Article Text

Download PDFPDF
Original article
Claudin-4-targeted optical imaging detects pancreatic cancer and its precursor lesions
  1. Albrecht Neesse1,2,
  2. Anke Hahnenkamp3,
  3. Heidi Griesmann1,
  4. Malte Buchholz1,
  5. Stefan A Hahn4,
  6. Abdelouahid Maghnouj4,
  7. Volker Fendrich5,
  8. Janine Ring3,
  9. Bence Sipos6,
  10. David A Tuveson2,
  11. Christoph Bremer7,
  12. Thomas M Gress1,
  13. Patrick Michl1
  1. 1Department of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Marburg, Germany
  2. 2Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
  3. 3Institute of Clinical Radiology, University Hospital of Münster, Münster, Germany
  4. 4Molecular GI Oncology, Department of Internal Medicine, Ruhr-University Bochum, Bochum, Germany
  5. 5Department of Visceral Surgery, University of Marburg, Marburg, Germany
  6. 6Department of Pathology and Neuropathology, University Hospital of Tuebingen, Tuebingen, Gemany
  7. 7Department of Radiology St Franziskus Hospital, Muenster, Germany
  1. Correspondence to Professor Dr Thomas Gress, Division of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Baldinger Strasse, 35043 Marburg, Germany; thomas.gress{at}med.uni-marburg.de

Abstract

Objectives Novel imaging methods based on specific molecular targets to detect both established neoplasms and their precursor lesions are highly desirable in cancer medicine. Previously, we identified claudin-4, an integral constituent of tight junctions, as highly expressed in various gastrointestinal tumours including pancreatic cancer. Here, we investigate the potential of targeting claudin-4 with a naturally occurring ligand to visualise pancreatic cancer and its precursor lesions in vitro and in vivo by near-infrared imaging approaches.

Design A non-toxic C-terminal fragment of the claudin-4 ligand Clostridium perfringens enterotoxin (C-CPE) was labelled with a cyanine dye (Cy5.5). Binding of the optical tracer was analysed on claudin-4 positive and negative cells in vitro, and tumour xenografts in vivo. In addition, two genetically engineered mouse models for pancreatic intraepithelial neoplasia (PanIN) and pancreatic cancer were used for in vivo validation. Optical imaging studies were conducted using 2D planar fluorescence reflectance imaging (FRI) technology and 3D fluorescence-mediated tomography (FMT).

Results In vitro, the peptide-dye conjugate showed high binding affinity to claudin-4 positive CAPAN1 cells, while claudin-4 negative HT1080 cells revealed little or no fluorescence. In vivo, claudin-4 positive tumour xenografts, endogenous pancreatic tumours, hepatic metastases, as well as preinvasive PanIN lesions, were visualised by FRI and FMT up to 48 h after injection showing a significantly higher average of fluorochrome concentration as compared with claudin-4 negative xenografts and normal pancreatic tissue.

Conclusions C-CPE-Cy5.5 combined with novel optical imaging methods enables non-invasive visualisation of claudin-4 positive murine pancreatic tumours and their precursor lesions, representing a promising modality for early diagnostic imaging.

  • Clostridium perfringens enterotoxin
  • fluorescence reflectance imaging
  • pancreatic cancer
  • PanIN
  • claudin-4
  • pancreatic fibrosis
  • pancreatic disease
  • pancreatic tumours
  • imaging
  • pancreatic damage
  • image analysis
  • adenocarcinoma
  • molecular biology
  • cancer genetics
  • pancreas
  • colorectal cancer genes
  • abdominal MRI
  • endoscopy
  • gene expression
  • pancreatitis
  • cancer
  • gastrointestinal cancer
  • cell migration
  • carcinogenesis

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Footnotes

  • AN and AH contributed equally to the manuscript.

  • Funding This work was supported in part by the Deutsche Krebshilfe (to PM and TG), Mildred Scheel Postdoctoral Fellowship by the Deutsche Krebshilfe (to AN), the Deutsche Forschungsgemeinschaft (DFG, KFO210 to PM and SFB656 to C.B.) and the German Ministry of Education and Research (BMBF, NGFN program of medical genome research ‘PaCa-Net’; project ID PKB-01GS08) to MB, TG, BS and SH), the European Union (6th framework program MolDiag-PaCa to TG. and the 7th framework program EPC-TM net to TG, PM, MB, DT), the state of Hessen (LOEWE, to PM, MB, TG) and the IZKF Core unit of the University of Muenster. This research was also supported by Cancer Research UK, The Li Ka Shing Foundation and Hutchison Whampoa Limited (DT).

  • Competing interests None.

  • Provenance and peer review Not commissioned; externally peer reviewed.

  • Data sharing statement All material and protocols published in this manuscript including also unpublished material and protocols will be made available to all other interested scientists upon request via email or surface mail.