)
- Mei Ding
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Xiang Dong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Yuan-Li Sun
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ahmet Kursat Azkur
- Department of Virology, Faculty of Veterinary Medicine, University of Kirikkale, Kirikkale, Turkey
| | - Dilek Azkur
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, University of Kirikkale, Kirikkale, Turkey
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Ya-Dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| |
Collapse
|
1621
|
Daian e Silva DSDO, da Fonseca FG. The Rise of Vectored Vaccines: A Legacy of the COVID-19 Global Crisis. Vaccines (Basel) 2021; 9:1101. [PMID: 34696209 PMCID: PMC8538930 DOI: 10.3390/vaccines9101101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 pandemic represents a milestone in vaccine research and development in a global context. A worldwide effort, as never seen before, involved scientists from all over the world in favor of the fast, accurate and precise construction and testing of immunogens against the new coronavirus, SARS-CoV-2. Among all the vaccine strategies put into play for study and validation, those based on recombinant viral vectors gained special attention due to their effectiveness, ease of production and the amplitude of the triggered immune responses. Some of these new vaccines have already been approved for emergency/full use, while others are still in pre- and clinical trials. In this article we will highlight what is behind adeno-associated vectors, such as those presented by the immunogens ChaAdOx1, Sputnik, Convidecia (CanSino, Tianjin, China), and Janssen (Johnson & Johnson, New Jersey, EUA), in addition to other promising platforms such as Vaccinia virus MVA, influenza virus, and measles virus, among others.
Collapse
Affiliation(s)
- Danielle Soares de Oliveira Daian e Silva
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- CT Vacinas, BH-TEC Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31310-260, Brazil
| |
Collapse
|
1622
|
Nestić D, Božinović K, Pehar I, Wallace R, Parker AL, Majhen D. The Revolving Door of Adenovirus Cell Entry: Not All Pathways Are Equal. Pharmaceutics 2021; 13:1585. [PMID: 34683878 PMCID: PMC8540258 DOI: 10.3390/pharmaceutics13101585] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Adenoviruses represent exceptional candidates for wide-ranging therapeutic applications, from vectors for gene therapy to oncolytics for cancer treatments. The first ever commercial gene therapy medicine was based on a recombinant adenovirus vector, while most recently, adenoviral vectors have proven critical as vaccine platforms in effectively controlling the global coronavirus pandemic. Here, we discuss factors involved in adenovirus cell binding, entry, and trafficking; how they influence efficiency of adenovirus-based vectors; and how they can be manipulated to enhance efficacy of genetically modified adenoviral variants. We focus particularly on endocytosis and how different adenovirus serotypes employ different endocytic pathways to gain cell entry, and thus, have different intracellular trafficking pathways that subsequently trigger different host antiviral responses. In the context of gene therapy, the final goal of the adenovirus vector is to efficiently deliver therapeutic transgenes into the target cell nucleus, thus allowing its functional expression. Aberrant or inefficient endocytosis can impede this goal, therefore, it should be considered when designing and constructing adenovirus-based vectors.
Collapse
Affiliation(s)
- Davor Nestić
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Ksenija Božinović
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Isabela Pehar
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Rebecca Wallace
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (R.W.); (A.L.P.)
| | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (R.W.); (A.L.P.)
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| |
Collapse
|
1623
|
Witek TJ. How the Global COVID-19 Pandemic Brought Drug and Vaccine Development into the Public Mainstream. Pharmaceut Med 2021; 35:287-295. [PMID: 34580837 PMCID: PMC8475305 DOI: 10.1007/s40290-021-00402-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 11/24/2022]
Abstract
Pharmaceutical development was at the forefront of efforts to prevent infection with the SARS-CoV-2 virus as well as to treat its often-devastating effects. Drug development, and its multifaceted and multi-disciplined activity toward effective vaccines and drugs, became part of everyday news. I review several key areas of vaccine and drug development that were brought into the public mainstream over the evolution of the pandemic. These include the unprecedented speed of vaccine discovery and development, issues uncovered from early clinical studies, and regulatory concepts that were highlighted throughout the development process. Among these was the importance of pharmacovigilance as each new agent was rapidly deployed to a mostly eager public. Critical challenges around production, packaging, and procurement of product for patient use were often centre stage. Finally, the ever-important need to transition not only from scientific concept to vaccine and drug, but from their authorized and approved use to their implementation in health systems to insure the intended effects both in individuals and populations.
Collapse
Affiliation(s)
- Theodore J Witek
- Institute of Health Policy Management & Evaluation, Dalla Lana School of Public Health, University of Toronto, Health Sciences Building, 155 College Street; Fourth Floor, Toronto, ON, M5T 3M6, Canada.
| |
Collapse
|
1624
|
Yan Z, Yang M, Lai CL. COVID-19 Vaccinations: A Comprehensive Review of Their Safety and Efficacy in Special Populations. Vaccines (Basel) 2021; 9:1097. [PMID: 34696205 PMCID: PMC8539110 DOI: 10.3390/vaccines9101097] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 has been spreading worldwide since late 2019. There is no definitive cure to date. Global vaccination programs are urgently required to confer herd immunity, reducing the incidence of COVID-19 infections and associated morbidity and mortality. However, a significant proportion of special populations are hesitant to receive vaccination due to their special conditions, namely, age (pediatrics and geriatrics), immunocompromised state, autoimmune diseases, chronic cardiovascular and pulmonary conditions, active or treated cancers, and pregnancy. This review aims to evaluate the existing evidence of COVID-19 vaccinations on these special populations and to provide clues to guide vaccination decision making to balance the benefits and risks of vaccinations.
Collapse
Affiliation(s)
- Zhipeng Yan
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Ming Yang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Ching-Lung Lai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
1625
|
Xu K, Dai L, Gao GF. Humoral and cellular immunity and the safety of COVID-19 vaccines: a summary of data published by 21 May 2021. Int Immunol 2021; 33:529-540. [PMID: 34491327 PMCID: PMC8499872 DOI: 10.1093/intimm/dxab061] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/06/2021] [Indexed: 01/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has caused millions of deaths, and serious consequences to public health, economies and societies. Rapid responses in vaccine development have taken place since the isolation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the release of the viral genome sequence. By 21 May 2021, 101 vaccines were under clinical trials, and published data were available for 18 of them. Clinical study results from some vaccines indicated good immunogenicity and acceptable reactogenicity. Here, we focus on these 18 vaccines that had published clinical data to dissect the induced humoral and cellular immune responses as well as their safety profiles and protection efficacy.
