Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.

Patient-derived xenograft (PDX) tumors are powerful tools to study cancer biology. However, the ability of PDX tumors to model the biological and histological diversity of pancreatic ductal adenocarcinoma (PDAC) is not well known. In this study, we subcutaneously implanted 133 primary and metastatic...

Full description

Bibliographic Details
Main Authors: Ilaria Pergolini, Vicente Morales-Oyarvide, Mari Mino-Kenudson, Kim C Honselmann, Matthew W Rosenbaum, Sabikun Nahar, Marina Kem, Cristina R Ferrone, Keith D Lillemoe, Nabeel Bardeesy, David P Ryan, Sarah P Thayer, Andrew L Warshaw, Carlos Fernández-Del Castillo, Andrew S Liss
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2017-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC5576681?pdf=render
id doaj-bf4d5669542f4d358dfa11126546b659
record_format Article
spelling doaj-bf4d5669542f4d358dfa11126546b6592020-11-24T21:54:06ZengPublic Library of Science (PLoS)PLoS ONE1932-62032017-01-01128e018285510.1371/journal.pone.0182855Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.Ilaria PergoliniVicente Morales-OyarvideMari Mino-KenudsonKim C HonselmannMatthew W RosenbaumSabikun NaharMarina KemCristina R FerroneKeith D LillemoeNabeel BardeesyDavid P RyanSarah P ThayerAndrew L WarshawCarlos Fernández-Del CastilloAndrew S LissPatient-derived xenograft (PDX) tumors are powerful tools to study cancer biology. However, the ability of PDX tumors to model the biological and histological diversity of pancreatic ductal adenocarcinoma (PDAC) is not well known. In this study, we subcutaneously implanted 133 primary and metastatic PDAC tumors into immunodeficient mice. Fifty-seven tumors were successfully engrafted and even after extensive passaging, the histology of poorly-, moderately-, and well-differentiated tumors was maintained in the PDX models. Moreover, the fibroblast and collagen contents in the stroma of patient tumors were recapitulated in the corresponding PDX models. Analysis of the clinicopathological features of patients revealed xenograft tumor engraftment was associated with lymphovascular invasion (P = 0.001) and worse recurrence-free (median, 7 vs. 16 months, log-rank P = 0.047) and overall survival (median, 13 vs. 21 months, log-rank P = 0.038). Among successful engraftments, median time of growth required for reimplantation into new mice was 151 days. Reflective of the inherent biological diversity between PDX tumors with rapid (<151 days) and slow growth, differences in their growth were maintained during extensive passaging. Rapid growth was additionally associated with lymph node metastasis (P = 0.022). The association of lymphovascular invasion and lymph node metastasis with PDX formation and rapid growth may reflect an underlying biological mechanism that allows these tumors to adapt and grow in a new environment. While the ability of PDX tumors to mimic the cellular and non-cellular features of the parental tumor stroma provides a valuable model to study the interaction of PDAC cells with the tumor microenvironment, the association of successful engraftment with adverse clinicopathological features suggests PDX models over represent more aggressive forms of this disease.http://europepmc.org/articles/PMC5576681?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Ilaria Pergolini
Vicente Morales-Oyarvide
Mari Mino-Kenudson
Kim C Honselmann
Matthew W Rosenbaum
Sabikun Nahar
Marina Kem
Cristina R Ferrone
Keith D Lillemoe
Nabeel Bardeesy
David P Ryan
Sarah P Thayer
Andrew L Warshaw
Carlos Fernández-Del Castillo
Andrew S Liss
spellingShingle Ilaria Pergolini
Vicente Morales-Oyarvide
Mari Mino-Kenudson
Kim C Honselmann
Matthew W Rosenbaum
Sabikun Nahar
Marina Kem
Cristina R Ferrone
Keith D Lillemoe
Nabeel Bardeesy
David P Ryan
Sarah P Thayer
Andrew L Warshaw
Carlos Fernández-Del Castillo
Andrew S Liss
Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
PLoS ONE
author_facet Ilaria Pergolini
Vicente Morales-Oyarvide
Mari Mino-Kenudson
Kim C Honselmann
Matthew W Rosenbaum
Sabikun Nahar
Marina Kem
Cristina R Ferrone
Keith D Lillemoe
Nabeel Bardeesy
David P Ryan
Sarah P Thayer
Andrew L Warshaw
Carlos Fernández-Del Castillo
Andrew S Liss
author_sort Ilaria Pergolini
title Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
title_short Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
title_full Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
title_fullStr Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
title_full_unstemmed Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
title_sort tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2017-01-01
description Patient-derived xenograft (PDX) tumors are powerful tools to study cancer biology. However, the ability of PDX tumors to model the biological and histological diversity of pancreatic ductal adenocarcinoma (PDAC) is not well known. In this study, we subcutaneously implanted 133 primary and metastatic PDAC tumors into immunodeficient mice. Fifty-seven tumors were successfully engrafted and even after extensive passaging, the histology of poorly-, moderately-, and well-differentiated tumors was maintained in the PDX models. Moreover, the fibroblast and collagen contents in the stroma of patient tumors were recapitulated in the corresponding PDX models. Analysis of the clinicopathological features of patients revealed xenograft tumor engraftment was associated with lymphovascular invasion (P = 0.001) and worse recurrence-free (median, 7 vs. 16 months, log-rank P = 0.047) and overall survival (median, 13 vs. 21 months, log-rank P = 0.038). Among successful engraftments, median time of growth required for reimplantation into new mice was 151 days. Reflective of the inherent biological diversity between PDX tumors with rapid (<151 days) and slow growth, differences in their growth were maintained during extensive passaging. Rapid growth was additionally associated with lymph node metastasis (P = 0.022). The association of lymphovascular invasion and lymph node metastasis with PDX formation and rapid growth may reflect an underlying biological mechanism that allows these tumors to adapt and grow in a new environment. While the ability of PDX tumors to mimic the cellular and non-cellular features of the parental tumor stroma provides a valuable model to study the interaction of PDAC cells with the tumor microenvironment, the association of successful engraftment with adverse clinicopathological features suggests PDX models over represent more aggressive forms of this disease.
url http://europepmc.org/articles/PMC5576681?pdf=render
work_keys_str_mv AT ilariapergolini tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT vicentemoralesoyarvide tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT mariminokenudson tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT kimchonselmann tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT matthewwrosenbaum tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT sabikunnahar tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT marinakem tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT cristinarferrone tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT keithdlillemoe tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT nabeelbardeesy tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT davidpryan tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT sarahpthayer tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT andrewlwarshaw tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT carlosfernandezdelcastillo tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
AT andrewsliss tumorengraftmentinpatientderivedxenograftsofpancreaticductaladenocarcinomaisassociatedwithadverseclinicopathologicalfeaturesandpoorsurvival
_version_ 1725868952134877184