Antibody-mediated targeting of iron oxide nanoparticles to the folate receptor alpha increases tumor cell association in vitro and in vivo

Christian Ndong,1 Seiko Toraya-Brown,2 Katsiaryna Kekalo,1 Ian Baker,1 Tillman U Gerngross,1,3,4 Steven N Fiering,2,5,6 Karl E Griswold1,3,6 1Thayer School of Engineering, Dartmouth, Hanover, NH, USA; 2Department of Microbiology and Immunology, Geisel School of Medicine at Dartmo...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Ndong C, Toraya-Brown S, Kekalo K, Baker I, Gerngross TU, Fiering SN, Griswold KE
Formato: article
Lenguaje:EN
Publicado: Dove Medical Press 2015
Materias:
Acceso en línea:https://doaj.org/article/2375a28548b0405b8e50bd05f06d4e82
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
Descripción
Sumario:Christian Ndong,1 Seiko Toraya-Brown,2 Katsiaryna Kekalo,1 Ian Baker,1 Tillman U Gerngross,1,3,4 Steven N Fiering,2,5,6 Karl E Griswold1,3,6 1Thayer School of Engineering, Dartmouth, Hanover, NH, USA; 2Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; 3Department of Biological Sciences, Dartmouth, Hanover, NH, USA; 4Department of Chemistry, Dartmouth, Hanover, NH, USA; 5Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; 6Norris Cotton Cancer Center, Lebanon, NH, USA Abstract: Active molecular targeting has become an important aspect of nanoparticle development for oncology indications. Here, we describe molecular targeting of iron oxide nanoparticles (IONPs) to the folate receptor alpha (FOLRα) using an engineered antibody fragment (Ffab). Compared to control nanoparticles targeting the non-relevant botulinum toxin, the Ffab-IONP constructs selectively accumulated on FOLRα-overexpressing cancer cells in vitro, where they exhibited the capacity to internalize into intracellular vesicles. Similarly, Ffab-IONPs homed to FOLRα-positive tumors upon intraperitoneal administration in an orthotopic murine xenograft model of ovarian cancer, whereas negative control particles showed no detectable tumor accumulation. Interestingly, Ffab-IONPs built with custom 120 nm nanoparticles exhibited lower in vitro targeting efficiency when compared to those built with commercially sourced 180 nm nanoparticles. In vivo, however, the two Ffab-IONP platforms achieved equivalent tumor homing, although the smaller 120 nm IONPs were more prone to liver sequestration. Overall, the results show that Ffab-mediated targeting of IONPs yields specific, high-level accumulation within cancer cells, and this fact suggests that Ffab-IONPs could have future utility in ovarian cancer diagnostics and therapy. Keywords: nanoparticle targeting, antibody fragment, biodistribution, ovarian cancer