Collapse
Affiliation(s)
- Kun Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan 571199, China
| | - Lianpan Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan 571199, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
1626
|
Swan DA, Goyal A, Bracis C, Moore M, Krantz E, Brown E, Cardozo-Ojeda F, Reeves DB, Gao F, Gilbert PB, Corey L, Cohen MS, Janes H, Dimitrov D, Schiffer JT. Mathematical Modeling of Vaccines That Prevent SARS-CoV-2 Transmission. Viruses 2021; 13:1921. [PMID: 34696352 PMCID: PMC8539635 DOI: 10.3390/v13101921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/01/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
SARS-CoV-2 vaccine clinical trials assess efficacy against disease (VEDIS), the ability to block symptomatic COVID-19. They only partially discriminate whether VEDIS is mediated by preventing infection completely, which is defined as detection of virus in the airways (VESUSC), or by preventing symptoms despite infection (VESYMP). Vaccine efficacy against transmissibility given infection (VEINF), the decrease in secondary transmissions from infected vaccine recipients, is also not measured. Using mathematical modeling of data from King County Washington, we demonstrate that if the Moderna (mRNA-1273QS) and Pfizer-BioNTech (BNT162b2) vaccines, which demonstrated VEDIS > 90% in clinical trials, mediate VEDIS by VESUSC, then a limited fourth epidemic wave of infections with the highly infectious B.1.1.7 variant would have been predicted in spring 2021 assuming rapid vaccine roll out. If high VEDIS is explained by VESYMP, then high VEINF would have also been necessary to limit the extent of this fourth wave. Vaccines which completely protect against infection or secondary transmission also substantially lower the number of people who must be vaccinated before the herd immunity threshold is reached. The limited extent of the fourth wave suggests that the vaccines have either high VESUSC or both high VESYMP and high VEINF against B.1.1.7. Finally, using a separate intra-host mathematical model of viral kinetics, we demonstrate that a 0.6 log vaccine-mediated reduction in average peak viral load might be sufficient to achieve 50% VEINF, which suggests that human challenge studies with a relatively low number of infected participants could be employed to estimate all three vaccine efficacy metrics.
Collapse
Affiliation(s)
- David A. Swan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Ashish Goyal
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Chloe Bracis
- TIMC-IMAG/BCM, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Mia Moore
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Elizabeth Krantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Elizabeth Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Fabian Cardozo-Ojeda
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Daniel B. Reeves
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Fei Gao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Myron S. Cohen
- Institute of Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Dobromir Dimitrov
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Department of Applied Mathematics, University of Washington, Seattle, WA 98195, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
1627
|
Windpessl M, Heine GH, Becker SL, Scheuer AL, Sester M, Kronbichler A. [The impact of immunosuppression and chronic kidney disease on immunogenicity of COVID-19 vaccines]. Dtsch Med Wochenschr 2021; 146:1277-1282. [PMID: 34553353 DOI: 10.1055/a-1550-7288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
HOW EFFECTIVE ARE THE APPROVED VACCINES IN KIDNEY DISEASES AND THOSE RECEIVING IMMUNOSUPPRESSION?: Several observational studies indicated that immunosuppression is associated with a weakened or absent humoral response. Patients with chronic kidney diseases or undergoing maintenance dialysis without immunosuppression have a reduced humoral response to COVID-19 vaccines. I HAD COVID-19. SHOULD I GET VACCINATED?: It is recommended to receive a booster after SARS-CoV-2 infection with a mRNA vaccine. IS A COVID-19 VACCINATION DESPITE ONGOING IMMUNOSUPPRESSION POSSIBLE?: Patients receiving immunosuppression have a reduced humoral response, and this is especially observed when anti-CD20 therapy is used. IS THERE A POSSIBILITY THAT THE VACCINE PROVOKES REJECTION OF MY TRANSPLANTED KIDNEY OR RELAPSE OF MY GLOMERULAR DISEASE?: Several reports were published in the last months highlighting new-onset diseases, recurrences and relapses of different glomerular diseases, which occurred after the receipt of the first or second vaccine dose. As a clear association of vaccines and induction of autoimmunity still needs to be established, most of these diseases are treatable, and COVID-19 in patients under immunosuppression is often fatal, there is a clear net benefit of vaccination. DO I HAVE A PERMANENT PROTECTION AFTER VACCINATION?: The antibody titers and likely the cellular immune response is significantly reduced in patients with kidney diseases, and titers are reducing at a fast pace under ongoing immunosuppression. A booster dose should be considered, especially taking into consideration the evolvement of virus variants and their impact on vaccine efficacy. AFTER THE FIRST SERIES OF VACCINATION, NO OR ONLY A MARGINAL AMOUNT OF ANTIBODIES WERE DETECTABLE. ARE THERE STRATEGIES TO IMPROVE VACCINE RESPONSE?: Many countries recommend the application of a third dose for vulnerable patient cohorts, especially because of a weakened response and their risk to develop a severe disease course of COVID-19. Prospective clinical trials are ongoing to test the ideal strategy to improve vaccine response in these cohorts.
Collapse
Affiliation(s)
- Martin Windpessl
- Innere Medizin IV, Klinikum Wels-Grieskirchen, Wels, Österreich.,Medizinische Fakultät, Kepler-Universitätsklinikum, Linz, Österreich
| | - Gunnar H Heine
- Medizinische Klinik II, AGAPLESION MARKUS-KRANKENHAUS, Frankfurt, Deutschland.,Universität des Saarlandes, Homburg, Deutschland
| | - Sören L Becker
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | - Anja L Scheuer
- Medizinische Klinik II, AGAPLESION MARKUS-KRANKENHAUS, Frankfurt, Deutschland.,Universität des Saarlandes, Homburg, Deutschland
| | - Martina Sester
- Abteilung für Transplantations- und Infektionsimmunologie, Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | | |
Collapse
|
1628
|
Torbati E, Krause KL, Ussher JE. The Immune Response to SARS-CoV-2 and Variants of Concern. Viruses 2021; 13:1911. [PMID: 34696342 PMCID: PMC8537260 DOI: 10.3390/v13101911] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/07/2023] Open
Abstract
At the end of 2019 a newly emerged betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was identified as the cause of an outbreak of severe pneumonia, subsequently termed COVID-19, in a number of patients in Wuhan, China. Subsequently, SARS-CoV-2 rapidly spread globally, resulting in a pandemic that has to date infected over 200 million individuals and resulted in more than 4.3 million deaths. While SARS-CoV-2 results in severe disease in 13.8%, with increasing frequency of severe disease with age, over 80% of infections are asymptomatic or mild. The immune response is an important determinant of outcome following SARS-CoV-2 infection. While B cell and T cell responses are associated with control of infection and protection against subsequent challenge with SARS-CoV-2, failure to control viral replication and the resulting hyperinflammation are associated with severe COVID-19. Towards the end of 2020, several variants of concern emerged that demonstrate increased transmissibility and/or evasion of immune responses from prior SARS-CoV-2 infection. This article reviews what is known about the humoral and cellular immune responses to SARS-CoV-2 and how mutation and structural/functional changes in the emerging variants of concern impact upon the immune protection from prior infection or vaccination.
