Your browser doesn't support javascript.
loading
A Novel CRISPR-Engineered, Stem Cell-Derived Cellular Vaccine
Krishnendu Chakraborty; Abishek Chandrashekar; Adam Sidaway; Elizabeth Latta; Jingyou Yu; Katherine McMahan; Cordelia Manickam; Kyle Kroll; Matthew Mosher; R. Keith Reeves; Rihab Gam; Elisa Arthofer; Modassir Choudhry; Dan H. Barouch; Tom Henley.
Afiliação
  • Krishnendu Chakraborty; Intima Bioscience
  • Abishek Chandrashekar; Beth Israel Deaconess Medical Center
  • Adam Sidaway; Intima Bioscience
  • Elizabeth Latta; Intima Bioscience
  • Jingyou Yu; Beth Israel Deaconess Medical Center
  • Katherine McMahan; Beth Israel Deaconess Medical Center
  • Cordelia Manickam; BIDMC/Harvard Medical School
  • Kyle Kroll; BIDMC/Harvard Medical School
  • Matthew Mosher; BIDMC/Harvard Medical School
  • R. Keith Reeves; BIDMC/Harvard Medical School
  • Rihab Gam; Intima Bioscience
  • Elisa Arthofer; Intima Bioscience
  • Modassir Choudhry; Intima Bioscience
  • Dan H. Barouch; Beth Israel Deaconess Medical Center
  • Tom Henley; Intima Bioscience
Preprint em En | PREPRINT-BIORXIV | ID: ppbiorxiv-474336
ABSTRACT
COVID-19 has forced rapid clinical translation of novel vaccine technologies, principally mRNA vaccines, that have resulted in meaningful efficacy and adequate safety in response to the global pandemic. Notwithstanding this success, there remains an opportunity for innovation in vaccine technology to address current limitations and meet the challenges of inevitable future pandemics. We describe a universal vaccine cell (UVC) rationally designed to mimic the natural physiologic immunity induced post viral infection of host cells. Induced pluripotent stem cells were CRISPR engineered to delete MHC-I expression and simultaneously overexpress a NK Ligand adjuvant to increase rapid cellular apoptosis which was hypothesized to enhance viral antigen presentation in the resulting immune microenvironment leading to a protective immune response. Cells were further engineered to express the parental variant WA1/2020 SARS-CoV-2 spike protein as a representative viral antigen prior to irradiation and cryopreservation. The cellular vaccine was then used to immunize non-human primates in a standard 2-dose, IM injected prime + boost vaccination with 1e8 cells per 1 ml dose resulting in robust neutralizing antibody responses (1e3 nAb titers) with decreasing levels at 6 months duration. Similar titers generated in this established NHP model have translated into protective human neutralizing antibody levels in SARS-Cov-2 vaccinated individuals. Animals vaccinated with WA1/2020 spike antigens were subsequently challenged with 1.0 x 105 TCID50 infectious Delta (B.1.617.2) SARS-CoV-2 in a heterologous challenge which resulted in an approximately 3-log order decrease in viral RNA load in the lungs. These heterologous viral challenge results reflect the ongoing real-world experience of original variant WA1/2020 spike antigen vaccinated populations exposed to rapidly emerging variants like Delta and now Omicron. This cellular vaccine is designed to be a rapidly scalable cell line with a modular poly-antigenic payload to allow for practical, large-scale clinical manufacturing and use in an evolving viral variant environment. Human clinical translation of the UVC is being actively explored for this and potential future pandemics.
Licença
cc_by_nc_nd
Texto completo: 1 Coleções: 09-preprints Base de dados: PREPRINT-BIORXIV Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2021 Tipo de documento: Preprint
Texto completo: 1 Coleções: 09-preprints Base de dados: PREPRINT-BIORXIV Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2021 Tipo de documento: Preprint
...