Skip to main content
Top
Published in: Endocrine 2/2017

01-08-2017 | Endocrine Methods and Techniques

Patient-derived xenograft in zebrafish embryos: a new platform for translational research in neuroendocrine tumors

Authors: Germano Gaudenzi, Manuela Albertelli, Alessandra Dicitore, Roberto Würth, Federico Gatto, Federica Barbieri, Franco Cotelli, Tullio Florio, Diego Ferone, Luca Persani, Giovanni Vitale

Published in: Endocrine | Issue 2/2017

Login to get access

Abstract

Preclinical research on neuroendocrine tumors usually involves immortalized cell lines and few animal models. In the present study we described an in vivo model based on patient-derived xenografts of neuroendocrine tumor cells in zebrafish (Danio rerio) embryos, allowing a rapid analysis of the angiogenic and invasive potential. Patient-derived neuroendocrine tumor cells were transplanted in 48 hours post-fertilization Tg(fli1a:EGFP) y1 zebrafish embryos that express enhanced green fluorescent protein in the entire vasculature. Neuroendocrine tumor cells, stained with CM-Dil, were injected into the subperidermal (perivitelline) space, close to the developing subintestinal venous plexus. A proper control group, represented by zebrafish injected with only D-PBS, was included in this study. Angiogenic and invasive potentials of each patient-derived xenograft were evaluated by both epifluorescence and confocal microscopes. Six out of eight neuroendocrine tumor samples were successfully transplanted in zebrafish embryos. Although the implanted tumor mass had a limited size (about 100 cells for embryos), patient-derived xenografts showed pro-angiogenic (5 cases) and invasive (6 cases) behaviors within 48 hours post injection. Patient-derived xenograft in zebrafish embryos appears to be a reliable in vivo preclinical model for neuroendocrine tumors, tumors with often limited cell availability. The rapidity of this procedure makes our model a promising platform to perform preclinical drug screening and opens a new scenario for personalized treatment in patients with neuroendocrine tumors.
Literature
3.
go back to reference A. Walenkamp, G. Crespo, F. Fierro Maya, R. Fossmark, P. Igaz, A. Rinke, G. Tamagno, G. Vitale, K. Oberg, T. Meyer, Hallmarks of gastrointestinal neuroendocrine tumours: implications for treatment. Endocr. Relat. Cancer 21(6), R445–R460 (2014). doi:10.1530/ERC-14-0106ERC-14-0106 CrossRefPubMed A. Walenkamp, G. Crespo, F. Fierro Maya, R. Fossmark, P. Igaz, A. Rinke, G. Tamagno, G. Vitale, K. Oberg, T. Meyer, Hallmarks of gastrointestinal neuroendocrine tumours: implications for treatment. Endocr. Relat. Cancer 21(6), R445–R460 (2014). doi:10.​1530/​ERC-14-0106ERC-14-0106 CrossRefPubMed
9.
go back to reference N.D. Lawson, B.M. Weinstein, In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev. Biol. 248(2), 307–318 (2002). doi:S0012160602907116CrossRefPubMed N.D. Lawson, B.M. Weinstein, In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev. Biol. 248(2), 307–318 (2002). doi:S0012160602907116CrossRefPubMed
13.
