Your browser doesn't support javascript.
loading
In vivo immuno-targeting of an extracellular epitope of membrane bound preferentially expressed antigen in melanoma (PRAME).
Pankov, Dmitry; Sjöström, Ludvig; Kalidindi, Teja; Lee, Sang-Gyu; Sjöström, Kjell; Gardner, Rui; McDevitt, Michael R; O'Reilly, Richard; Thorek, Daniel L J; Larson, Steven M; Veach, Darren; Ulmert, David.
Afiliação
  • Pankov D; Immunology Program, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Sjöström L; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Kalidindi T; Division of Oncology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden.
  • Lee SG; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Sjöström K; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Gardner R; Innovagen AB, Lund, Sweden.
  • McDevitt MR; Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • O'Reilly R; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Thorek DLJ; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
  • Larson SM; Immunology Program, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Veach D; Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Ulmert D; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
Oncotarget ; 8(39): 65917-65931, 2017 Sep 12.
Article em En | MEDLINE | ID: mdl-29029482
ABSTRACT
Preferentially Expressed Antigen in Melanoma (PRAME) is a cancer/testis antigen that is overexpressed in a broad range of malignancies, while absent in most healthy human tissues, making it an attractive diagnostic cancer biomarker and therapeutic target. Although commonly viewed as an intracellular protein, we have demonstrated that PRAME has a membrane bound form with an external epitope targetable with conventional antibodies. We generated a polyclonal antibody (Membrane associated PRAME Antibody 1, MPA1) against an extracellular peptide sequence of PRAME. Binding of MPA1 to recombinant PRAME was evaluated by Enzyme-Linked Immunosorbent Assay (ELISA). Flow cytometry and confocal immunofluorescence microscopy of MPA1 was performed on multiple tumor cell lines. Reverse Transcription Polymerase Chain Reaction (RT-PCR) for PRAME was conducted to compare protein and transcriptional expression levels. We demonstrated a robust proof-of-concept for PRAME targeting in vivo by radiolabeling MPA1 with zirconium-89 (89Zr-DFO-MPA1) and demonstrating high specific uptake in PRAME expressing tumors. To our knowledge, this is the first time a cancer testis antigen has been targeted using conventional antibody technologies. Thus, PRAME can be exploited for multiple clinical applications, including targeted therapy, diagnostic imaging and treatment guidance in a wide-range of malignancies, with minimal off-target toxicity.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Guideline Idioma: En Revista: Oncotarget Ano de publicação: 2017 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Guideline Idioma: En Revista: Oncotarget Ano de publicação: 2017 Tipo de documento: Article País de afiliação: Estados Unidos