Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Nature ; 630(8018): 950-960, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749479

ABSTRACT

Immune imprinting is a phenomenon in which prior antigenic experiences influence responses to subsequent infection or vaccination1,2. The effects of immune imprinting on serum antibody responses after boosting with variant-matched SARS-CoV-2 vaccines remain uncertain. Here we characterized the serum antibody responses after mRNA vaccine boosting of mice and human clinical trial participants. In mice, a single dose of a preclinical version of mRNA-1273 vaccine encoding Wuhan-1 spike protein minimally imprinted serum responses elicited by Omicron boosters, enabling generation of type-specific antibodies. However, imprinting was observed in mice receiving an Omicron booster after two priming doses of mRNA-1273, an effect that was mitigated by a second booster dose of Omicron vaccine. In both SARS-CoV-2-infected and uninfected humans who received two Omicron-matched boosters after two or more doses of the prototype mRNA-1273 vaccine, spike-binding and neutralizing serum antibodies cross-reacted with Omicron variants as well as more distantly related sarbecoviruses. Because serum neutralizing responses against Omicron strains and other sarbecoviruses were abrogated after pre-clearing with Wuhan-1 spike protein, antibodies induced by XBB.1.5 boosting in humans focus on conserved epitopes targeted by the antecedent mRNA-1273 primary series. Thus, the antibody response to Omicron-based boosters in humans is imprinted by immunizations with historical mRNA-1273 vaccines, but this outcome may be beneficial as it drives expansion of cross-neutralizing antibodies that inhibit infection of emerging SARS-CoV-2 variants and distantly related sarbecoviruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , mRNA Vaccines , Adult , Animals , Female , Humans , Male , Mice , 2019-nCoV Vaccine mRNA-1273/administration & dosage , 2019-nCoV Vaccine mRNA-1273/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , China , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Cross Reactions/immunology , Epitopes, B-Lymphocyte/immunology , mRNA Vaccines/administration & dosage , mRNA Vaccines/genetics , mRNA Vaccines/immunology , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccination
2.
Front Immunol ; 15: 1381508, 2024.
Article in English | MEDLINE | ID: mdl-38690272

ABSTRACT

Seasonal influenza remains a serious global health problem, leading to high mortality rates among the elderly and individuals with comorbidities. Vaccination is generally accepted as the most effective strategy for influenza prevention. While current influenza vaccines are effective, they still have limitations, including narrow specificity for certain serological variants, which may result in a mismatch between vaccine antigens and circulating strains. Additionally, the rapid variability of the virus poses challenges in providing extended protection beyond a single season. Therefore, mRNA technology is particularly promising for influenza prevention, as it enables the rapid development of multivalent vaccines and allows for quick updates of their antigenic composition. mRNA vaccines have already proven successful in preventing COVID-19 by eliciting rapid cellular and humoral immune responses. In this study, we present the development of a trivalent mRNA vaccine candidate, evaluate its immunogenicity using the hemagglutination inhibition assay, ELISA, and assess its efficacy in animals. We demonstrate the higher immunogenicity of the mRNA vaccine candidate compared to the inactivated split influenza vaccine and its enhanced ability to generate a cross-specific humoral immune response. These findings highlight the potential mRNA technology in overcoming current limitations of influenza vaccines and hold promise for ensuring greater efficacy in preventing seasonal influenza outbreaks.


Subject(s)
Immunity, Humoral , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , mRNA Vaccines , Animals , Female , Humans , Mice , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Humoral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice, Inbred BALB C , mRNA Vaccines/administration & dosage , mRNA Vaccines/chemistry , mRNA Vaccines/genetics , mRNA Vaccines/immunology , Seasons , Time Factors , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
3.
J Control Release ; 370: 287-301, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679162