Collapse
Affiliation(s)
- Elham Torbati
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand;
- Vaccine Alliance Aotearoa New Zealand, Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Kurt L. Krause
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand;
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - James E. Ussher
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand;
- Vaccine Alliance Aotearoa New Zealand, Malaghan Institute of Medical Research, Wellington 6242, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
1629
|
Affiliation(s)
- Jake Scott
- Division of Infectious Diseases and Geographic Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Aaron Richterman
- Division of Infectious Diseases, University of Pennsylvania, Philadelphia, PA, USA
| | - Muge Cevik
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK
| |
Collapse
|
1630
|
Movassaghi M, Chung R, Anderson CB, Stein M, Saenger Y, Faiena I. Overcoming Immune Resistance in Prostate Cancer: Challenges and Advances. Cancers (Basel) 2021; 13:cancers13194757. [PMID: 34638243 PMCID: PMC8507531 DOI: 10.3390/cancers13194757] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Immunotherapy has changed the landscape of treatment modalities available for many different types of malignancies. However, the factors that influence the success of immunotherapeutics have not been as clearly seen in advanced prostate cancer, likely due to immunosuppressive factors that exist within the prostate cancer tumor microenvironment. While there have been many immunotherapeutics used for prostate cancer, the majority have targeted a single immunosuppressive mechanism resulting in limited clinical efficacy. More recent research centered on elucidating the key mechanisms of immune resistance in the prostate tumor microenvironment has led to the discovery of a range of new treatment targets. With that in mind, many clinical trials have now set out to evaluate combination immunotherapeutic strategies in patients with advanced prostate cancer, in the hopes of circumventing the immunosuppressive mechanisms. Abstract The use of immunotherapy has become a critical treatment modality in many advanced cancers. However, immunotherapy in prostate cancer has not been met with similar success. Multiple interrelated mechanisms, such as low tumor mutational burden, immunosuppressive cells, and impaired cellular immunity, appear to subvert the immune system, creating an immunosuppressive tumor microenvironment and leading to lower treatment efficacy in advanced prostate cancer. The lethality of metastatic castrate-resistant prostate cancer is driven by the lack of therapeutic regimens capable of generating durable responses. Multiple strategies are currently being tested to overcome immune resistance including combining various classes of treatment modalities. Several completed and ongoing trials have shown that combining vaccines or checkpoint inhibitors with hormonal therapy, radiotherapy, antibody–drug conjugates, chimeric antigen receptor T cell therapy, or chemotherapy may enhance immune responses and induce long-lasting clinical responses without significant toxicity. Here, we review the current state of immunotherapy for prostate cancer, as well as tumor-specific mechanisms underlying therapeutic resistance, with a comprehensive look at the current preclinical and clinical immunotherapeutic strategies aimed at overcoming the immunosuppressive tumor microenvironment and impaired cellular immunity that have largely limited the utility of immunotherapy in advanced prostate cancer.
Collapse
Affiliation(s)
- Miyad Movassaghi
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| | - Rainjade Chung
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Christopher B. Anderson
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Mark Stein
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Yvonne Saenger
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Izak Faiena
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| |
Collapse
|
1631
|
Günl F, Mecate-Zambrano A, Rehländer S, Hinse S, Ludwig S, Brunotte L. Shooting at a Moving Target-Effectiveness and Emerging Challenges for SARS-CoV-2 Vaccine Development. Vaccines (Basel) 2021; 9:1052. [PMID: 34696160 PMCID: PMC8540924 DOI: 10.3390/vaccines9101052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023] Open
Abstract
Since late 2019 the newly emerged pandemic SARS-CoV-2, the causative agent of COVID-19, has hit the world with recurring waves of infections necessitating the global implementation of non-pharmaceutical interventions, including strict social distancing rules, the wearing of masks and the isolation of infected individuals in order to restrict virus transmissions and prevent the breakdown of our healthcare systems. These measures are not only challenging on an economic level but also have a strong impact on social lifestyles. Using traditional and novel technologies, highly efficient vaccines against SARS-CoV-2 were developed and underwent rapid clinical evaluation and approval to accelerate the immunization of the world population, aiming to end the pandemic and return to normality. However, the emergence of virus variants with improved transmission, enhanced fitness and partial immune escape from the first generation of vaccines poses new challenges, which are currently being addressed by scientists and pharmaceutical companies all over the world. In this ongoing pandemic, the evaluation of SARS-CoV-2 vaccines underlies diverse unpredictable dynamics, posed by the first broad application of the mRNA vaccine technology and their compliance, the occurrence of unexpected side effects and the rapid emergence of variations in the viral antigen. However, despite these hurdles, we conclude that the available SARS-CoV-2 vaccines are very safe and efficiently protect from severe COVID-19 and are thereby the most powerful tools to prevent further harm to our healthcare systems, economics and individual lives. This review summarizes the unprecedented pathways of vaccine development and approval during the ongoing SARS-CoV-2 pandemic. We focus on the real-world effectiveness and unexpected positive and negative side effects of the available vaccines and summarize the timeline of the applied adaptations to the recommended vaccination strategies in the light of emerging virus variants. Finally, we highlight upcoming strategies to improve the next generations of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Franziska Günl
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Angeles Mecate-Zambrano
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Selina Rehländer
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Saskia Hinse
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Linda Brunotte
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| |
Collapse
|
1632
|
Haimei MA. Concern About the Adverse Effects of Thrombocytopenia and Thrombosis After Adenovirus-Vectored COVID-19 Vaccination. Clin Appl Thromb Hemost 2021; 27:10760296211040110. [PMID: 34541935 PMCID: PMC8642058 DOI: 10.1177/10760296211040110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Since the outbreak of Covid-19 in December, 2019, scientists worldwide have been
committed to developing COVID-19 vaccines. Only when most people have immunity
to SARS-CoV-2, COVID-19 can reduce even wholly overcome. So far, nine kinds of
COVID-19 vaccines have passed the phase III clinical trials and have approved
for use. At the same time, adverse reactions after COVID-19 vaccination have
also reported. This paper focuses on the adverse effects of thrombosis and
thrombocytopenia caused by the COVID-19 vaccine, especially the
adenovirus-vector vaccine from AstraZeneca and Pfizer, and discusses its
mechanism and possible countermeasures.
