Anti-GRP-R monoclonal antibody antitumor therapy against neuroblastoma

PLoS One. 2022 Dec 16;17(12):e0277956. doi: 10.1371/journal.pone.0277956. eCollection 2022.

Abstract

Standard treatment for patients with high-risk neuroblastoma remains multimodal therapy including chemoradiation, surgical resection, and autologous stem cell rescue. Immunotherapy has demonstrated success in treating many types of cancers; however, its use in pediatric solid tumors has been limited by low tumor mutation burdens. Gastrin-releasing peptide receptor (GRP-R) is overexpressed in numerous malignancies, including poorly-differentiated neuroblastoma. Monoclonal antibodies (mAbs) to GRP-R have yet to be developed but could serve as a potential novel immunotherapy. This preclinical study aims to evaluate the efficacy of a novel GRP-R mAb immunotherapy against neuroblastoma. We established four candidate anti-GRP-R mAbs by screening a single-chain variable fragment (scFv) library. GRP-R mAb-1 demonstrated the highest efficacy with the lowest EC50 at 4.607 ng/ml against GRP-R expressing neuroblastoma cells, blocked the GRP-ligand activation of GRP-R and its downstream PI3K/AKT signaling. This resulted in functional inhibition of cell proliferation and anchorage-independent growth, indicating that mAb-1 has an antagonist inhibitory role on GRP-R. To examine the antibody-dependent cellular cytotoxicity (ADCC) of GRP-R mAb-1 on neuroblastoma, we co-cultured neuroblastoma cells with natural killer (NK) cells versus GRP-R mAb-1 treatment alone. GRP-R mAb-1 mediated ADCC effects on neuroblastoma cells and induced release of IFNγ by NK cells under co-culture conditions in vitro. The cytotoxic effects of mAb-1 were confirmed with the secretion of cytotoxic granzyme B from NK cells and the reduction of mitotic tumor cells in vivo using a murine tumor xenograft model. In summary, GRP-R mAb-1 demonstrated efficacious anti-tumor effects on neuroblastoma cells in preclinical models. Importantly, GRP-R mAb-1 may be an efficacious, novel immunotherapy in the treatment of high-risk neuroblastoma patients.

Publication types

  • Research Support, Non-U.S. Gov't
  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Antibodies, Monoclonal / pharmacology
  • Antibodies, Monoclonal / therapeutic use
  • Cell Line, Tumor
  • Child
  • Humans
  • Mice
  • Neuroblastoma* / metabolism
  • Phosphatidylinositol 3-Kinases / metabolism
  • Receptors, Bombesin* / metabolism

Substances

  • Receptors, Bombesin
  • Phosphatidylinositol 3-Kinases
  • Antibodies, Monoclonal

Grants and funding

This work was supported by a grant from the National Institutes of Health (R01 DK61470). This work was supported in part by the Welch Foundation grant AU-0042-20030616 (ZA), and Cancer Prevention and Research Institute of Texas (CPRIT) Grants RP150230, RP150551, and RP190561 (ZA). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.