Your browser doesn't support javascript.
loading
A Rationally Designed Fully Human EGFRvIII:CD3-Targeted Bispecific Antibody Redirects Human T Cells to Treat Patient-derived Intracerebral Malignant Glioma.
Gedeon, Patrick C; Schaller, Teilo H; Chitneni, Satish K; Choi, Bryan D; Kuan, Chien-Tsun; Suryadevara, Carter M; Snyder, David J; Schmittling, Robert J; Szafranski, Scott E; Cui, Xiuyu; Healy, Patrick N; Herndon, James E; McLendon, Roger E; Keir, Stephen T; Archer, Gary E; Reap, Elizabeth A; Sanchez-Perez, Luis; Bigner, Darell D; Sampson, John H.
Afiliação
  • Gedeon PC; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Schaller TH; Department of Biomedical Engineering, Duke University, Durham, North Carolina.
  • Chitneni SK; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Choi BD; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Kuan CT; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Suryadevara CM; Department of Pathology, Duke University Medical Center, Durham, North Carolina.
  • Snyder DJ; Department of Radiology, Duke University Medical Center, Durham, North Carolina.
  • Schmittling RJ; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Szafranski SE; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Cui X; Department of Pathology, Duke University Medical Center, Durham, North Carolina.
  • Healy PN; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Herndon JE; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • McLendon RE; Department of Pathology, Duke University Medical Center, Durham, North Carolina.
  • Keir ST; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Archer GE; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Reap EA; Department of Pathology, Duke University Medical Center, Durham, North Carolina.
  • Sanchez-Perez L; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Bigner DD; The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
  • Sampson JH; Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
Clin Cancer Res ; 24(15): 3611-3631, 2018 08 01.
Article em En | MEDLINE | ID: mdl-29703821
ABSTRACT

Purpose:

Conventional therapy for malignant glioma fails to specifically target tumor cells. In contrast, substantial evidence indicates that if appropriately redirected, T cells can precisely eradicate tumors. Here we report the rational development of a fully human bispecific antibody (hEGFRvIII-CD3 bi-scFv) that redirects human T cells to lyse malignant glioma expressing a tumor-specific mutation of the EGFR (EGFRvIII).Experimental

Design:

We generated a panel of bispecific single-chain variable fragments and optimized design through successive rounds of screening and refinement. We tested the ability of our lead construct to redirect naïve T cells and induce target cell-specific lysis. To test for efficacy, we evaluated tumor growth and survival in xenogeneic and syngeneic models of glioma. Tumor penetrance following intravenous drug administration was assessed in highly invasive, orthotopic glioma models.

Results:

A highly expressed bispecific antibody with specificity to CD3 and EGFRvIII was generated (hEGFRvIII-CD3 bi-scFv). Antibody-induced T-cell activation, secretion of proinflammatory cytokines, and proliferation was robust and occurred exclusively in the presence of target antigen. hEGFRvIII-CD3 bi-scFv was potent and target-specific, mediating significant lysis of multiple malignant glioma cell lines and patient-derived malignant glioma samples that heterogeneously express EGFRvIII. In both subcutaneous and orthotopic models, well-engrafted, patient-derived malignant glioma was effectively treated despite heterogeneity of EGFRvIII expression; intravenous hEGFRvIII-CD3 bi-scFv administration caused significant regression of tumor burden (P < 0.0001) and significantly extended survival (P < 0.0001). Similar efficacy was obtained in highly infiltrative, syngeneic glioma models, and intravenously administered hEGFRvIII-CD3 bi-scFv localized to these orthotopic tumors.

Conclusions:

We have developed a clinically translatable bispecific antibody that redirects human T cells to safely and effectively treat malignant glioma. On the basis of these results, we have developed a clinical study of hEGFRvIII-CD3 bi-scFv for patients with EGFRvIII-positive malignant glioma. Clin Cancer Res; 24(15); 3611-31. ©2018 AACR.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Complexo CD3 / Receptores ErbB / Glioma / Imunoterapia Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Complexo CD3 / Receptores ErbB / Glioma / Imunoterapia Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2018 Tipo de documento: Article