ABSTRACT

The success of lipid nanoparticles (LNPs) in treating COVID-19 promotes further research of mRNA vaccines for cancer vaccination. Aiming at overcoming the constraints of currently available mRNA carriers, various alternative nano-vectors have been developed for delivering tumor antigen encoding mRNA and showed versatility to induce potent anti-tumor immunity. The rationally designed nano-vaccines increase the immune activation capacity of the mRNA vaccines by promoting crucial aspects including mRNA stability, cellular uptake, endosomal escape and targeting of immune cells or organs. Herein, we summarized the research progress of various mRNA based nano-vaccines that have been reported for cancer vaccination, including LNPs, lipid enveloped hybrid nanoparticles, polymeric nanoparticles etc. Several strategies that have been reported for further enhancing the immune stimulation efficacy of mRNA nano-vaccines, including developing nano-vaccines for co-delivering adjuvants, combination of immune checkpoint inhibitors, and optimizing the injection routes for boosting immune responses, have been reviewed. The progress of mRNA nano-vaccines in clinical trials and the prospect of the mRNA vaccines for cancer vaccination are also discussed.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , mRNA Vaccines , Humans , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Neoplasms/therapy , Neoplasms/immunology , Nanoparticles/administration & dosage , Animals , mRNA Vaccines/administration & dosage , RNA, Messenger/administration & dosage , RNA, Messenger/immunology , COVID-19/prevention & control , COVID-19/immunology , Drug Delivery Systems/methods , Lipids/chemistry , Liposomes
4.
Cell ; 187(8): 1822-1822.e1, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38608649

ABSTRACT

In November 2023, Japan's Ministry of Health, Labour and Welfare granted regulatory approval of ARCT-154, a self-amplifying RNA COVID-19 vaccine developed by Arcturus Therapeutics. Clinical trials showed comparable safety and efficacy using a lower dose compared to the mRNA vaccine BNT162b2. To view this Bench-to-Bedside, open or download the PDF.


Subject(s)
COVID-19 Vaccines , mRNA Vaccines , Humans , BNT162 Vaccine , COVID-19/prevention & control , mRNA Vaccines/administration & dosage , mRNA Vaccines/adverse effects
5.
J Neurol ; 271(3): 1063-1071, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38233678

ABSTRACT

BACKGROUND: Case-reports/series and cohorts of Guillain-Barré syndrome (GBS) associated with COVID-19 vaccination have been reported. METHODS: A systematic review and meta-analysis of cohort studies of GBS after COVID-19 vaccination was carried out. Incidence and incidence rate ratio for a number of vaccine doses and risk of GBS, also considering the specific vaccine technology, were calculated in a random-effects model. RESULTS: Of 554 citations retrieved, 518 were discarded as irrelevant. We finally included 15 studies. The random effect model yielded, regardless of the vaccine technology, 1.25 (95%CI 0.21; 2.83) GBS cases per million of COVID-19 vaccine doses, 3.93 (2.54; 5.54) cases per million doses for adenovirus-vectored vaccines and 0.69 (0.38; 1.06) cases per million doses for mRNA vaccines. The GBS risk was 2.6 times increased with the first dose. Regardless of the vaccine technology, the GBS risk was not increased but disaggregating the data it was 2.37 (1.67; 3.36) times increased for adenovirus-vectored vaccines and 0.32 (0.23; 0.47) for mRNA vaccines. Mortality for GBS after vaccination was 0.10 per million doses and 4.6 per GBS cases. CONCLUSIONS: Adenovirus-vectored vaccines showed a 2.4 times increased risk of GBS that was about seven times higher compared with mRNA-based vaccines. The decreased GBS risk associated with mRNA vaccines was possibly due to an elicited reduction of infections, including SARS-CoV-2, associated with GBS during the vaccination period. How adenovirus-vectored COVID-19 vaccines may trigger GBS is unclear and further studies should investigate the relationship between vaccine technologies and GBS risk.