Collapse
Affiliation(s)
- M A Haimei
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
1633
|
Patarroyo ME, Patarroyo MA, Alba MP, Pabon L, Rugeles MT, Aguilar-Jimenez W, Florez L, Bermudez A, Rout AK, Griesinger C, Suarez CF, Aza-Conde J, Reyes C, Avendaño C, Samacá J, Camargo A, Silva Y, Forero M, Gonzalez E. The First Chemically-Synthesised, Highly Immunogenic Anti-SARS-CoV-2 Peptides in DNA Genotyped Aotus Monkeys for Human Use. Front Immunol 2021; 12:724060. [PMID: 34539660 PMCID: PMC8446425 DOI: 10.3389/fimmu.2021.724060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Thirty-five peptides selected from functionally-relevant SARS-CoV-2 spike (S), membrane (M), and envelope (E) proteins were suitably modified for immunising MHC class II (MHCII) DNA-genotyped Aotus monkeys and matched with HLA-DRβ1* molecules for use in humans. This was aimed at producing the first minimal subunit-based, chemically-synthesised, immunogenic molecules (COLSARSPROT) covering several HLA alleles. They were predicted to cover 48.25% of the world’s population for 6 weeks (short-term) and 33.65% for 15 weeks (long-lasting) as they induced very high immunofluorescent antibody (IFA) and ELISA titres against S, M and E parental native peptides, SARS-CoV-2 neutralising antibodies and host cell infection. The same immunological methods that led to identifying new peptides for inclusion in the COLSARSPROT mixture were used for antigenicity studies. Peptides were analysed with serum samples from patients suffering mild or severe SARS-CoV-2 infection, thereby increasing chemically-synthesised peptides’ potential coverage for the world populations up to 62.9%. These peptides’ 3D structural analysis (by 1H-NMR acquired at 600 to 900 MHz) suggested structural-functional immunological association. This first multi-protein, multi-epitope, minimal subunit-based, chemically-synthesised, highly immunogenic peptide mixture highlights such chemical synthesis methodology’s potential for rapidly obtaining very pure, highly reproducible, stable, cheap, easily-modifiable peptides for inducing immune protection against COVID-19, covering a substantial percentage of the human population.
Collapse
Affiliation(s)
- Manuel E Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,Universidad Santo Tomás, Bogotá, Colombia
| | - Manuel A Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Martha P Alba
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Laura Pabon
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - María T Rugeles
- Grupo Inmunovirología, Universidad de Antioquia, Medellín, Colombia
| | | | - Lizdany Florez
- Grupo Inmunovirología, Universidad de Antioquia, Medellín, Colombia
| | - Adriana Bermudez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Ashok K Rout
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian Griesinger
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Carlos F Suarez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Jorge Aza-Conde
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - César Reyes
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Catalina Avendaño
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá, Colombia
| | - Jhoan Samacá
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Anny Camargo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Yolanda Silva
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Martha Forero
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Edgardo Gonzalez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| |
Collapse
|
1634
|
Hargrave A, Mustafa AS, Hanif A, Tunio JH, Hanif SNM. Current Status of HIV-1 Vaccines. Vaccines (Basel) 2021; 9:1026. [PMID: 34579263 PMCID: PMC8471857 DOI: 10.3390/vaccines9091026] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
HIV-1 infection and its progression to AIDS remains a significant global health challenge, particularly for low-income countries. Developing a vaccine to prevent HIV-1 infections has proven to be immensely challenging with complex biological acquisition and infection, unforeseen clinical trial disappointments, and funding issues. This paper discusses important landmarks of progress in HIV-1 vaccine development, various vaccine strategies, and clinical trials.
Collapse
Affiliation(s)
- Anna Hargrave
- Department of Biomedical Sciences, Kentucky College of Osteopathic Medicine, University of Pikeville, Pikeville, KY 41501, USA;
| | - Abu Salim Mustafa
- Department of Microbiology, College of Medicine, Kuwait University, Kuwait City 12037, Kuwait;
| | - Asma Hanif
- Department of Restorative Sciences, College of Dentistry, Kuwait University, Kuwait City 12037, Kuwait;
| | - Javed H. Tunio
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Shumaila Nida M. Hanif
- Department of Biomedical Sciences, Kentucky College of Osteopathic Medicine, University of Pikeville, Pikeville, KY 41501, USA;
| |
Collapse
|
1635
|
Malcangi G, Inchingolo AD, Inchingolo AM, Santacroce L, Marinelli G, Mancini A, Vimercati L, Maggiore ME, D’Oria MT, Hazballa D, Bordea IR, Xhajanka E, Scarano A, Farronato M, Tartaglia GM, Giovanniello D, Nucci L, Serpico R, Sammartino G, Capozzi L, Parisi A, Di Domenico M, Lorusso F, Contaldo M, Inchingolo F, Dipalma G. COVID-19 Infection in Children, Infants and Pregnant Subjects: An Overview of Recent Insights and Therapies. Microorganisms 2021; 9:1964. [PMID: 34576859 PMCID: PMC8469368 DOI: 10.3390/microorganisms9091964] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The SARS-CoV-2 pandemic has involved a severe increase of cases worldwide in a wide range of populations. The aim of the present investigation was to evaluate recent insights about COVID-19 infection in children, infants and pregnant subjects. METHODS a literature overview was performed including clinical trials, in vitro studies, reviews and published guidelines regarding the present paper topic. A descriptive synthesis was performed to evaluate recent insights and the effectiveness of therapies for SARS-CoV-2 infection in children, infants and pregnant subjects. RESULTS Insufficient data are available regarding the relationship between COVID-19 and the clinical risk of spontaneous abortion and premature foetus death. A decrease in the incidence of COVID-19 could be correlated to a minor expression of ACE2 in childrens' lungs. At present, a modulation of the dose-effect posology for children and infants is necessary. CONCLUSIONS Pregnant vertical transmission has been hypothesised for SARS-CoV-2 infection. Vaccines are necessary to achieve mass immunity for children and also pregnant subjects.
Collapse
Affiliation(s)
- Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Grazia Marinelli
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Maria Elena Maggiore
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Denisa Hazballa
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
- Kongresi Elbasanit, Rruga: Aqif Pasha, 3001 Elbasan, Albania
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Edit Xhajanka
- Department of Dental Prosthesis, Medical University of Tirana, Rruga e Dibrës, U.M.T., 1001 Tirana, Albania;
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Marco Farronato
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | | | - Ludovica Nucci
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Rosario Serpico
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Gilberto Sammartino
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy;
| | - Loredana Capozzi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, 71121 Foggia, Italy; (L.C.); (A.P.)
