LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 36

Search options

  1. Article ; Online: The safety and immunogenicity of two Zika virus mRNA vaccine candidates in healthy flavivirus baseline seropositive and seronegative adults: the results of two randomised, placebo-controlled, dose-ranging, phase 1 clinical trials.

    Essink, Brandon / Chu, Laurence / Seger, William / Barranco, Elizabeth / Le Cam, Nancy / Bennett, Hamilton / Faughnan, Veronica / Pajon, Rolando / Paila, Yamuna D / Bollman, Brooke / Wang, Steven / Dooley, Jacqueline / Kalidindi, Shiva / Leav, Brett

    The Lancet. Infectious diseases

    2023  Volume 23, Issue 5, Page(s) 621–633

    Abstract: Background: Developing a safe and immunogenic vaccine against Zika virus remains an unmet medical need. We did two phase 1 studies that evaluated the safety and immunogenicity of two mRNA-based Zika virus vaccines (mRNA-1325 and mRNA-1893) in adults.: ...

    Abstract Background: Developing a safe and immunogenic vaccine against Zika virus remains an unmet medical need. We did two phase 1 studies that evaluated the safety and immunogenicity of two mRNA-based Zika virus vaccines (mRNA-1325 and mRNA-1893) in adults.
    Methods: Two randomised, placebo-controlled, dose-ranging, multicentre, phase 1 trials, one of mRNA-1325 (mRNA-1325 trial) and one of mRNA-1893 (mRNA-1893 trial), were done. For both studies, eligible participants were healthy adults (aged 18-49 years) who were flavivirus seronegative or flavivirus seropositive at baseline. Participants in the mRNA-1325 trial, which was done at three centres in the USA, were randomly assigned centrally (1:4), using a randomisation table, to the placebo group or one of three mRNA-1325 dose groups (10, 25, or 100 μg). All participants received two doses. The mRNA-1325 vaccine encoded the premembrane and envelope E structural proteins (prME) from a Micronesia 2007 Zika virus isolate. Participants in the mRNA-1893 trial, which was done at three centres in the USA and one centre in Puerto Rico, were randomly assigned (1:4) to the placebo group or one of four mRNA-1893 dose groups (10, 30, 100, or 250 μg) using centralised interactive response technology. All participants in the mRNA-1893 trial received dose one on day 1 and then dose two on day 29. The mRNA-1893 vaccine encoded the prME from the RIO-U1 Zika virus isolate. Safety was the primary outcome of each study, which was evaluated in the respective safety populations (mRNA-1325 trial: participants who received at least one dose and provided safety data; mRNA-1893 trial: participants who received at least one dose) and the solicited safety population (mRNA-1893 trial only: received at least 1 dose and contributed solicited adverse reaction data). Endpoints in both trials included solicited adverse reactions within 7 days after vaccination and unsolicited adverse events within 28 days after vaccination. The secondary outcome of both trials was immunogenicity assessed by Zika virus-specific neutralising antibodies (nAbs) in the per-protocol populations in either trial (participants with no major protocol deviations received full dose[s] of assigned dose level within the acceptable time window, had samples drawn within acceptable time window, and had prevaccination and corresponding post-vaccination serum samples for testing). These were descriptive studies, with no formal hypothesis testing in either trial. Both trials are registered with ClinicalTrials.gov, NCT03014089 (mRNA-1325 trial) and NCT04064905 (mRNA-1893 trial).
    Findings: The mRNA-1325 trial was done from Dec 14, 2016, to Aug 16, 2018. 90 participants were enrolled: 53 (59%) participants were women and 37 (41%) were men; 84 (93%) were White; and 74 (82%) were not Hispanic or Latino. All three dose levels of mRNA-1325 (10, 25, and 100 μg) were generally well tolerated, but the vaccine elicited poor Zika virus-specific nAb responses. At 28 days after dose two, geometric mean titres (GMTs) were highest for mRNA-1325 10 μg (10·3 [95% CI 5·9-18·2]). The mRNA-1893 trial was done from July 23, 2019, to March 22, 2021. 120 participants (70 [58%] women and 50 [42%] men) were enrolled, most participants were White (89 [74%]), and not Hispanic or Latino (91 [76%]). In the mRNA-1893 trial, solicited adverse reactions in participants who received a vaccine were mostly grade 1 or 2 and occurred more frequently at higher dose levels and after dose two. No participants withdrew due to an unsolicited treatment-emergent adverse event and most of these events were not treatment related. On day 57, all evaluated mRNA-1893 dose levels induced robust Zika virus-specific nAb responses, independent of flavivirus serostatus, that persisted until month 13. At day 57 in participants who were flavivirus seronegative, plaque reduction neutralisation titre test nAb GMTs were highest for mRNA-1893 100 μg (454·2 [330·0-619·6]); in participants who were flavivirus seropositive, GMTs were highest for mRNA-1893 10 μg (224·1 [43·5-1153·5]) and mRNA-1893 100 μg (190·5 [19·2-1887·2]).
    Interpretation: These findings support the continued development of mRNA-1893 against Zika virus, which was well tolerated at all evaluated dose levels and induced strong Zika virus-specific serum nAb responses after two doses, regardless of baseline flavivirus serostatus.
    Funding: Biomedical Advanced Research and Development Authority and Moderna.
    MeSH term(s) Male ; Adult ; Humans ; Female ; Zika Virus/genetics ; Flavivirus ; Double-Blind Method ; Vaccination ; Puerto Rico ; Immunogenicity, Vaccine ; Zika Virus Infection/prevention & control ; Antibodies, Viral
    Chemical Substances Antibodies, Viral
    Language English
    Publishing date 2023-01-19
    Publishing country United States
    Document type Randomized Controlled Trial ; Clinical Trial, Phase I ; Journal Article ; Research Support, U.S. Gov't, Non-P.H.S. ; Research Support, Non-U.S. Gov't
    ZDB-ID 2061641-7
    ISSN 1474-4457 ; 1473-3099
    ISSN (online) 1474-4457
    ISSN 1473-3099
    DOI 10.1016/S1473-3099(22)00764-2
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: A preliminary report of a randomized controlled phase 2 trial of the safety and immunogenicity of mRNA-1273 SARS-CoV-2 vaccine.

