LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 3 of total 3

Search options

  1. Article ; Online: Low-dose self-amplifying mRNA COVID-19 vaccine drives strong protective immunity in non-human primates against SARS-CoV-2 infection.

    Rappaport, Amy R / Hong, Sue-Jean / Scallan, Ciaran D / Gitlin, Leonid / Akoopie, Arvin / Boucher, Gregory R / Egorova, Milana / Espinosa, J Aaron / Fidanza, Mario / Kachura, Melissa A / Shen, Annie / Sivko, Gloria / Van Abbema, Anne / Veres, Robert L / Jooss, Karin

    Nature communications

    2022  Volume 13, Issue 1, Page(s) 3289

    Abstract: The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self- ... ...

    Abstract The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self-amplifying mRNA (SAM) vaccine encoding a prefusion stabilized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein and demonstrate strong cellular and humoral immune responses at low doses in mice and rhesus macaques. The homologous prime-boost vaccination regimen of SAM at 3, 10 and 30 μg induced potent neutralizing antibody (nAb) titers in rhesus macaques following two SAM vaccinations at all dose levels, with the 10 μg dose generating geometric mean titers (GMT) 48-fold greater than the GMT of a panel of SARS-CoV-2 convalescent human sera. Spike-specific T cell responses were observed with all tested vaccine regimens. SAM vaccination provided protective efficacy against SARS-CoV-2 challenge as both a homologous prime-boost and as a single boost following ChAd prime, demonstrating reduction of viral replication in both the upper and lower airways. The SAM vaccine is currently being evaluated in clinical trials as both a homologous prime-boost regimen at low doses and as a boost following heterologous prime.
    MeSH term(s) Animals ; Antibodies, Neutralizing ; Antibodies, Viral ; COVID-19/prevention & control ; COVID-19 Vaccines ; Humans ; Macaca mulatta/genetics ; Mice ; RNA, Messenger ; SARS-CoV-2/genetics ; Spike Glycoprotein, Coronavirus/genetics ; Vaccination
    Chemical Substances Antibodies, Neutralizing ; Antibodies, Viral ; COVID-19 Vaccines ; RNA, Messenger ; Spike Glycoprotein, Coronavirus ; spike protein, SARS-CoV-2
    Language English
    Publishing date 2022-06-07
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-022-31005-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: phase 1 trial interim results.

    Rappaport, Amy R / Kyi, Chrisann / Lane, Monica / Hart, Meghan G / Johnson, Melissa L / Henick, Brian S / Liao, Chih-Yi / Mahipal, Amit / Shergill, Ardaman / Spira, Alexander I / Goldman, Jonathan W / Scallan, Ciaran D / Schenk, Desiree / Palmer, Christine D / Davis, Matthew J / Kounlavouth, Sonia / Kemp, Lindsey / Yang, Aaron / Li, Yaojun John /
    Likes, Molly / Shen, Annie / Boucher, Gregory R / Egorova, Milana / Veres, Robert L / Espinosa, J Aaron / Jaroslavsky, Jason R / Kraemer Tardif, Lauren D / Acrebuche, Lindsey / Puccia, Christopher / Sousa, Leiliane / Zhou, Rita / Bae, Kyounghwa / Hecht, J Randolph / Carbone, David P / Johnson, Benny / Allen, Andrew / Ferguson, Andrew R / Jooss, Karin

    Nature medicine

    2024  Volume 30, Issue 4, Page(s) 1013–1022

    Abstract: Therapeutic vaccines that elicit cytotoxic T cell responses targeting tumor-specific neoantigens hold promise for providing long-term clinical benefit to patients with cancer. Here we evaluated safety and tolerability of a therapeutic vaccine encoding 20 ...