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, 71121 Foggia, Italy; (L.C.); (A.P.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| |
Collapse
|
1636
|
Detection of SARS-CoV-2 antibodies formed in response to the BNT162b2 and mRNA-1237 mRNA vaccine by commercial antibody tests. Vaccine 2021; 39:5563-5570. [PMID: 34454782 PMCID: PMC8354789 DOI: 10.1016/j.vaccine.2021.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 01/11/2023]
Abstract
Background With rapid approval of SARS-CoV-2 vaccines, the ability of clinical laboratories to detect vaccine-induced antibodies with available high-throughput commercial assays is unknown. We aimed to determine if commercial serology assays can detect vaccine-induced antibodies (VIAs) and understand the vaccination response. Methods This cohort study recruited healthcare workers and residents of long-term care facilities (receiving the BNT162b2 and mRNA-1273 products, respectively) who underwent serum collection pre-vaccination (BNT162b2 group), 2-weeks post vaccination (both groups), and pre-2nd dose (both groups). Sera were tested for the presence of SARS-CoV-2 IgG using four commercial assays (Abbott SARS-CoV-2 IgG, Abbott SARS-CoV-2 IgG II Quant, DiaSorin Trimeric S IgG, and GenScript cPASS) to detect VIAs. Secondary outcomes included description of post-vaccination antibody response and correlation with neutralizing titers. Results 225 participants (177 receiving BNT162b2 and 48 receiving mRNA-1273) were included (median age 41 years; 66–78% female). Nucleocapsid IgG was found in 4.1% and 21.9% of the BNT162b2 (baseline) and mRNA-1273 (2-weeks post first dose). All anti-spike assays detected antibodies post-vaccination, with an average increase of 87.2% (range 73.8–94.3%; BNT162b2), and 25.2% (range 23.8–26.7%; mRNA-1273) between the first and last sampling time points (all p < 0.05). Neutralizing antibodies were detected at all post-vaccine timepoints for both vaccine arms, with increasing titers over time (all p < 0.05). Conclusions Anti-spike vaccine-induced SARS-CoV-2 IgG are detectable by commercially available high-throughput assays and increases over time. Prior to second dose of vaccination, neutralizing antibodies are detectable in 73–89% of individuals, suggesting most individuals would have some degree of protection from subsequent infection.
Collapse
|
1637
|
Chiuppesi F, Nguyen VH, Park Y, Contreras H, Karpinski V, Faircloth K, Nguyen J, Kha M, Johnson D, Martinez J, Iniguez A, Zhou Q, Kaltcheva T, Frankel P, Kar S, Sharma A, Andersen H, Lewis MG, Shostak Y, Wussow F, Diamond DJ. Synthetic Multiantigen MVA Vaccine COH04S1 Protects Against SARS-CoV-2 in Syrian Hamsters and Non-Human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34545366 DOI: 10.1101/2021.09.15.460487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Second-generation COVID-19 vaccines could contribute to establish protective immunity against SARS-CoV-2 and its emerging variants. We developed COH04S1, a synthetic multiantigen Modified Vaccinia Ankara-based SARS-CoV-2 vaccine that co-expresses spike and nucleocapsid antigens. Here, we report COH04S1 vaccine efficacy in animal models. We demonstrate that intramuscular or intranasal vaccination of Syrian hamsters with COH04S1 induces robust Th1-biased antigen-specific humoral immunity and cross-neutralizing antibodies (NAb) and protects against weight loss, lower respiratory tract infection, and lung injury following intranasal SARS-CoV-2 challenge. Moreover, we demonstrate that single-dose or two-dose vaccination of non-human primates with COH04S1 induces robust antigen-specific binding antibodies, NAb, and Th1-biased T cells, protects against both upper and lower respiratory tract infection following intranasal/intratracheal SARS-CoV-2 challenge, and triggers potent post-challenge anamnestic antiviral responses. These results demonstrate COH04S1-mediated vaccine protection in animal models through different vaccination routes and dose regimens, complementing ongoing investigation of this multiantigen SARS-CoV-2 vaccine in clinical trials.
Collapse
|
1638
|
Sengupta S, Bhattacharyya D, Kasle G, Karmakar S, Sahu O, Ganguly A, Addya S, Das Sarma J. Potential Immunomodulatory Properties of Biologically Active Components of Spices Against SARS-CoV-2 and Pan β-Coronaviruses. Front Cell Infect Microbiol 2021; 11:729622. [PMID: 34513735 PMCID: PMC8431827 DOI: 10.3389/fcimb.2021.729622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced COVID-19 has emerged as a defining global health crisis in current times. Data from the World Health Organization shows demographic variations in COVID-19 severity and lethality. Diet may play a significant role in providing beneficial host cell factors contributing to immunity against deadly SARS-CoV-2 pathogenesis. Spices are essential components of the diet that possess anti-inflammatory, antioxidant, and antiviral properties. Hyperinflammation, an aberrant systemic inflammation associated with pneumonia, acute respiratory failure, and multiorgan dysfunction, is a major clinical outcome in COVID-19. Knowing the beneficial properties of spices, we hypothesize that spice-derived bioactive components can modulate host immune responses to provide protective immunity in COVID-19. This study emphasizes that biologically active components of spices might alleviate the sustained pro-inflammatory condition by inhibiting the activity of tumor necrosis factor-alpha (TNF-α), interleukins (IL6, IL8), and chemokine (CCL2) known to be elevated in COVID-19. Spices may potentially prevent the tissue damage induced by oxidative stress and pro-inflammatory mediators during SARS-CoV-2 infection. The current study also highlights the effects of spices on the antioxidant pathways mediated by Nrf2 (nuclear factor erythroid 2-related factor 2) and Hmox1 (heme oxygenase 1) to restore oxidative homeostasis and protect from aberrant tissue damage. Taken together, the anti-inflammatory and antioxidant activities of bioactive components of spices may hold a promise to target the cellular pathways for developing antivirals against SARS-CoV-2 and pan β-coronaviruses.