go back to reference T. Florio, F. Barbieri, R. Spaziante, G. Zona, L.J. Hofland, P.M. van Koetsveld, R.A. Feelders, G.K. Stalla, M. Theodoropoulou, M.D. Culler, J. Dong, J.E. Taylor, J.P. Moreau, A. Saveanu, G. Gunz, H. Dufour, P. Jaquet, Efficacy of a dopamine-somatostatin chimeric molecule, BIM-23A760, in the control of cell growth from primary cultures of human non-functioning pituitary adenomas: a multi-center study. Endocr. Relat. Cancer 15(2), 583–596 (2008). doi:10.1677/ERC-07-027115/2/583 CrossRefPubMed T. Florio, F. Barbieri, R. Spaziante, G. Zona, L.J. Hofland, P.M. van Koetsveld, R.A. Feelders, G.K. Stalla, M. Theodoropoulou, M.D. Culler, J. Dong, J.E. Taylor, J.P. Moreau, A. Saveanu, G. Gunz, H. Dufour, P. Jaquet, Efficacy of a dopamine-somatostatin chimeric molecule, BIM-23A760, in the control of cell growth from primary cultures of human non-functioning pituitary adenomas: a multi-center study. Endocr. Relat. Cancer 15(2), 583–596 (2008). doi:10.​1677/​ERC-07-027115/​2/​583 CrossRefPubMed
14.
go back to reference A. Mohamed, M.P. Blanchard, M. Albertelli, F. Barbieri, T. Brue, P. Niccoli, J.R. Delpero, G. Monges, S. Garcia, D. Ferone, T. Florio, A. Enjalbert, V. Moutardier, A. Schonbrunn, C. Gerard, A. Barlier, A. Saveanu, Pasireotide and octreotide antiproliferative effects and sst2 trafficking in human pancreatic neuroendocrine tumor cultures. Endocr. Relat. Cancer 21(5), 691–704 (2014). doi:10.1530/ERC-14-0086ERC-14-0086 CrossRefPubMed A. Mohamed, M.P. Blanchard, M. Albertelli, F. Barbieri, T. Brue, P. Niccoli, J.R. Delpero, G. Monges, S. Garcia, D. Ferone, T. Florio, A. Enjalbert, V. Moutardier, A. Schonbrunn, C. Gerard, A. Barlier, A. Saveanu, Pasireotide and octreotide antiproliferative effects and sst2 trafficking in human pancreatic neuroendocrine tumor cultures. Endocr. Relat. Cancer 21(5), 691–704 (2014). doi:10.​1530/​ERC-14-0086ERC-14-0086 CrossRefPubMed
15.
go back to reference S. Nicoli, M. Presta, The zebrafish/tumor xenograft angiogenesis assay. Nat. Protoc. 2(11), 2918–2923 (2007). doi:nprot.2007.412 [pii]10.1038/nprot.2007.412CrossRefPubMed S. Nicoli, M. Presta, The zebrafish/tumor xenograft angiogenesis assay. Nat. Protoc. 2(11), 2918–2923 (2007). doi:nprot.2007.412 [pii]10.1038/nprot.2007.412CrossRefPubMed
22.
go back to reference C.L. Morton, P.J. Houghton, Establishment of human tumor xenografts in immunodeficient mice. Nat. Protoc. 2(2), 247–250 (2007). doi:nprot.2007.25 [pii]10.1038/nprot.2007.25CrossRefPubMed C.L. Morton, P.J. Houghton, Establishment of human tumor xenografts in immunodeficient mice. Nat. Protoc. 2(2), 247–250 (2007). doi:nprot.2007.25 [pii]10.1038/nprot.2007.25CrossRefPubMed
23.
go back to reference M.J. Puchner, D.K. Ludecke, W. Saeger, H.D. Herrmann, Use of athymic nude mice for in vivo studies of human growth-hormone-secreting pituitary adenomas. Horm. Res. 35(5), 198–204 (1991)CrossRefPubMed M.J. Puchner, D.K. Ludecke, W. Saeger, H.D. Herrmann, Use of athymic nude mice for in vivo studies of human growth-hormone-secreting pituitary adenomas. Horm. Res. 35(5), 198–204 (1991)CrossRefPubMed
24.
go back to reference J.H. Yang, J. Hu, L. Wan, L.J. Chen, Barbigerone inhibits tumor angiogenesis, growth and metastasis in melanoma. Asian Pac. J. Cancer Prev. 15(1), 167–174 (2014)CrossRefPubMed J.H. Yang, J. Hu, L. Wan, L.J. Chen, Barbigerone inhibits tumor angiogenesis, growth and metastasis in melanoma. Asian Pac. J. Cancer Prev. 15(1), 167–174 (2014)CrossRefPubMed
Metadata
Title
Patient-derived xenograft in zebrafish embryos: a new platform for translational research in neuroendocrine tumors
Authors
Germano Gaudenzi
Manuela Albertelli
Alessandra Dicitore
Roberto Würth
Federico Gatto
Federica Barbieri
Franco Cotelli
Tullio Florio
Diego Ferone
Luca Persani
Giovanni Vitale
Publication date
01-08-2017
Publisher
Springer US
Published in
Endocrine / Issue 2/2017
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-016-1048-9

Other articles of this Issue 2/2017

Endocrine 2/2017 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine