Unknown

Dataset Information

0

Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma.


ABSTRACT: Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13R?2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13R?2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13R?2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13R?2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13R?2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.

SUBMITTER: Hegde M 

PROVIDER: S-EPMC3831041 | biostudies-literature | 2013 Nov

REPOSITORIES: biostudies-literature

altmetric image

Publications

Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma.

Hegde Meenakshi M   Corder Amanda A   Chow Kevin K H KK   Mukherjee Malini M   Ashoori Aidin A   Kew Yvonne Y   Zhang Yi Jonathan YJ   Baskin David S DS   Merchant Fatima A FA   Brawley Vita S VS   Byrd Tiara T TT   Krebs Simone S   Wu Meng Fen MF   Liu Hao H   Heslop Helen E HE   Gottschalk Stephen S   Yvon Eric E   Ahmed Nabil N  

Molecular therapy : the journal of the American Society of Gene Therapy 20130813 11


Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER  ...[more]

Similar Datasets

| S-EPMC6467811 | biostudies-literature
| S-EPMC5805388 | biostudies-literature
| S-EPMC7904624 | biostudies-literature
| S-EPMC6541619 | biostudies-literature
| S-EPMC6167529 | biostudies-literature
| S-EPMC5626814 | biostudies-literature
| S-EPMC5754236 | biostudies-literature
| S-EPMC3247626 | biostudies-literature
| S-EPMC4489991 | biostudies-literature
| S-EPMC2762661 | biostudies-literature