Establishment of the first well-differentiated human pancreatic neuroendocrine tumor model

D Benten, Y Behrang, L Unrau, V Weissmann… - Molecular Cancer …, 2018 - AACR
D Benten, Y Behrang, L Unrau, V Weissmann, G Wolters-Eisfeld, S Burdak-Rothkamm
Molecular Cancer Research, 2018AACR
Clinical options for systemic therapy of neuroendocrine tumors (NET) are limited.
Development of new drugs requires suitable representative in vitro and in vivo model
systems. So far, the unavailability of a human model with a well-differentiated phenotype
and typical growth characteristics has impaired preclinical research in NET. Herein, we
establish and characterize a lymph node–derived cell line (NT-3) from a male patient with
well-differentiated pancreatic NET. Neuroendocrine differentiation and tumor biology was …
Abstract
Clinical options for systemic therapy of neuroendocrine tumors (NET) are limited. Development of new drugs requires suitable representative in vitro and in vivo model systems. So far, the unavailability of a human model with a well-differentiated phenotype and typical growth characteristics has impaired preclinical research in NET. Herein, we establish and characterize a lymph node–derived cell line (NT-3) from a male patient with well-differentiated pancreatic NET. Neuroendocrine differentiation and tumor biology was compared with existing NET cell lines BON and QGP-1. In vivo growth was assessed in a xenograft mouse model. The neuroendocrine identity of NT-3 was verified by expression of multiple NET-specific markers, which were highly expressed in NT-3 compared with BON and QGP-1. In addition, NT-3 expressed and secreted insulin. Until now, this well-differentiated phenotype is stable since 58 passages. The proliferative labeling index, measured by Ki-67, of 14.6% ± 1.0% in NT-3 is akin to the original tumor (15%–20%), and was lower than in BON (80.6% ± 3.3%) and QGP-1 (82.6% ± 1.0%). NT-3 highly expressed somatostatin receptors (SSTRs: 1, 2, 3, and 5). Upon subcutaneous transplantation of NT-3 cells, recipient mice developed tumors with an efficient tumor take rate (94%) and growth rate (139% ± 13%) by 4 weeks. Importantly, morphology and neuroendocrine marker expression of xenograft tumors resembled the original human tumor.
Implications: High expression of somatostatin receptors and a well-differentiated phenotype as well as a slow growth rate qualify the new cell line as a relevant model to study neuroendocrine tumor biology and to develop new tumor treatments. Mol Cancer Res; 16(3); 496–507. ©2018 AACR.
AACR