    Abstract Therapeutic vaccines that elicit cytotoxic T cell responses targeting tumor-specific neoantigens hold promise for providing long-term clinical benefit to patients with cancer. Here we evaluated safety and tolerability of a therapeutic vaccine encoding 20 shared neoantigens derived from selected common oncogenic driver mutations as primary endpoints in an ongoing phase 1/2 study in patients with advanced/metastatic solid tumors. Secondary endpoints included immunogenicity, overall response rate, progression-free survival and overall survival. Eligible patients were selected if their tumors expressed one of the human leukocyte antigen-matched tumor mutations included in the vaccine, with the majority of patients (18/19) harboring a mutation in KRAS. The vaccine regimen, consisting of a chimp adenovirus (ChAd68) and self-amplifying mRNA (samRNA) in combination with the immune checkpoint inhibitors ipilimumab and nivolumab, was shown to be well tolerated, with observed treatment-related adverse events consistent with acute inflammation expected with viral vector-based vaccines and immune checkpoint blockade, the majority grade 1/2. Two patients experienced grade 3/4 serious treatment-related adverse events that were also dose-limiting toxicities. The overall response rate was 0%, and median progression-free survival and overall survival were 1.9 months and 7.9 months, respectively. T cell responses were biased toward human leukocyte antigen-matched TP53 neoantigens encoded in the vaccine relative to KRAS neoantigens expressed by the patients' tumors, indicating a previously unknown hierarchy of neoantigen immunodominance that may impact the therapeutic efficacy of multiepitope shared neoantigen vaccines. These data led to the development of an optimized vaccine exclusively targeting KRAS-derived neoantigens that is being evaluated in a subset of patients in phase 2 of the clinical study. ClinicalTrials.gov registration: NCT03953235 .
    MeSH term(s) Humans ; Antigens, Neoplasm ; Cancer Vaccines/adverse effects ; HLA Antigens ; Immune Checkpoint Inhibitors/therapeutic use ; Neoplasms/drug therapy ; Neoplasms/pathology ; Proto-Oncogene Proteins p21(ras)/genetics ; Vaccines/therapeutic use
    Chemical Substances Antigens, Neoplasm ; Cancer Vaccines ; HLA Antigens ; Immune Checkpoint Inhibitors ; Proto-Oncogene Proteins p21(ras) (EC 3.6.5.2) ; Vaccines
    Language English
    Publishing date 2024-03-27
    Publishing country United States
    Document type Clinical Trial, Phase I ; Journal Article
    ZDB-ID 1220066-9
    ISSN 1546-170X ; 1078-8956
    ISSN (online) 1546-170X
    ISSN 1078-8956
    DOI 10.1038/s41591-024-02851-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: A self-amplifying mRNA COVID-19 vaccine drives potent and broad immune responses at low doses that protects non-human primates against SARS-CoV-2

    Rappaport, Amy R / Hong, Sue-Jean / Scallan, Ciaran D / Gitlin, Leonid / Akoopie, Arvin / Boucher, Gregory R / Egorova, Milana / Espinosa, J Aaron / Fidanza, Mario / Kachura, Melissa A / Shen, Annie / Sivko, Gloria / Van Abbema, Anne / Veres, Robert L / Jooss, Karin

    bioRxiv

    Abstract: The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self- ... ...

    Abstract The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self-amplifying mRNA (SAM) vaccine encoding a prefusion stabilized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein and demonstrate potent cellular and humoral immune responses at low doses in mice and rhesus macaques. The homologous prime-boost vaccination regimen of SAM at 3, 10 and 30 μg induced potent neutralizing antibody titers in rhesus macaques following two SAM vaccinations at all dose levels, with the 10 μg dose generating geometric mean titers (GMT) 48-fold greater than the GMT of a panel of SARS-CoV-2 convalescent human sera. Spike-specific T cell responses were observed at all dose levels. SAM vaccination provided protective efficacy against SARS-CoV-2 challenge as both a homologous prime-boost and as a single boost following ChAd prime, demonstrating reduction of viral replication in both the upper and lower airways. Protection was most effective with a SAM prime-boost vaccination regimen at 10 and 30 μg and with a ChAd/SAM heterologous prime-boost regimen. The SAM vaccine is currently being evaluated in clinical trials as both a homologous prime-boost regimen at low doses and as a boost following heterologous prime.
    Keywords covid19
    Language English
    Publishing date 2021-11-09
    Publisher Cold Spring Harbor Laboratory
    Document type Article ; Online
    DOI 10.1101/2021.11.08.467773
    Database COVID19

    Kategorien

To top