    Chu, Laurence / McPhee, Roderick / Huang, Wenmei / Bennett, Hamilton / Pajon, Rolando / Nestorova, Biliana / Leav, Brett

    Vaccine

    2021  Volume 39, Issue 20, Page(s) 2791–2799

    Abstract: Background: Vaccines are urgently needed to prevent the global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We assessed the safety and immunogenicity of vaccine candidate mRNA-1273, encoding the prefusion-stabilized spike ... ...

    Abstract Background: Vaccines are urgently needed to prevent the global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We assessed the safety and immunogenicity of vaccine candidate mRNA-1273, encoding the prefusion-stabilized spike protein of SARS-CoV-2.
    Methods: This phase 2, randomized, observer-blind, placebo-controlled trial was conducted at 8 sites in the USA, in healthy adults aged ≥18 years with no known history or risk of SARS-CoV-2 infection, and had not previously received an investigational CoV vaccine or treatment. Participants were stratified into two age cohorts (≥18-<55 and ≥55) and were randomly assigned (1:1:1) to either 50 or 100 µg of mRNA-1273, or placebo administered as two intramuscular injections 28 days apart. The primary outcomes were safety, reactogenicity, and immunogenicity assessed by anti-SARS-CoV-2-spike binding antibody level (bAb). Secondary outcome was immunogenicity assessed by SARS-CoV-2 neutralizing antibody (nAb) response.
    Results: Between 29 May and 8 July 2020, 600 participants were randomized, 300 per age cohort. The most common solicited adverse reactions were pain at injection site, headache, and fatigue following each vaccination in both age cohorts. One serious adverse event deemed unrelated by the site investigator occurred 33 days post-vaccination one. mRNA-1273 induced bAb and nAb by 28 days post-vaccination one that were higher at the 100 µg dose relative to the 50 µg dose; this difference was less apparent post-vaccination two. Binding antibodies and nAb increased substantially by 14 days following the second vaccination (day 43) to levels exceeding those of convalescent sera and remained elevated through day 57.
    Conclusions: Vaccination with mRNA-1273 resulted in significant immune responses to SARS-CoV-2 in participants 18 years and older, with an acceptable safety profile, confirming the safety and immunogenicity of 50 and 100 µg mRNA-1273 given as a 2 dose-regimen. ClinicalTrials.gov; NCT04405076.
    MeSH term(s) Adolescent ; Adult ; Antibodies, Viral ; COVID-19/therapy ; COVID-19 Vaccines ; Double-Blind Method ; Humans ; Immunization, Passive ; Immunogenicity, Vaccine ; RNA, Messenger ; SARS-CoV-2 ; Vaccines ; COVID-19 Serotherapy
    Chemical Substances Antibodies, Viral ; COVID-19 Vaccines ; RNA, Messenger ; Vaccines
    Language English
    Publishing date 2021-02-09
    Publishing country Netherlands
    Document type Clinical Trial, Phase II ; Journal Article ; Randomized Controlled Trial ; Research Support, N.I.H., Extramural
    ZDB-ID 605674-x
    ISSN 1873-2518 ; 0264-410X
    ISSN (online) 1873-2518
    ISSN 0264-410X
    DOI 10.1016/j.vaccine.2021.02.007
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: An optimized messenger RNA vaccine candidate protects non-human primates from Zika virus infection.

    Bollman, Brooke / Nunna, Naveen / Bahl, Kapil / Hsiao, Chiaowen Joyce / Bennett, Hamilton / Butler, Scott / Foreman, Bryant / Burgomaster, Katherine E / Aleshnick, Maya / Kong, Wing-Pui / Fisher, Brian E / Ruckwardt, Tracy J / Morabito, Kaitlyn M / Graham, Barney S / Dowd, Kimberly A / Pierson, Theodore C / Carfi, Andrea

    NPJ vaccines

    2023  Volume 8, Issue 1, Page(s) 58

    Abstract: Zika virus (ZIKV), an arbovirus transmitted by mosquitoes, was identified as a cause of congenital disease during a major outbreak in the Americas in 2016. Vaccine design strategies relied on limited available isolate sequence information due to the ... ...

    Abstract Zika virus (ZIKV), an arbovirus transmitted by mosquitoes, was identified as a cause of congenital disease during a major outbreak in the Americas in 2016. Vaccine design strategies relied on limited available isolate sequence information due to the rapid response necessary. The first-generation ZIKV mRNA vaccine, mRNA-1325, was initially generated and, as additional strain sequences became available, a second mRNA vaccine, mRNA-1893, was developed. Herein, we compared the immune responses following mRNA-1325 and mRNA-1893 vaccination and reported that mRNA-1893 generated comparable neutralizing antibody titers to mRNA-1325 at 1/20th of the dose and provided complete protection from ZIKV challenge in non-human primates. In-depth characterization of these vaccines indicated that the observed immunologic differences could be attributed to a single amino acid residue difference that compromised mRNA-1325 virus-like particle formation.
    Language English
    Publishing date 2023-04-20
    Publishing country England
    Document type Journal Article
    ISSN 2059-0105
    ISSN (online) 2059-0105
    DOI 10.1038/s41541-023-00656-4
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Serum Neutralizing Activity of mRNA-1273 against SARS-CoV-2 Variants.

    Choi, Angela / Koch, Matthew / Wu, Kai / Dixon, Groves / Oestreicher, Judy / Legault, Holly / Stewart-Jones, Guillaume B E / Colpitts, Tonya / Pajon, Rolando / Bennett, Hamilton / Carfi, Andrea / Edwards, Darin K

    Journal of virology

    2021  Volume 95, Issue 23, Page(s) e0131321

    Abstract: The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to growing concerns over increased transmissibility and the ability of some variants to partially escape immunity. Sera from participants immunized on a prime- ... ...

    Abstract The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to growing concerns over increased transmissibility and the ability of some variants to partially escape immunity. Sera from participants immunized on a prime-boost schedule with the mRNA-1273 COVID-19 vaccine were tested for neutralizing activity against several SARS-CoV-2 variants, including variants of concern (VOCs) and variants of interest (VOIs), compared to neutralization of the wild-type SARS-CoV-2 virus (designated D614G). Results showed minimal, statistically nonsignificant effects on neutralization titers against the B.1.1.7 (Alpha) variant (1.2-fold reduction compared with D614G); other VOCs, such as B.1.351 (Beta, including B.1.351-v1, B.1.351-v2, and B.1.351-v3), P.1 (Gamma), and B.1.617.2 (Delta), showed significantly decreased neutralization titers ranging from 2.1-fold to 8.4-fold reductions compared with D614G, although all remained susceptible to mRNA-1273-elicited serum neutralization.
    MeSH term(s) Adult ; Antibodies, Neutralizing/immunology ; Antibodies, Viral/immunology ; COVID-19/immunology ; COVID-19/prevention & control ; COVID-19 Vaccines/immunology ; Female ; Humans ; Male ; Mutation ; Neutralization Tests ; Pandemics/prevention & control ; SARS-CoV-2/immunology ; Spike Glycoprotein, Coronavirus/immunology ; Vaccination
    Chemical Substances Antibodies, Neutralizing ; Antibodies, Viral ; COVID-19 Vaccines ; Spike Glycoprotein, Coronavirus ; mRNA-1273 vaccine (EPK39PL4R4)
    Language English
    Publishing date 2021-09-22
    Publishing country United States
    Document type Clinical Trial, Phase I ; Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, P.H.S.
    ZDB-ID 80174-4
    ISSN 1098-5514 ; 0022-538X
    ISSN (online) 1098-5514
    ISSN 0022-538X
    DOI 10.1128/JVI.01313-21
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants.

    Wu, Kai / Werner, Anne P / Moliva, Juan I / Koch, Matthew / Choi, Angela / Stewart-Jones, Guillaume B E / Bennett, Hamilton / Boyoglu-Barnum, Seyhan / Shi, Wei / Graham, Barney S / Carfi, Andrea / Corbett, Kizzmekia S / Seder, Robert A / Edwards, Darin K

    bioRxiv : the preprint server for biology

    2021  

    Abstract: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative infection of a global pandemic that has led to more than 2 million deaths worldwide. The Moderna mRNA-1273 vaccine has demonstrated ~94% efficacy in a Phase 3 study and has ... ...

    Abstract Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative infection of a global pandemic that has led to more than 2 million deaths worldwide. The Moderna mRNA-1273 vaccine has demonstrated ~94% efficacy in a Phase 3 study and has been approved under Emergency Use Authorization. The emergence of SARS-CoV-2 variants with mutations in the spike protein, most recently circulating isolates from the United Kingdom (B.1.1.7) and Republic of South Africa (B.1.351), has led to lower neutralization from convalescent serum by pseudovirus neutralization (PsVN) assays and resistance to certain monoclonal antibodies. Here, using two orthogonal VSV and lentivirus PsVN assays expressing spike variants of 20E (EU1), 20A.EU2, D614G-N439, mink cluster 5, B.1.1.7, and B.1.351 variants, we assessed the neutralizing capacity of sera from human subjects or non-human primates (NHPs) that received mRNA-1273. No significant impact on neutralization against the B.1.1.7 variant was detected in either case, however reduced neutralization was measured against the mutations present in B.1.351. Geometric mean titer (GMT) of human sera from clinical trial participants in VSV PsVN assay using D614G spike was 1/1852. VSV pseudoviruses with spike containing K417N-E484K-N501Y-D614G and full B.1.351 mutations resulted in 2.7 and 6.4-fold GMT reduction, respectively, when compared to the D614G VSV pseudovirus. Importantly, the VSV PsVN GMT of these human sera to the full B.1.351 spike variant was still 1/290, with all evaluated sera able to fully neutralize. Similarly, sera from NHPs immunized with 30 or 100μg of mRNA-1273 had VSV PsVN GMTs of ~ 1/323 or 1/404, respectively, against the full B.1.351 spike variant with a ~ 5 to 10-fold reduction compared to D614G. Individual mutations that are characteristic of the B.1.1.7 and B.1.351 variants had a similar impact on neutralization when tested in VSV or in lentivirus PsVN assays. Despite the observed decreases, the GMT of VSV PsVN titers in human vaccinee sera against the B.1.351 variant remained at ~1/300. Taken together these data demonstrate reduced but still significant neutralization against the full B.1.351 variant following mRNA-1273 vaccination.
    Language English
    Publishing date 2021-01-25
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2021.01.25.427948
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Serum Neutralizing Activity Elicited by mRNA-1273 Vaccine.

    Wu, Kai / Werner, Anne P / Koch, Matthew / Choi, Angela / Narayanan, Elisabeth / Stewart-Jones, Guillaume B E / Colpitts, Tonya / Bennett, Hamilton / Boyoglu-Barnum, Seyhan / Shi, Wei / Moliva, Juan I / Sullivan, Nancy J / Graham, Barney S / Carfi, Andrea / Corbett, Kizzmekia S / Seder, Robert A / Edwards, Darin K

    The New England journal of medicine

    2021  Volume 384, Issue 15, Page(s) 1468–1470

    MeSH term(s) 2019-nCoV Vaccine mRNA-1273 ; Antibodies, Neutralizing/blood ; Antibodies, Viral/blood ; COVID-19/immunology ; COVID-19/prevention & control ; COVID-19 Vaccines/immunology ; Humans ; Mutation ; SARS-CoV-2/genetics ; SARS-CoV-2/immunology ; Spike Glycoprotein, Coronavirus/genetics ; Spike Glycoprotein, Coronavirus/immunology ; Statistics, Nonparametric ; Vaccines, Synthetic/immunology ; mRNA Vaccines
    Chemical Substances Antibodies, Neutralizing ; Antibodies, Viral ; COVID-19 Vaccines ; Spike Glycoprotein, Coronavirus ; Vaccines, Synthetic ; spike protein, SARS-CoV-2 ; 2019-nCoV Vaccine mRNA-1273 (EPK39PL4R4)
    Language English
    Publishing date 2021-03-17
    Publishing country United States
    Document type Clinical Trial, Phase I ; Letter
    ZDB-ID 207154-x
    ISSN 1533-4406 ; 0028-4793
    ISSN (online) 1533-4406
    ISSN 0028-4793
    DOI 10.1056/NEJMc2102179
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: An mpox virus mRNA-lipid nanoparticle vaccine confers protection against lethal orthopoxviral challenge.

    Freyn, Alec W / Atyeo, Caroline / Earl, Patricia L / Americo, Jeffrey L / Chuang, Gwo-Yu / Natarajan, Harini / Frey, Tiffany R / Gall, Jason G / Moliva, Juan I / Hunegnaw, Ruth / Asthagiri Arunkumar, Guha / Ogega, Clinton O / Nasir, Arshan / Santos, Genesis / Levin, Rafael H / Meni, Anusha / Jorquera, Patricia A / Bennett, Hamilton / Johnson, Joshua A /
    Durney, Michael A / Stewart-Jones, Guillaume / Hooper, Jay W / Colpitts, Tonya M / Alter, Galit / Sullivan, Nancy J / Carfi, Andrea / Moss, Bernard

    Science translational medicine

    2023  Volume 15, Issue 716, Page(s) eadg3540

    Abstract: Mpox virus (MPXV) caused a global outbreak in 2022. Although smallpox vaccines were rapidly deployed to curb spread and disease among those at highest risk, breakthrough disease was noted after complete immunization. Given the threat of additional ... ...

    Abstract Mpox virus (MPXV) caused a global outbreak in 2022. Although smallpox vaccines were rapidly deployed to curb spread and disease among those at highest risk, breakthrough disease was noted after complete immunization. Given the threat of additional zoonotic events and the virus's evolving ability to drive human-to-human transmission, there is an urgent need for an MPXV-specific vaccine that confers protection against evolving MPXV strains and related orthopoxviruses. Here, we demonstrate that an mRNA-lipid nanoparticle vaccine encoding a set of four highly conserved MPXV surface proteins involved in virus attachment, entry, and transmission can induce MPXV-specific immunity and heterologous protection against a lethal vaccinia virus (VACV) challenge. Compared with modified vaccinia virus Ankara (MVA), which forms the basis for the current MPXV vaccine, immunization with an mRNA-based MPXV vaccine generated superior neutralizing activity against MPXV and VACV and more efficiently inhibited spread between cells. We also observed greater Fc effector T
    MeSH term(s) Humans ; Monkeypox virus/genetics ; Vaccinia virus/genetics ; Smallpox Vaccine/genetics ; Viral Vaccines ; Antigens, Viral ; RNA, Messenger/genetics
    Chemical Substances Lipid Nanoparticles ; Smallpox Vaccine ; Viral Vaccines ; Antigens, Viral ; RNA, Messenger
    Language English
    Publishing date 2023-10-04
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2518854-9
    ISSN 1946-6242 ; 1946-6234
    ISSN (online) 1946-6242
    ISSN 1946-6234
    DOI 10.1126/scitranslmed.adg3540
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Serum Neutralizing Activity of mRNA-1273 against SARS-CoV-2 Variants

    Choi, Angela / Koch, Matthew / Wu, Kai / Dixon, Groves / Oestreicher, Judith / Legault, Holly / Stewart-Jones, Guillaume B.E. / Colpitts, Tonya / Pajon, Rolando / Bennett, Hamilton / Carfi, Andrea / Edwards, Darin K

    bioRxiv

    Abstract: The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to growing concerns over increased transmissibility and the ability of some variants to partially escape immunity. Sera from participants immunized on a prime- ... ...

    Abstract The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to growing concerns over increased transmissibility and the ability of some variants to partially escape immunity. Sera from participants immunized on a prime-boost schedule with the mRNA-1273 COVID-19 vaccine were tested for neutralizing activity against several SARS-CoV-2 variants, including variants of concern (VOCs) and variants of interest (VOIs), compared to neutralization of the wild-type SARS-CoV-2 virus (designated as D614G). Results showed minimal effects on neutralization titers against the B.1.1.7 (Alpha) variant (1.2-fold reduction compared with D614G); other VOCs such as B.1.351 (Beta, including B.1.351-v1, B.1.351-v2, and B.1.351-v3), B.1.617.2 (Delta), and P.1 (Gamma) showed decreased neutralization titers ranging from 2.1-fold to 8.4-fold reductions compared with D614G, although all remained susceptible to mRNA-1273-elicited serum neutralization.
    Keywords covid19
    Language English
    Publishing date 2021-06-28
    Publisher Cold Spring Harbor Laboratory
    Document type Article ; Online
    DOI 10.1101/2021.06.28.449914
    Database COVID19

    Kategorien

  9. Article: Variant SARS-CoV-2 mRNA vaccines confer broad neutralization as primary or booster series in mice

    Wu, Kai / Choi, Angela / Koch, Matthew / Elbashir, Sayda / Ma, LingZhi / Lee, Diana / Woods, Angela / Henry, Carole / Palandjian, Charis / Hill, Anna / Jani, Hardik / Quinones, Julian / Nunna, Naveen / O'Connell, Sarah / McDermott, Adrian B. / Falcone, Samantha / Narayanan, Elisabeth / Colpitts, Tonya / Bennett, Hamilton /
    Corbett, Kizzmekia S. / Seder, Robert / Graham, Barney S. / Stewart-Jones, Guillaume B.E. / Carfi, Andrea / Edwards, Darin K.

    Vaccine. 2021 Dec. 17, v. 39, no. 51

    2021  

    Abstract: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic. Safe and effective COVID-19 vaccines are now available, including mRNA-1273, which has shown 94% efficacy in prevention of symptomatic COVID-19 ... ...

