1. Academic Validation
  2. The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions

The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions

  • Clin Cancer Res. 2021 Jun 1;27(11):3154-3166. doi: 10.1158/1078-0432.CCR-20-3605.
Igor Odintsov 1 2 Allan J W Lui 1 Whitney J Sisso 1 Eric Gladstone 1 2 Zebing Liu 3 Lukas Delasos 1 Renate I Kurth 1 Exequiel M Sisso 4 Morana Vojnic 1 2 Inna Khodos 5 Marissa S Mattar 5 Elisa de Stanchina 5 Shawn M Leland 6 Marc Ladanyi 7 2 Romel Somwar 7 2
Affiliations

Affiliations

  • 1 Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
  • 2 Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • 3 Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
  • 4 Development Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • 5 Anti-tumor Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • 6 Elevation Oncology, Inc. New York, New York.
  • 7 Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. ladanyim@mskcc.org somwarr@mskcc.org.
Abstract

Purpose: Oncogenic fusions involving the neuregulin 1 (NRG1) gene are found in approximately 0.2% of cancers of diverse histologies. The resulting chimeric NRG1 proteins bind predominantly to HER3, leading to HER3-HER2 dimerization and activation of downstream growth and survival pathways. HER3 is, therefore, a rational target for therapy in NRG1 fusion-driven cancers.

Experimental design: We developed novel patient-derived and isogenic models of NRG1-rearranged cancers and examined the effect of the anti-HER3 antibody, seribantumab, on growth and activation of signaling networks in vitro and in vivo.

Results: Seribantumab inhibited NRG1-stimulated growth of MCF-7 cells and growth of patient-derived breast (MDA-MB-175-VII, DOC4-NRG1 fusion) and lung (LUAD-0061AS3, SLC3A2-NRG1 fusion) Cancer cells harboring NRG1 fusions or NRG1 amplification (HCC-95). In addition, seribantumab inhibited growth of isogenic HBEC cells expressing a CD74-NRG1 fusion (HBECp53-CD74-NRG1) and induced Apoptosis in MDA-MB-175-VII and LUAD-0061AS3 cells. Induction of proapoptotic proteins and reduced expression of the cell-cycle regulator, cyclin D1, were observed in seribantumab-treated cells. Treatment of MDA-MB-175-VII, LUAD-0061AS3, and HBECp53-CD74-NRG1 cells with seribantumab reduced phosphorylation of EGFR, HER2, HER3, HER4, and known downstream signaling molecules, such as Akt and ERK1/2. Significantly, administration of seribantumab to mice bearing LUAD-0061AS3 patient-derived xenograft (PDX) and OV-10-0050 (ovarian Cancer with CLU-NRG1 fusion) PDX tumors induced regression of tumors by 50%-100%. Afatinib was much less effective at blocking tumor growth.

Conclusions: Seribantumab treatment blocked activation of the four ERBB family members and of downstream signaling, leading to inhibition of NRG1 fusion-dependent tumorigenesis in vitro and in vivo in breast, lung, and ovarian patient-derived Cancer models.

Figures
Products