Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model

Standard

Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model. / Benten, Daniel; Behrang, Yasmin; Unrau, Ludmilla; Weissmann, Victoria; Wolters-Eisfeld, Gerrit; Burdak-Rothkamm, Susanne; Stahl, Felix R; Anlauf, Martin; Grabowski, Patricia; Möbs, Markus; Dieckhoff, Jan; Sipos, Bence; Fahl, Martina; Eggers, Corinna; Perez, Daniel; Bockhorn, Maximilian; Izbicki, Jakob R; Lohse, Ansgar W; Schrader, Jörg.

in: MOL CANCER RES, Jahrgang 16, Nr. 3, 01.03.2018, S. 496-507.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Benten, D, Behrang, Y, Unrau, L, Weissmann, V, Wolters-Eisfeld, G, Burdak-Rothkamm, S, Stahl, FR, Anlauf, M, Grabowski, P, Möbs, M, Dieckhoff, J, Sipos, B, Fahl, M, Eggers, C, Perez, D, Bockhorn, M, Izbicki, JR, Lohse, AW & Schrader, J 2018, 'Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model', MOL CANCER RES, Jg. 16, Nr. 3, S. 496-507. https://doi.org/10.1158/1541-7786.MCR-17-0163

APA

Benten, D., Behrang, Y., Unrau, L., Weissmann, V., Wolters-Eisfeld, G., Burdak-Rothkamm, S., Stahl, F. R., Anlauf, M., Grabowski, P., Möbs, M., Dieckhoff, J., Sipos, B., Fahl, M., Eggers, C., Perez, D., Bockhorn, M., Izbicki, J. R., Lohse, A. W., & Schrader, J. (2018). Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model. MOL CANCER RES, 16(3), 496-507. https://doi.org/10.1158/1541-7786.MCR-17-0163

Vancouver

Bibtex

@article{d11e53354dbb475fb31b63ec50b6ca7c,
title = "Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model",
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. {\textcopyright}2018 AACR.",
keywords = "Journal Article",
author = "Daniel Benten and Yasmin Behrang and Ludmilla Unrau and Victoria Weissmann and Gerrit Wolters-Eisfeld and Susanne Burdak-Rothkamm and Stahl, {Felix R} and Martin Anlauf and Patricia Grabowski and Markus M{\"o}bs and Jan Dieckhoff and Bence Sipos and Martina Fahl and Corinna Eggers and Daniel Perez and Maximilian Bockhorn and Izbicki, {Jakob R} and Lohse, {Ansgar W} and J{\"o}rg Schrader",
note = "{\textcopyright}2018 American Association for Cancer Research.",
year = "2018",
month = mar,
day = "1",
doi = "10.1158/1541-7786.MCR-17-0163",
language = "English",
volume = "16",
pages = "496--507",
journal = "MOL CANCER RES",
issn = "1541-7786",
publisher = "American Association for Cancer Research Inc.",
number = "3",

}

RIS

TY - JOUR

T1 - Establishment of the First Well-differentiated Human Pancreatic Neuroendocrine Tumor Model

AU - Benten, Daniel

AU - Behrang, Yasmin

AU - Unrau, Ludmilla

AU - Weissmann, Victoria

AU - Wolters-Eisfeld, Gerrit

AU - Burdak-Rothkamm, Susanne

AU - Stahl, Felix R

AU - Anlauf, Martin

AU - Grabowski, Patricia

AU - Möbs, Markus

AU - Dieckhoff, Jan

AU - Sipos, Bence

AU - Fahl, Martina

AU - Eggers, Corinna

AU - Perez, Daniel

AU - Bockhorn, Maximilian

AU - Izbicki, Jakob R

AU - Lohse, Ansgar W

AU - Schrader, Jörg

N1 - ©2018 American Association for Cancer Research.

PY - 2018/3/1

Y1 - 2018/3/1

N2 - 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.

AB - 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.

KW - Journal Article

U2 - 10.1158/1541-7786.MCR-17-0163

DO - 10.1158/1541-7786.MCR-17-0163

M3 - SCORING: Journal article

C2 - 29330294

VL - 16

SP - 496

EP - 507

JO - MOL CANCER RES

JF - MOL CANCER RES

SN - 1541-7786

IS - 3

ER -