    Abstract Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic. Safe and effective COVID-19 vaccines are now available, including mRNA-1273, which has shown 94% efficacy in prevention of symptomatic COVID-19 disease. However, the emergence of SARS-CoV-2 variants has led to concerns of viral escape from vaccine-induced immunity. Several variants have shown decreased susceptibility to neutralization by vaccine-induced immunity, most notably B.1.351 (Beta), although the overall impact on vaccine efficacy remains to be determined. Here, we present the initial evaluation in mice of 2 updated mRNA vaccines designed to target SARS-CoV-2 variants: (1) monovalent mRNA-1273.351 encodes for the spike protein found in B.1.351 and (2) mRNA-1273.211 comprising a 1:1 mix of mRNA-1273 and mRNA-1273.351. Both vaccines were evaluated as a 2-dose primary series in mice; mRNA-1273.351 was also evaluated as a booster dose in animals previously vaccinated with mRNA-1273. The results demonstrated that a primary vaccination series of mRNA-1273.351 was effective at increasing neutralizing antibody titers against B.1.351, while mRNA-1273.211 was effective at providing broad cross-variant neutralization. A third (booster) dose of mRNA-1273.351 significantly increased both wild-type and B.1.351-specific neutralization titers. Both mRNA-1273.351 and mRNA-1273.211 are being evaluated in pre-clinical challenge and clinical studies.
    Keywords COVID-19 infection ; Severe acute respiratory syndrome coronavirus 2 ; etiological agents ; immunity ; neutralization ; pandemic ; vaccination ; vaccines
    Language English
    Dates of publication 2021-1217
    Size p. 7394-7400.
    Publishing place Elsevier Ltd
    Document type Article
    ZDB-ID 605674-x
    ISSN 1873-2518 ; 0264-410X
    ISSN (online) 1873-2518
    ISSN 0264-410X
    DOI 10.1016/j.vaccine.2021.11.001
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  10. Article: Variant SARS-CoV-2 mRNA vaccines confer broad neutralization as primary or booster series in mice.

    Wu, Kai / Choi, Angela / Koch, Matthew / Elbashir, Sayda / Ma, LingZhi / Lee, Diana / Woods, Angela / Henry, Carole / Palandjian, Charis / Hill, Anna / Jani, Hardik / Quinones, Julian / Nunna, Naveen / Oâ Connell, Sarah / McDermott, Adrian B / Falcone, Samantha / Narayanan, Elisabeth / Colpitts, Tonya / Bennett, Hamilton /
    Corbett, Kizzmekia S / Seder, Robert / Graham, Barney S / Stewart-Jones, Guillaume Be / Carfi, Andrea / Edwards, Darin K

    bioRxiv : the preprint server for biology

    2021  

    Abstract: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic. Safe and effective COVID-19 vaccines are now available, including mRNA-1273, which has shown 94% efficacy in prevention of symptomatic COVID-19 ... ...

    Abstract Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic. Safe and effective COVID-19 vaccines are now available, including mRNA-1273, which has shown 94% efficacy in prevention of symptomatic COVID-19 disease. However, the emergence of SARS-CoV-2 variants has led to concerns of viral escape from vaccine-induced immunity. Several variants have shown decreased susceptibility to neutralization by vaccine-induced immunity, most notably B.1.351 (Beta), although the overall impact on vaccine efficacy remains to be determined. Here, we present the initial evaluation in mice of 2 updated mRNA vaccines designed to target SARS-CoV-2 variants: (1) monovalent mRNA-1273.351 encodes for the spike protein found in B.1.351 and (2) mRNA-1273.211 comprising a 1:1 mix of mRNA-1273 and mRNA-1273.351. Both vaccines were evaluated as a 2-dose primary series in mice; mRNA-1273.351 was also evaluated as a booster dose in animals previously vaccinated with mRNA-1273. The results demonstrated that a primary vaccination series of mRNA-1273.351 was effective at increasing neutralizing antibody titers against B.1.351, while mRNA-1273.211 was effective at providing broad cross-variant neutralization. A third (booster) dose of mRNA-1273.351 significantly increased both wild-type and B.1.351-specific neutralization titers. Both mRNA-1273.351 and mRNA-1273.211 are being evaluated in pre-clinical challenge and clinical studies.
    Language English
    Publishing date 2021-10-07
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2021.04.13.439482
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top