Cargando…

Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma

Discrimination of pancreatic ductal adenocarcinoma (PDAC) from chronic pancreatitis (CP) or peritumoral inflammation is challenging, both at preoperative imaging and during surgery, but it is crucial for proper therapy selection. Tumor-specific molecular imaging aims to enhance this discrimination a...

Descripción completa

Detalles Bibliográficos
Autores principales: Tummers, Willemieke S., Farina-Sarasqueta, Arantza, Boonstra, Martin C., Prevoo, Hendrica A., Sier, Cornelis F., Mieog, Jan S., Morreau, Johannes, van Eijck, Casper H., Kuppen, Peter J., van de Velde, Cornelis J., Bonsing, Bert A., Vahrmeijer, Alexander L., Swijnenburg, Rutger-Jan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5593604/
https://www.ncbi.nlm.nih.gov/pubmed/28915633
http://dx.doi.org/10.18632/oncotarget.18232
_version_ 1783263071431557120
author Tummers, Willemieke S.
Farina-Sarasqueta, Arantza
Boonstra, Martin C.
Prevoo, Hendrica A.
Sier, Cornelis F.
Mieog, Jan S.
Morreau, Johannes
van Eijck, Casper H.
Kuppen, Peter J.
van de Velde, Cornelis J.
Bonsing, Bert A.
Vahrmeijer, Alexander L.
Swijnenburg, Rutger-Jan
author_facet Tummers, Willemieke S.
Farina-Sarasqueta, Arantza
Boonstra, Martin C.
Prevoo, Hendrica A.
Sier, Cornelis F.
Mieog, Jan S.
Morreau, Johannes
van Eijck, Casper H.
Kuppen, Peter J.
van de Velde, Cornelis J.
Bonsing, Bert A.
Vahrmeijer, Alexander L.
Swijnenburg, Rutger-Jan
author_sort Tummers, Willemieke S.
collection PubMed
description Discrimination of pancreatic ductal adenocarcinoma (PDAC) from chronic pancreatitis (CP) or peritumoral inflammation is challenging, both at preoperative imaging and during surgery, but it is crucial for proper therapy selection. Tumor-specific molecular imaging aims to enhance this discrimination and to help select and stratify patients for resection. We evaluated various biomarkers for the specific identification of PDAC and associated lymph node metastases. Using immunohistochemistry (IHC), expression levels and patterns were investigated of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), Cathepsin E (Cath E), epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (c-MET), thymocyte differentiation antigen 1 (Thy1), and urokinase-type plasminogen activator receptor (uPAR). In a first cohort, multiple types of pancreatic tissue were evaluated (n=62); normal pancreatic tissue (n=8), CP (n=7), PDAC (n=9), tumor associated lymph nodes (n=32), and PDAC after neoadjuvant radiochemotherapy (n=6). In a second cohort, tissues were investigated (n=55) with IHC and immunofluorescence (IF) for concordance of biomarker expression in all tissue types, obtained from an individual patient. Integrin αvβ6 and CEACAM5 showed significantly higher expression levels in PDAC versus normal pancreatic tissue (P=0.001 and P<0.001, respectively) and CP (P=0.003 and P<0.001, respectively). Avβ6 and CEACAM5 expression identified tumor-positive lymph nodes correctly in 84% and 68%, respectively, and in 100% of tumor-negative nodes for both biomarkers. In conclusion, αvβ6 and CEACAM5 are excellent biomarkers to differentiate PDAC from surrounding tissue and to identify lymph node metastases. Individually or combined, these biomarkers are promising targets for tumor-specific molecular imaging of PDAC.
format Online
Article
Text
id pubmed-5593604
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-55936042017-09-14 Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma Tummers, Willemieke S. Farina-Sarasqueta, Arantza Boonstra, Martin C. Prevoo, Hendrica A. Sier, Cornelis F. Mieog, Jan S. Morreau, Johannes van Eijck, Casper H. Kuppen, Peter J. van de Velde, Cornelis J. Bonsing, Bert A. Vahrmeijer, Alexander L. Swijnenburg, Rutger-Jan Oncotarget Research Paper Discrimination of pancreatic ductal adenocarcinoma (PDAC) from chronic pancreatitis (CP) or peritumoral inflammation is challenging, both at preoperative imaging and during surgery, but it is crucial for proper therapy selection. Tumor-specific molecular imaging aims to enhance this discrimination and to help select and stratify patients for resection. We evaluated various biomarkers for the specific identification of PDAC and associated lymph node metastases. Using immunohistochemistry (IHC), expression levels and patterns were investigated of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), Cathepsin E (Cath E), epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (c-MET), thymocyte differentiation antigen 1 (Thy1), and urokinase-type plasminogen activator receptor (uPAR). In a first cohort, multiple types of pancreatic tissue were evaluated (n=62); normal pancreatic tissue (n=8), CP (n=7), PDAC (n=9), tumor associated lymph nodes (n=32), and PDAC after neoadjuvant radiochemotherapy (n=6). In a second cohort, tissues were investigated (n=55) with IHC and immunofluorescence (IF) for concordance of biomarker expression in all tissue types, obtained from an individual patient. Integrin αvβ6 and CEACAM5 showed significantly higher expression levels in PDAC versus normal pancreatic tissue (P=0.001 and P<0.001, respectively) and CP (P=0.003 and P<0.001, respectively). Avβ6 and CEACAM5 expression identified tumor-positive lymph nodes correctly in 84% and 68%, respectively, and in 100% of tumor-negative nodes for both biomarkers. In conclusion, αvβ6 and CEACAM5 are excellent biomarkers to differentiate PDAC from surrounding tissue and to identify lymph node metastases. Individually or combined, these biomarkers are promising targets for tumor-specific molecular imaging of PDAC. Impact Journals LLC 2017-05-26 /pmc/articles/PMC5593604/ /pubmed/28915633 http://dx.doi.org/10.18632/oncotarget.18232 Text en Copyright: © 2017 Tummers et al. http://creativecommons.org/licenses/by/3.0/ This article is distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0/) (CC-BY), which permits unrestricted use and redistribution provided that the original author and source are credited.
spellingShingle Research Paper
Tummers, Willemieke S.
Farina-Sarasqueta, Arantza
Boonstra, Martin C.
Prevoo, Hendrica A.
Sier, Cornelis F.
Mieog, Jan S.
Morreau, Johannes
van Eijck, Casper H.
Kuppen, Peter J.
van de Velde, Cornelis J.
Bonsing, Bert A.
Vahrmeijer, Alexander L.
Swijnenburg, Rutger-Jan
Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title_full Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title_fullStr Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title_full_unstemmed Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title_short Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
title_sort selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5593604/
https://www.ncbi.nlm.nih.gov/pubmed/28915633
http://dx.doi.org/10.18632/oncotarget.18232
work_keys_str_mv AT tummerswillemiekes selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT farinasarasquetaarantza selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT boonstramartinc selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT prevoohendricaa selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT siercornelisf selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT mieogjans selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT morreaujohannes selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT vaneijckcasperh selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT kuppenpeterj selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT vandeveldecornelisj selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT bonsingberta selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT vahrmeijeralexanderl selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma
AT swijnenburgrutgerjan selectionofoptimalmoleculartargetsfortumorspecificimaginginpancreaticductaladenocarcinoma