Collapse
Affiliation(s)
- Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Debina Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Grishma Kasle
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Souvik Karmakar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Omkar Sahu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Anirban Ganguly
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Sankar Addya
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| |
Collapse
|
1639
|
Huang Y, Borisov O, Kee JJ, Carpp LN, Wrin T, Cai S, Sarzotti-Kelsoe M, McDanal C, Eaton A, Pajon R, Hural J, Posavad CM, Gill K, Karuna S, Corey L, McElrath MJ, Gilbert PB, Petropoulos CJ, Montefiori DC. Calibration of Two Validated SARS-CoV-2 Pseudovirus Neutralization Assays for COVID-19 Vaccine Evaluation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.09.09.21263049. [PMID: 34545372 PMCID: PMC8452111 DOI: 10.1101/2021.09.09.21263049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Vaccine-induced neutralizing antibodies (nAbs) are key biomarkers considered to be associated with vaccine efficacy. In United States Government-sponsored phase 3 efficacy trials of COVID-19 vaccines, nAbs are measured by two different validated pseudovirus-based SARS-CoV-2 neutralization assays, with each trial using one of the two assays. Here we describe and compare the nAb titers obtained in the two assays. We observe that one assay consistently yielded higher nAb titers than the other when both assays were performed on the World Health Organization’s anti-SARS-CoV-2 immunoglobulin International Standard, COVID-19 convalescent sera, and mRNA-1273 vaccinee sera. To overcome the challenge this difference in readout poses in comparing/combining data from the two assays, we evaluate three calibration approaches and show that readouts from the two assays can be calibrated to a common scale. These results may aid decision-making based on data from these assays for the evaluation and licensure of new or adapted COVID-19 vaccines.
Collapse
Affiliation(s)
- Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Oleg Borisov
- Biomedical Advanced Research and Development Authority, Washington DC, USA
| | - Jia Jin Kee
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lindsay N. Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Terri Wrin
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Suqin Cai
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Marcella Sarzotti-Kelsoe
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Charlene McDanal
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Amanda Eaton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christine M. Posavad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Katherine Gill
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
1640
|
Tsatsakis A, Vakonaki E, Tzatzarakis M, Flamourakis M, Nikolouzakis TK, Poulas K, Papazoglou G, Hatzidaki E, Papanikolaou NC, Drakoulis N, Iliaki E, Goulielmos GN, Kallionakis M, Lazopoulos G, Kteniadakis S, Alegkakis A, Farsalinos K, Spandidos DA. Immune response (IgG) following full inoculation with BNT162b2 COVID‑19 mRNA among healthcare professionals. Int J Mol Med 2021; 48:200. [PMID: 34515322 PMCID: PMC8448546 DOI: 10.3892/ijmm.2021.5033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Soon after the beginning of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic in December, 2019, numerous research teams, assisted by vast capital investments, achieved vaccine development in a fraction of time. However, almost 8 months following the initiation of the European vaccination programme, the need for prospective monitoring of the vaccine-induced immune response, its determinants and related side-effects remains a priority. The present study aimed to quantify the immune response following full vaccination with the BNT162b2 coronavirus disease 2019 (COVID-19) mRNA vaccine by measuring the levels of immunoglobulin G (IgG) titers in healthcare professionals. Moreover, common side-effects and factors associated with IgG titers were identified. For this purpose, blood samples from 517 individuals were obtained and analysed. Blood sampling was performed at a mean period of 69.0±23.5 days following the second dose of the vaccine. SARS-CoV-2 IgG titers had an overall mean value of 4.23±2.76. Females had higher titers than males (4.44±2.70 and 3.89 ±2.84, respectively; P=0.007), while non-smokers had higher titers than smokers (4.48±2.79 and 3.80±2.64, respectively; P=0.003). An older age was also associated with lower antibody titers (P<0.001). Moreover, the six most prevalent adverse effects were pain at the injection site (72.1%), generalized fatigue (40.5%), malaise (36.3%), myalgia (31,0%), headache (25.8%) and dizziness/weakness (21.6%). The present study demonstrated that the immune response after receiving the BNT162b2 COVID-19 mRNA vaccine is dependent on various modifiable and non-modifiable factors. Overall, the findings of the present study highlight two key aspects of the vaccination programs: First, the need for prospective immunosurveillance studies in order to estimate the duration of immunity, and second, the need to identify those individuals who are at a greater risk of developing low IgG titers in order to evaluate the need for a third dose of the vaccine.
Collapse
Affiliation(s)
- Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Elena Vakonaki
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Manolis Tzatzarakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Matthaios Flamourakis
- Department of General Surgery, Venizeleion General Hospital, 71409 Heraklion, Greece
| | | | - Konstantinos Poulas
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26500 Rio‑Patras, Greece
| | - Georgios Papazoglou
- Department of Emergency Medicine, Venizeleion General Hospital, 71409 Heraklion, Greece
| | - Eleftheria Hatzidaki
- Department of Neonatology and Neonatal Intensive Care Unit, University Hospital of Heraklion, 71500 Heraklion, Greece
| | | | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Evangelia Iliaki
- Department of Internal Medicine, Venizeleion General Hospital, 71409 Heraklion, Greece
| | - Georgios N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Manolis Kallionakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Georgios Lazopoulos
- Department of Cardiothoracic Surgery, University Hospital of Heraklion, 71500 Heraklion, Greece
| | | | - Athanasios Alegkakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Konstantinos Farsalinos
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26500 Rio‑Patras, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
1641
|
Cancer or COVID-19? A Review of Recommendations for COVID-19 Vaccination in Cancer Patients. Curr Treat Options Oncol 2021; 22:95. [PMID: 34515857 PMCID: PMC8436191 DOI: 10.1007/s11864-021-00903-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
While emergency use is authorized for numerous COVID-19 vaccines and the high-risk population including cancer patients or those with immunosuppression due to disease or therapy is prioritized, data on this group’s specific safety and efficacy of these vaccines remains limited. Safety data from clinical trials and population data may be extrapolated, and these vaccines may be used for cancer patients. However, concerns of efficacy due to the variable immune response in patients with active cancers undergoing active therapy and cancer survivors with chronic immunosuppression remain. The authors aim to discuss the current recommendations for use of COVID-19 vaccination in patients with cancer.
Collapse
|
1642
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections trigger viral RNA sensors such as TLR7 and RIG-I, thereby leading to production of type I interferon (IFN) and other inflammatory mediators. Expression of viral proteins in the context of this inflammation leads to stereotypical antigen-specific antibody and T cell responses that clear the virus. Immunity is then maintained through long-lived antibody-secreting plasma cells and by memory B and T cells that can initiate anamnestic responses. Each of these steps is consistent with prior knowledge of acute RNA virus infections. Yet there are certain concepts, while not entirely new, that have been resurrected by the biology of severe SARS-CoV-2 infections and deserve further attention. These include production of anti-IFN autoantibodies, early inflammatory processes that slow adaptive humoral immunity, immunodominance of antibody responses, and original antigenic sin. Moreover, multiple different vaccine platforms allow for comparisons of pathways that promote robust and durable adaptive immunity.