Subject(s)
COVID-19 Vaccines , COVID-19 , Guillain-Barre Syndrome , Humans , COVID-19/prevention & control , COVID-19/complications , COVID-19 Vaccines/adverse effects , Guillain-Barre Syndrome/chemically induced , Guillain-Barre Syndrome/epidemiology , mRNA Vaccines/administration & dosage , mRNA Vaccines/adverse effects , Vaccination/adverse effects
6.
Nature ; 626(7998): 385-391, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096903

ABSTRACT

A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunity, Mucosal , Immunization, Secondary , Macaca mulatta , SARS-CoV-2 , Animals , Humans , Administration, Intranasal , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Cytokines/immunology , Immunity, Mucosal/immunology , Immunization, Secondary/methods , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Injections, Intramuscular , Killer Cells, Natural/immunology , Lung/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology , SARS-CoV-2/classification , SARS-CoV-2/immunology , Trachea/immunology , Trachea/virology
8.
Am J Public Health ; 113(9): 947-951, 2023 09.
Article in English | MEDLINE | ID: mdl-37410982

ABSTRACT

To assess factors associated with timely second-dose completion, we analyzed COVID-19 vaccine data from community-based and mobile vaccine clinics in Maryland. Overall, 85.3% of patients received a timely second dose. Factors associated with a timely second dose included Latino ethnicity (adjusted odds ratio [AOR] = 1.5; 95% confidence interval [CI] = 1.1, 2.0) and receipt of the first dose at community-based vaccine clinics (AOR = 2.1; 95% CI = 1.8, 2.5). Future health initiatives for underserved communities should focus on establishing vaccine clinics in trusted community spaces with culturally sensitive support. (Am J Public Health. 2023;113(9):947-951. https://doi.org/10.2105/AJPH.2023.307338).


Subject(s)
COVID-19 Vaccines , COVID-19 , mRNA Vaccines , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Community Health Centers , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , Maryland/epidemiology , Mobile Health Units , mRNA Vaccines/administration & dosage , mRNA Vaccines/adverse effects , Pandemics
9.
Arch. argent. pediatr ; 121(3): e202202757, jun. 2023. tab
Article in English, Spanish | LILACS, BINACIS | ID: biblio-1436138

ABSTRACT

Los niños cursan mayormente la infección por el virus SARS-CoV-2 en forma leve. Sin embargo, de forma muy infrecuente algunos pueden desarrollar una patología con marcada gravedad denominada síndrome inflamatorio multisistémico en niños relacionado temporalmente con COVID-19 (SIM-C). Dado su reciente surgimiento, aún hay aspectos de su fisiopatología que se desconocen. La posibilidad de recidiva en caso de reinfección o ante la vacunación contra SARS-CoV-2 son nuevos interrogantes a los que nos enfrentamos. Reportamos una serie de casos de 4 pacientes adolescentes que cursaron SIM-C y meses después han sido vacunados contra SARS-CoV-2 con plataformas ARN mensajero (ARNm) sin presentar recurrencia de la enfermedad ni efectos adversos cardiológicos


In most cases, children with SARS-CoV-2 have a mild infection. However, very rarely, some children may develop a severe disease called multisystem inflammatory syndrome in children temporally associated with COVID-19 (MIS-C). Given its recent emergence, some aspects of its pathophysiology are still unknown. The possibility of recurrence in case of reinfection or SARS-CoV-2 vaccination are new questions we are facing. Here we report a case series of 4 adolescent patients who developed MIS-C and, months later, received the SARS-CoV-2 vaccine with messenger RNA (mRNA) platforms without disease recurrence or cardiac adverse events.


Subject(s)
Humans , Male , Female , Adolescent , COVID-19 Vaccines/administration & dosage , COVID-19/complications , COVID-19/prevention & control , Vaccination , SARS-CoV-2 , mRNA Vaccines/administration & dosage
10.
Nat Commun ; 14(1): 1541, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36973247

ABSTRACT

Several studies have reported associations between COVID-19 vaccination and risk of cardiac diseases, especially in young people; the impact on mortality, however, remains unclear. We use national, linked electronic health data in England to assess the impact of COVID-19 vaccination and positive SARS-CoV-2 tests on the risk of cardiac and all-cause mortality in young people (12 to 29 years) using a self-controlled case series design. Here, we show there is no significant increase in cardiac or all-cause mortality in the 12 weeks following COVID-19 vaccination compared to more than 12 weeks after any dose. However, we find an increase in cardiac death in women after a first dose of non mRNA vaccines. A positive SARS-CoV-2 test is associated with increased cardiac and all-cause mortality among people vaccinated or unvaccinated at time of testing.


