Preclinical optimization of a GPC2-targeting CAR T-cell therapy for neuroblastoma

Although most patients with newly diagnosed high-risk neuroblastoma (NB) achieve remission after initial therapy, more than 50% experience late relapses caused by minimal residual disease (MRD) and succumb to their cancer. Therapeutic strategies to target MRD may benefit these children. We developed...

Full description

Saved in:
Bibliographic Details
Published inJournal for immunotherapy of cancer Vol. 11; no. 1; p. e005881
Main Authors Sun, Ming, Cao, Yingying, Okada, Reona, Reyes-González, Jeyshka M, Stack, Hannah G, Qin, Haiying, Li, Nan, Seibert, Charlie, Kelly, Michael C, Ruppin, Eytan, Ho, Mitchell, Thiele, Carol J, Nguyen, Rosa
Format Journal Article
LanguageEnglish
Published England BMJ Publishing Group LTD 01.01.2023
BMJ Publishing Group
SeriesOriginal research
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Although most patients with newly diagnosed high-risk neuroblastoma (NB) achieve remission after initial therapy, more than 50% experience late relapses caused by minimal residual disease (MRD) and succumb to their cancer. Therapeutic strategies to target MRD may benefit these children. We developed a new chimeric antigen receptor (CAR) targeting glypican-2 (GPC2) and conducted iterative preclinical engineering of the CAR structure to maximize its anti-tumor efficacy before clinical translation. We evaluated different GPC2-CAR constructs by measuring the CAR activity in vitro. NOD-SCID mice engrafted orthotopically with human NB cell lines or patient-derived xenografts and treated with human CAR T cells served as in vivo models. Mechanistic studies were performed using single-cell RNA-sequencing. Applying stringent in vitro assays and orthotopic in vivo NB models, we demonstrated that our single-chain variable fragment, CT3, integrated into a CAR vector with a CD28 hinge, CD28 transmembrane, and 4-1BB co-stimulatory domain (CT3.28H.BBζ) elicits the best preclinical anti-NB activity compared with other tested CAR constructs. This enhanced activity was associated with an enrichment of CD8 effector T cells in the tumor-microenvironment and upregulation of several effector molecules such as , , , and . Finally, we also showed that the CT3.28H.BBζ CAR we developed was more potent than a recently clinically tested GD2-targeted CAR to control NB growth in vivo. Given the robust preclinical activity of CT3.28H.BBζ, these results form a promising basis for further clinical testing in children with NB.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2051-1426
2051-1426
DOI:10.1136/jitc-2022-005881