Collapse
Affiliation(s)
- Dominik Schenten
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, United States.
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, United States.
| |
Collapse
|
1643
|
Cairoli E, Espinosa G. Autoimmune diseases and vaccines against COVID-19. Decision making in uncertain scenarios. MEDICINA CLINICA (ENGLISH ED.) 2021; 157:247-252. [PMID: 34395909 PMCID: PMC8346352 DOI: 10.1016/j.medcle.2021.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/13/2021] [Indexed: 12/01/2022]
Affiliation(s)
- Ernesto Cairoli
- Unidad de Enfermedades Autoinmunes, Hospital Evangélico y Centro Asistencial del Sindicato Médico del Uruguay (CASMU), Montevideo, Uruguay.,Laboratorio de Inmunorregulación e Inflamación, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gerard Espinosa
- Servicio de Enfermedades Autoinmunes, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
1644
|
Kates OS. The right to bare arms. Transpl Infect Dis 2021; 23:e13723. [PMID: 34494707 DOI: 10.1111/tid.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Olivia S Kates
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
1645
|
Zabaleta N, Dai W, Bhatt U, Hérate C, Maisonnasse P, Chichester JA, Sanmiguel J, Estelien R, Michalson KT, Diop C, Maciorowski D, Dereuddre-Bosquet N, Cavarelli M, Gallouët AS, Naninck T, Kahlaoui N, Lemaitre J, Qi W, Hudspeth E, Cucalon A, Dyer CD, Pampena MB, Knox JJ, LaRocque RC, Charles RC, Li D, Kim M, Sheridan A, Storm N, Johnson RI, Feldman J, Hauser BM, Contreras V, Marlin R, Tsong Fang RH, Chapon C, van der Werf S, Zinn E, Ryan A, Kobayashi DT, Chauhan R, McGlynn M, Ryan ET, Schmidt AG, Price B, Honko A, Griffiths A, Yaghmour S, Hodge R, Betts MR, Freeman MW, Wilson JM, Le Grand R, Vandenberghe LH. An AAV-based, room-temperature-stable, single-dose COVID-19 vaccine provides durable immunogenicity and protection in non-human primates. Cell Host Microbe 2021; 29:1437-1453.e8. [PMID: 34428428 PMCID: PMC8346325 DOI: 10.1016/j.chom.2021.08.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022]
Abstract
The SARS-CoV-2 pandemic has affected more than 185 million people worldwide resulting in over 4 million deaths. To contain the pandemic, there is a continued need for safe vaccines that provide durable protection at low and scalable doses and can be deployed easily. Here, AAVCOVID-1, an adeno-associated viral (AAV), spike-gene-based vaccine candidate demonstrates potent immunogenicity in mouse and non-human primates following a single injection and confers complete protection from SARS-CoV-2 challenge in macaques. Peak neutralizing antibody titers are sustained at 1 year and complemented by functional memory T cell responses. The AAVCOVID vector has no relevant pre-existing immunity in humans and does not elicit cross-reactivity to common AAVs used in gene therapy. Vector genome persistence and expression wanes following injection. The single low-dose requirement, high-yield manufacturability, and 1-month stability for storage at room temperature may make this technology well suited to support effective immunization campaigns for emerging pathogens on a global scale.
Collapse
Affiliation(s)
- Nerea Zabaleta
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Wenlong Dai
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Urja Bhatt
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Cécile Hérate
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Jessica A Chichester
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julio Sanmiguel
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Reynette Estelien
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kristofer T Michalson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cheikh Diop
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Dawid Maciorowski
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Nidhal Kahlaoui
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Wenbin Qi
- Novartis Gene Therapies, San Diego, CA, USA
| | | | - Allison Cucalon
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Cecilia D Dyer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Betina Pampena
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James J Knox
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina C LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Richelle C Charles
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dan Li
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Maya Kim
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Abigail Sheridan
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Nadia Storm
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA
| | - Rebecca I Johnson
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Blake M Hauser
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Vanessa Contreras
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Raphaël Ho Tsong Fang
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Catherine Chapon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Sylvie van der Werf
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur, CNRS UMR 3569, Université de Paris, Paris, France; National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | - Eric Zinn
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Aisling Ryan
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Dione T Kobayashi
- Translational Innovation Fund, Mass General Brigham Innovation, Cambridge, MA, USA
| | - Ruchi Chauhan
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Marion McGlynn
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | | | - Anna Honko
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony Griffiths
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | - Michael R Betts
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mason W Freeman
- Center for Computational & Integrative Biology, Department of Medicine, and Translational Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James M Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France.
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
1646
|
Janes H, Gao F, Luedtke A. Discussion on "Estimating vaccine efficacy over time after a randomized study is unblinded" by Anastasios A. Tsiatis and Marie Davidian. Biometrics 2021; 78:841-843. [PMID: 34492117 PMCID: PMC8653041 DOI: 10.1111/biom.13542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/24/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Fei Gao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
1647
|
Dangi T, Class J, Palacio N, Richner JM, Penaloza MacMaster P. Combining spike- and nucleocapsid-based vaccines improves distal control of SARS-CoV-2. Cell Rep 2021; 36:109664. [PMID: 34450033 PMCID: PMC8367759 DOI: 10.1016/j.celrep.2021.109664] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/13/2021] [Accepted: 08/13/2021] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 infection causes respiratory insufficiency and neurological manifestations, including loss of smell and psychiatric disorders, and can be fatal. Most vaccines are based on the spike antigen alone, and although they have shown efficacy at preventing severe disease and death, they do not always confer sterilizing immunity. Here, we interrogate whether SARS-CoV-2 vaccines could be improved by incorporating nucleocapsid as an antigen. We show that, after 72 h of challenge, a spike-based vaccine confers acute protection in the lung, but not in the brain. However, combining a spike-based vaccine with a nucleocapsid-based vaccine confers acute protection in both the lung and brain. These findings suggest that nucleocapsid-specific immunity can improve the distal control of SARS-CoV-2, warranting the inclusion of nucleocapsid in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jacob Class
- Department of Microbiology & Immunology, University of Illinois, Chicago College of Medicine, Chicago, IL 60612, USA
| | - Nicole Palacio
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Justin M Richner
- Department of Microbiology & Immunology, University of Illinois, Chicago College of Medicine, Chicago, IL 60612, USA.
