Cookies on this website

We use cookies to ensure that we give you the best experience on our website. If you click 'Accept all cookies' we'll assume that you are happy to receive all cookies and you won't see this message again. If you click 'Reject all non-essential cookies' only necessary cookies providing core functionality such as security, network management, and accessibility will be enabled. Click 'Find out more' for information on how to change your cookie settings.

Introduction: Overexpression of tight junction protein claudin-4 has been detected in primary and metastatic pancreatic cancer tissue, and is associated with better prognosis in patients. Non-invasive measurement of claudin-4 expression by imaging methods could provide a means for accelerating detection and stratifying patients into risk groups. Clostridium perfringens enterotoxin (CPE) is a natural ligand for claudin-4 and holds potential as a targeting vector for molecular imaging of claudin-4 overexpression. A glutathione S-transferases (GST)-tagged version of the C-terminus of CPE (cCPE) was previously used to delineate claudin-4 overexpression by SPECT, but showed modest binding affinity and slow blood clearance in vivo. Materials and Methods: Based on the crystal structure of cCPE, a series of smaller-sized cCPE194-319 mutants with putatively improved binding affinity for claudin-4 were generated by site-directed mutagenesis (SDM). All peptides were conjugated site-specifically on a C-terminal cysteine using maleimide-DTPA to enable radiolabelling with 111In. The binding affinity of all radioconjugates was evaluated in claudin-4-expressing PSN-1 cells and HT1080 negative controls. The specificity of all cCPE mutants to claudin-4 was assessed in HT1080 cells stably transfected with hCLDN4. SPECT/CT imaging of BALB/c Nude mice bearing PSN-1 or HT1080 tumour xenografts was performed to determine the claudin-4-targeting ability of these peptides in vivo. Results: Uptake of all cCPE-based radioconjugates was significantly higher in PSN-1 cells compared to HT1080 negative controls. All peptides showed a marked improvement in affinity for claudin-4 in vitro when compared to previously reported values (KD values of 2.2 ± 0.8, 3 ± 0.1, 4.2 ± 0.5 nM, 10 ± 0.9 and 9.7 ± 0.7 nM). Blood clearance of [111In]In-cCPE194-319, as measured by SPECT, was considerably faster when compared to that of [111In]In-cCPE.GST (t1/2<1 min). All radiopeptides showed significantly higher accumulation in PSN-1 xenografts than HT1080 tumours at 90 min post-injection of the tracer ([111In]In-cCPE194-319: 2.7 ± 0.8 vs 0.4 ± 0.1 %ID/g, P<0.001; [111In]In-S313A: 2.3 ± 0.9 vs 0.5 ± 0.1 %ID/g, P<0.01; [111In]In- S307A+N309A+S313A: 2 ± 0.4 vs 0.3 ± 0.1 %ID/g, P<0.01; [111In]In- D284A: 2 ± 0.2 vs 0.7 ± 0.1 %ID/g, P<0.05; [111In]In- L254F+K257D: 6.3 ± 0.9 vs 0.7 ± 0.2 %ID/g, P<0.001). Conclusion: These optimised cCPE-based SPECT imaging agents show great promise as claudin-4-targeting vectors for in vivo imaging of claudin-4 overexpression in pancreatic cancer.

Original publication

DOI

10.2967/jnumed.120.243113

Type

Journal article

Journal

J Nucl Med

Publication Date

15/05/2020

Keywords

Animal Imaging, Oncology: Pancreas, Pancreatic Ductal Adenocarcinoma, Radiopharmaceuticals, cCPE, claudin