Subject(s)
COVID-19 Testing , COVID-19 Vaccines , COVID-19 , Cause of Death , SARS-CoV-2 , Vaccination , Adolescent , Adult , Female , Humans , Male , Young Adult , Age Factors , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/mortality , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , Electronic Health Records , England/epidemiology , Heart Diseases/epidemiology , Heart Diseases/mortality , Incidence , mRNA Vaccines/administration & dosage , mRNA Vaccines/adverse effects , Risk Assessment , SARS-CoV-2/isolation & purification , Sex Factors , Time Factors , Vaccination/adverse effects , Child , Hospitalization
12.
Mol Cells ; 46(1): 41-47, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36697236

ABSTRACT

The rapid development of mRNA vaccines has contributed to the management of the current coronavirus disease 2019 (COVID-19) pandemic, suggesting that this technology may be used to manage future outbreaks of infectious diseases. Because the antigens targeted by mRNA vaccines can be easily altered by simply changing the sequence present in the coding region of mRNA structures, it is more appropriate to develop vaccines, especially during rapidly developing outbreaks of infectious diseases. In addition to allowing rapid development, mRNA vaccines have great potential in inducing successful antigen-specific immunity by expressing target antigens in cells and simultaneously triggering immune responses. Indeed, the two COVID-19 mRNA vaccines approved by the U.S. Food and Drug Administration have shown significant efficacy in preventing infections. The ability of mRNAs to produce target proteins that are defective in specific diseases has enabled the development of options to treat intractable diseases. Clinical applications of mRNA vaccines/therapeutics require strategies to safely deliver the RNA molecules into targeted cells. The present review summarizes current knowledge about mRNA vaccines/ therapeutics, their clinical applications, and their delivery strategies.


Subject(s)
COVID-19 Vaccines , mRNA Vaccines , Humans , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/genetics , mRNA Vaccines/administration & dosage , Open Reading Frames , Pandemics , United States
13.
Arch Argent Pediatr ; 121(3): e202202757, 2023 06 01.
Article in English, Spanish | MEDLINE | ID: mdl-36194695

ABSTRACT

In most cases, children with SARS-CoV-2 have a mild infection. However, very rarely, some children may develop a severe disease called multisystem inflammatory syndrome in children temporally associated with COVID-19 (MIS-C). Given its recent emergence, some aspects of its pathophysiology are still unknown. The possibility of recurrence in case of reinfection or SARS-CoV-2 vaccination are new questions we are facing. Here we report a case series of 4 adolescent patients who developed MIS-C and, months later, received the SARS-CoV-2 vaccine with messenger RNA (mRNA) platforms without disease recurrence or cardiac adverse events.


Los niños cursan mayormente la infección por el virus SARS-CoV-2 en forma leve. Sin embargo, de forma muy infrecuente algunos pueden desarrollar una patología con marcada gravedad denominada síndrome inflamatorio multisistémico en niños relacionado temporalmente con COVID-19 (SIM-C). Dado su reciente surgimiento, aún hay aspectos de su fisiopatología que se desconocen. La posibilidad de recidiva en caso de reinfección o ante la vacunación contra SARS-CoV-2 son nuevos interrogantes a los que nos enfrentamos. Reportamos una serie de casos de 4 pacientes adolescentes que cursaron SIM-C y meses después han sido vacunados contra SARS-CoV-2 con plataformas ARN mensajero (ARNm) sin presentar recurrencia de la enfermedad ni efectos adversos cardiológicos.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adolescent , Humans , COVID-19/complications , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , mRNA Vaccines/administration & dosage , SARS-CoV-2 , Vaccination
14.
Science ; 376(6594): 680-681, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549401

ABSTRACT

New delivery systems aim to increase vaccine potency and reduce side effects.


