Your browser doesn't support javascript.
Design and proof of concept for targeted phage-based COVID-19 vaccination strategies with a streamlined cold-free supply chain.
Staquicini, Daniela I; Tang, Fenny H F; Markosian, Christopher; Yao, Virginia J; Staquicini, Fernanda I; Dodero-Rojas, Esteban; Contessoto, Vinícius G; Davis, Deodate; O'Brien, Paul; Habib, Nazia; Smith, Tracey L; Bruiners, Natalie; Sidman, Richard L; Gennaro, Maria L; Lattime, Edmund C; Libutti, Steven K; Whitford, Paul C; Burley, Stephen K; Onuchic, José N; Arap, Wadih; Pasqualini, Renata.
  • Staquicini DI; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Tang FHF; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Markosian C; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Yao VJ; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Staquicini FI; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Dodero-Rojas E; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Contessoto VG; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Davis D; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • O'Brien P; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Habib N; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Smith TL; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005.
  • Bruiners N; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005.
  • Sidman RL; Department of Physics, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054, Brazil.
  • Gennaro ML; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Lattime EC; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Libutti SK; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Whitford PC; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Burley SK; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Onuchic JN; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
  • Arap W; Rutgers Cancer Institute of New Jersey, Newark, NJ 07101.
  • Pasqualini R; Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: covidwho-1301236
ABSTRACT
Development of effective vaccines against coronavirus disease 2019 (COVID-19) is a global imperative. Rapid immunization of the entire human population against a widespread, continually evolving, and highly pathogenic virus is an unprecedented challenge, and different vaccine approaches are being pursued. Engineered filamentous bacteriophage (phage) particles have unique potential in vaccine development due to their inherent immunogenicity, genetic plasticity, stability, cost-effectiveness for large-scale production, and proven safety profile in humans. Herein we report the development and initial evaluation of two targeted phage-based vaccination approaches against SARS-CoV-2 dual ligand peptide-targeted phage and adeno-associated virus/phage (AAVP) particles. For peptide-targeted phage, we performed structure-guided antigen design to select six solvent-exposed epitopes of the SARS-CoV-2 spike (S) protein. One of these epitopes displayed on the major capsid protein pVIII of phage induced a specific and sustained humoral response when injected in mice. These phage were further engineered to simultaneously display the peptide CAKSMGDIVC on the minor capsid protein pIII to enable their transport from the lung epithelium into the systemic circulation. Aerosolization of these "dual-display" phage into the lungs of mice generated a systemic and specific antibody response. In the second approach, targeted AAVP particles were engineered to deliver the entire S protein gene under the control of a constitutive CMV promoter. This induced tissue-specific transgene expression, stimulating a systemic S protein-specific antibody response in mice. With these proof-of-concept preclinical experiments, we show that both targeted phage- and AAVP-based particles serve as robust yet versatile platforms that can promptly yield COVID-19 vaccine prototypes for translational development.
Subject(s)
Keywords

Full text: Available Collection: International databases Database: MEDLINE Main subject: Bacteriophages / Immunization Programs / COVID-19 Vaccines / COVID-19 Type of study: Experimental Studies / Prognostic study Topics: Vaccines Limits: Animals Language: English Year: 2021 Document Type: Article

Similar

MEDLINE

...
LILACS

LIS


Full text: Available Collection: International databases Database: MEDLINE Main subject: Bacteriophages / Immunization Programs / COVID-19 Vaccines / COVID-19 Type of study: Experimental Studies / Prognostic study Topics: Vaccines Limits: Animals Language: English Year: 2021 Document Type: Article