| | - Pablo Penaloza MacMaster
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
1648
|
Jabłońska K, Aballéa S, Toumi M. The real-life impact of vaccination on COVID-19 mortality in Europe and Israel. Public Health 2021; 198:230-237. [PMID: 34482101 PMCID: PMC8413007 DOI: 10.1016/j.puhe.2021.07.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/14/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES This study aimed at estimating the real-life impact of vaccination on COVID-19 mortality, with adjustment for SARS-CoV-2 variants spread and other factors across Europe and Israel. STUDY DESIGN Time series analysis. METHODS Time series analysis of the daily number of COVID-19 deaths was performed using non-linear Poisson mixed regression models. Variables such as variants' frequency, demographics, climate, health, and mobility characteristics of thirty-two countries between January 2020 and April 2021 were considered as potentially relevant adjustment factors. RESULTS The analysis revealed that vaccination efficacy in terms of protection against deaths was 72%, with a lower reduction of the number of deaths for B.1.1.7 vs non-B.1.1.7 variants (70% and 78%, respectively). Other factors significantly related to mortality were arrivals at airports, mobility change from the prepandemic level, and temperature. CONCLUSIONS Our study confirms a strong effectiveness of COVID-19 vaccination based on real-life public data, although lower than expected from clinical trials. This suggests the absence of indirect protection for non-vaccinated individuals. Results also show that vaccination effectiveness against mortality associated with the B.1.1.7 variant is slightly lower than that with other variants. Lastly, this analysis confirms the role of mobility reduction, within and between countries, as an effective way to reduce COVID-19 mortality and suggests the possibility of seasonal variations in COVID-19 incidence.
Collapse
Affiliation(s)
| | - Samuel Aballéa
- Creativ-Ceutical, Westblaak 90, Rotterdam, 3012KM, the Netherlands.
| | - Mondher Toumi
- Faculty of Medicine, Public Health Department, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13005, France.
| |
Collapse
|
1649
|
[SARS-CoV-2-update on skin manifestations, predictive markers and cutaneous reactions after vaccination]. Hautarzt 2021; 72:929-932. [PMID: 34480208 PMCID: PMC8415434 DOI: 10.1007/s00105-021-04881-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 12/03/2022]
|
1650
|
D’Amelio R, Asero R, Cassatella MA, Laganà B, Lunardi C, Migliorini P, Nisini R, Parronchi P, Quinti I, Racanelli V, Senna G, Vacca A, Maggi E. Anti-COVID-19 Vaccination in Patients with Autoimmune-Autoinflammatory Disorders and Primary/Secondary Immunodeficiencies: The Position of the Task Force on Behalf of the Italian Immunological Societies. Biomedicines 2021; 9:1163. [PMID: 34572349 PMCID: PMC8465958 DOI: 10.3390/biomedicines9091163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023] Open
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic has represented an unprecedented challenge for humankind from health, economic, and social viewpoints. In February 2020, Italy was the first western country to be deeply hit by the pandemic and suffered the highest case/fatality rate among western countries. Brand new anti-COVID-19 vaccines have been developed and made available in <1-year from the viral sequence publication. Patients with compromised immune systems, such as autoimmune-autoinflammatory disorders (AIAIDs), primary (PIDs) and secondary (SIDs) immunodeficiencies, have received careful attention for a long time regarding their capacity to safely respond to traditional vaccines. The Italian Immunological Societies, therefore, have promptly faced the issues of safety, immunogenicity, and efficacy/effectiveness of the innovative COVID-19 vaccines, as well as priority to vaccine access, in patients with AIADs, PIDs, and SIDs, by organizing an ad-hoc Task Force. Patients with AIADs, PIDs, and SIDs: (1) Do not present contraindications to COVID-19 vaccines if a mRNA vaccine is used and administered in a stabilized disease phase without active infection. (2) Should usually not discontinue immunosuppressive therapy, which may be modulated depending on the patient's clinical condition. (3) When eligible, should have a priority access to vaccination. In fact, immunizing these patients may have relevant social/health consequences, since these patients, if infected, may develop chronic infection, which prolongs viral spread and facilitates the emergence of viral variants.
Collapse
Affiliation(s)
- Raffaele D’Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Via di Grottarossa 1035-1039, 00189 Rome, Italy;
| | - Riccardo Asero
- Ambulatorio di Allergologia, Clinica S. Carlo di Paderno Dugnano, Via Ospedale 21, 20037 Milano, Italy;
| | - Marco Antonio Cassatella
- Sezione di Patologia Generale, Dipartimento di Medicina, Università di Verona, Strada Le Grazie 4, 37134 Verona, Italy;
| | - Bruno Laganà
- UOC Medicina Interna, Dipartimento di Medicina Clinica e Molecolare, AOU S. Andrea, Sapienza Università di Roma, Via di Grottarossa 1035-1039, 00189 Rome, Italy;
| | - Claudio Lunardi
- Responsabile Unità di Malattie Autoimmunitarie, Dipartimento di Medicina, AOU Policlinico G.B. Rossi, Borgo Roma, Università di Verona, Piazzale Ludovico Antonio Scuro 10, 37134 Verona, Italy;
| | - Paola Migliorini
- Direttore Unità Operativa di Immunoallergologia Clinica, Dipartimento di Medicina Clinica e Sperimentale, Azienda Ospedaliero Universitaria Pisana, Università di Pisa, Via Roma 67, 56126 Pisa, Italy;
| | - Roberto Nisini
- Direttore Reparto Immunologia, Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Paola Parronchi
- Direttore SOD Immunologia e Terapie Cellulari, Dipartimento di Medicina Sperimentale e Clinica, AOU Careggi, Università di Firenze, Largo Brambilla 3, 50134 Firenze, Italy;
| | - Isabella Quinti
- Responsabile UOD Centro di Riferimento Regionale per le Immunodeficienze, Dipartimento di Medicina Molecolare, AOU Policlinico Umberto I, Sapienza Università di Roma, Viale dell’Università 37, 00161 Rome, Italy;
| | - Vito Racanelli
- UOC Medicina Interna “Guido Baccelli”, Dipartimento di Scienze Biomediche ed Oncologia Umana, AOU Policlinico, Università di Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Gianenrico Senna
- Direttore USD Allergologia, Dipartimento di Medicina, AOU Policlinico G.B. Rossi, Borgo Roma, Università di Verona, Piazzale Ludovico Antonio Scuro 10, 37134 Verona, Italy;
| | - Angelo Vacca
- Direttore UOC Medicina Interna “Guido Baccelli”, Dipartimento di Scienze Biomediche ed Oncologia Umana, AOU Policlinico, Università di Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Enrico Maggi
- Unità di Immunità Traslazionale, Dipartimento di Immunologia, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di S. Paolo 15, 00146 Rome, Italy
| |
Collapse
|