Subject(s)
Lipids , Nanoparticles , mRNA Vaccines , Humans , Lipids/chemistry , Nanoparticles/adverse effects , Nanoparticles/chemistry , mRNA Vaccines/administration & dosage , mRNA Vaccines/chemistry
15.
Nature ; 607(7917): 128-134, 2022 07.
Article in English | MEDLINE | ID: mdl-35447027

ABSTRACT

The Omicron variant of SARS-CoV-2 infected many vaccinated and convalescent individuals1-3. Despite the reduced protection from infection, individuals who received three doses of an mRNA vaccine were highly protected from more serious consequences of infection4. Here we examine the memory B cell repertoire in a longitudinal cohort of individuals receiving three mRNA vaccine doses5,6. We find that the third dose is accompanied by an increase in, and evolution of, receptor-binding domain (RBD)-specific memory B cells. The increase is due to expansion of memory B cell clones that were present after the second dose as well as the emergence of new clones. The antibodies encoded by these cells showed significantly increased potency and breadth when compared with antibodies obtained after the second dose. Notably, the increase in potency was especially evident among newly developing clones of memory cells, which differed from persisting clones in targeting more conserved regions of the RBD. Overall, more than 50% of the analysed neutralizing antibodies in the memory compartment after the third mRNA vaccine dose neutralized the Omicron variant. Thus, individuals receiving three doses of an mRNA vaccine have a diverse memory B cell repertoire that can respond rapidly and produce antibodies capable of clearing even diversified variants such as Omicron. These data help to explain why a third dose of a vaccine that was not specifically designed to protect against variants is effective against variant-induced serious disease.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Memory B Cells , SARS-CoV-2 , mRNA Vaccines , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Humans , Memory B Cells/immunology , RNA, Messenger/genetics , SARS-CoV-2/genetics , SARS-CoV-2/immunology , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology
16.
Nat Commun ; 13(1): 1162, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246536

ABSTRACT

Mass vaccination is effective in reducing SARS-CoV-2 infections among vaccinated individuals. However, it remains unclear how effectively COVID-19 vaccines prevent people from spreading the virus to their close contacts. Using nationwide administrative datasets on SARS-CoV-2 infections, vaccination records, demographics, and unique household IDs, we conducted an observational cohort study to estimate the direct and indirect effectiveness of mRNA-based COVID-19 vaccines in reducing infections among vaccinated healthcare workers and their unvaccinated household members. Our estimates for adults imply indirect effectiveness of 39.1% (95% CI: -7.1% to 65.3%) two weeks and 39.0% (95% CI: 18.9% to 54.0%) eight weeks after the second dose. We find that the indirect effect of mRNA-based COVID-19 vaccines within households is smaller for unvaccinated children than for adults and statistically insignificant. Here, we show that mRNA-based COVID-19 vaccines are associated with a reduction in SARS-CoV-2 infections not only among vaccinated individuals but also among unvaccinated adult household members in a real-world setting.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Health Personnel/statistics & numerical data , SARS-CoV-2/immunology , Vaccination/statistics & numerical data , mRNA Vaccines/immunology , Adolescent , Adult , Aged , COVID-19/epidemiology , COVID-19/virology , COVID-19 Vaccines/administration & dosage , Child , Cohort Studies , Female , Finland/epidemiology , Humans , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Pandemics/prevention & control , Registries/statistics & numerical data , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Vaccination/methods , Young Adult , mRNA Vaccines/administration & dosage
17.
Cell Rep ; 38(9): 110429, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35216664

ABSTRACT

Continuous emergence of SARS-CoV-2 variants of concern (VOCs) is fueling the COVID-19 pandemic. Omicron (B.1.1.529) rapidly spread worldwide. The large number of mutations in its Spike raise concerns about a major antigenic drift that could significantly decrease vaccine efficacy and infection-induced immunity. A long interval between BNT162b2 mRNA doses elicits antibodies that efficiently recognize Spikes from different VOCs. Here, we evaluate the recognition of Omicron Spike by plasma from a cohort of SARS-CoV-2 naive and previously infected individuals who received their BNT162b2 mRNA vaccine 16 weeks apart. Omicron Spike is recognized less efficiently than D614G, Alpha, Beta, Gamma, and Delta Spikes. We compare with plasma activity from participants receiving a short (4 weeks) interval regimen. Plasma from individuals of the long-interval cohort recognize and neutralize better the Omicron Spike compared with those who received a short interval. Whether this difference confers any clinical benefit against Omicron remains unknown.


Subject(s)
Antibodies, Neutralizing/blood , BNT162 Vaccine/administration & dosage , Immunization Schedule , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adult , Aged , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Antibodies, Viral/analysis , Antibodies, Viral/blood , Antibodies, Viral/immunology , BNT162 Vaccine/immunology , Cohort Studies , Female , HEK293 Cells , Humans , Immunization, Secondary/methods , Male , Middle Aged , Quebec , SARS-CoV-2/pathogenicity , Time Factors , Vaccination/methods , Vaccine Potency , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Young Adult , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology
18.
J Phys Chem Lett ; 13(5): 1314-1322, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35107010

ABSTRACT

With the global outbreak of SARS-CoV-2, mRNA vaccines became the first type of COVID-19 vaccines to enter clinical trials because of their facile production, low cost, and relative safety, which initiated great advances in mRNA therapeutic techniques. However, the development of mRNA therapeutic techniques still confronts some challenges. First, in vitro transcribed mRNA molecules can be easily degraded by ribonuclease (RNase), resulting in their low stability. Next, the negative charge of mRNA molecules prevents them from direct cell entry. Therefore, finding efficient and safe delivery technology could be the key issue to improve mRNA therapeutic techniques. In this Perspective, we mainly discuss the problems of the existing mRNA-based delivery nanoplatforms, including safety evaluation, administration routes, and preparation technology. Moreover, we also propose some views on strategies to further improve mRNA delivery technology.


Subject(s)
COVID-19 Vaccines/administration & dosage , Nanoparticle Drug Delivery System , RNA, Messenger/administration & dosage , Vaccines, Synthetic/administration & dosage , mRNA Vaccines/administration & dosage , Drug Stability , Drug Storage , High-Throughput Screening Assays , Humans , Vaccine Development
20.
MMWR Morb Mortal Wkly Rep ; 71(7): 255-263, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35176007

ABSTRACT

CDC recommends that all persons aged ≥12 years receive a booster dose of COVID-19 mRNA vaccine ≥5 months after completion of a primary mRNA vaccination series and that immunocompromised persons receive a third primary dose.* Waning of vaccine protection after 2 doses of mRNA vaccine has been observed during the period of the SARS-CoV-2 B.1.617.2 (Delta) variant predominance† (1-5), but little is known about durability of protection after 3 doses during periods of Delta or SARS-CoV-2 B.1.1.529 (Omicron) variant predominance. A test-negative case-control study design using data from eight VISION Network sites§ examined vaccine effectiveness (VE) against COVID-19 emergency department/urgent care (ED/UC) visits and hospitalizations among U.S. adults aged ≥18 years at various time points after receipt of a second or third vaccine dose during two periods: Delta variant predominance and Omicron variant predominance (i.e., periods when each variant accounted for ≥50% of sequenced isolates).¶ Persons categorized as having received 3 doses included those who received a third dose in a primary series or a booster dose after a 2 dose primary series (including the reduced-dosage Moderna booster). The VISION Network analyzed 241,204 ED/UC encounters** and 93,408 hospitalizations across 10 states during August 26, 2021-January 22, 2022. VE after receipt of both 2 and 3 doses was lower during the Omicron-predominant than during the Delta-predominant period at all time points evaluated. During both periods, VE after receipt of a third dose was higher than that after a second dose; however, VE waned with increasing time since vaccination. During the Omicron period, VE against ED/UC visits was 87% during the first 2 months after a third dose and decreased to 66% among those vaccinated 4-5 months earlier; VE against hospitalizations was 91% during the first 2 months following a third dose and decreased to 78% ≥4 months after a third dose. For both Delta- and Omicron-predominant periods, VE was generally higher for protection against hospitalizations than against ED/UC visits. All eligible persons should remain up to date with recommended COVID-19 vaccinations to best protect against COVID-19-associated hospitalizations and ED/UC visits.


Subject(s)
Ambulatory Care/statistics & numerical data , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Hospitalization/statistics & numerical data , SARS-CoV-2/immunology , Vaccine Efficacy , mRNA Vaccines/administration & dosage , Adult , Aged , Aged, 80 and over , Case-Control Studies , Emergency Service, Hospital , Female , Humans , Male , Middle Aged , Time Factors , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...