Please use this identifier to cite or link to this item: http://bura.brunel.ac.uk/handle/2438/29407
Title: [<sup>123</sup>I]CC1: A PARP-Targeting, Auger Electron–Emitting Radiopharmaceutical for Radionuclide Therapy of Cancer
Other Titles: [123I]CC1: A PARP-Targeting, Auger Electron–Emitting Radiopharmaceutical for Radionuclide Therapy of Cancer
Authors: Chan, CY
Chen, Z
Guibbal, F
Dias, G
Destro, G
O’Neill, E
Veal, M
Lau, D
Mosley, M
Wilson, TC
Gouverneur, V
Cornelissen, B
Keywords: PARP;radionuclide therapy;radiopharmaceuticals;<sup>123</sup>I;Auger
Issue Date: 28-Sep-2023
Publisher: Society of Nuclear Medicine and Molecular Imaging
Citation: Chan, C.Y. et al. (2023) '[<sup>123</sup>I]CC1: A PARP-Targeting, Auger Electron–Emitting Radiopharmaceutical for Radionuclide Therapy of Cancer', Journal of Nuclear Medicine, 64 (12), pp. 1965 - 1971. doi: 10.2967/jnumed.123.265429.
Abstract: Poly(adenosine diphosphate ribose) polymerase (PARP) has emerged as an effective therapeutic strategy against cancer that targets the DNA damage repair enzyme. PARP-targeting compounds radiolabeled with an Auger electron–emitting radionuclide can be trapped close to damaged DNA in tumor tissue, where high ionizing potential and short range lead Auger electrons to kill cancer cells through the creation of complex DNA damage, with minimal damage to surrounding normal tissue. Here, we report on [<sup>123</sup>I]CC1, an <sup>123</sup>I-labeled PARP inhibitor for radioligand therapy of cancer. Methods: Copper-mediated <sup>123</sup>I iododeboronation of a boronic pinacol ester precursor afforded [<sup>123</sup>I]CC1. The level and specificity of cell uptake and the therapeutic efficacy of [<sup>123</sup>I]CC1 were determined in human breast carcinoma, pancreatic adenocarcinoma, and glioblastoma cells. Tumor uptake and tumor growth inhibition of [<sup>123</sup>I]CC1 were assessed in mice bearing human cancer xenografts (MDA-MB-231, PSN1, and U87MG). Results: In vitro and in vivo studies showed selective uptake of [<sup>123</sup>I]CC1 in all models. Significantly reduced clonogenicity, a proxy for tumor growth inhibition by ionizing radiation in vivo, was observed in vitro after treatment with as little as 10 Bq [<sup>123</sup>I]CC1. Biodistribution at 1 h after intravenous administration showed PSN1 tumor xenograft uptake of 0.9 ± 0.06 percentage injected dose per gram of tissue. Intravenous administration of a relatively low amount of [<sup>123</sup>I]CC1 (3 MBq) was able to significantly inhibit PSN1 xenograft tumor growth but was less effective in xenografts that expressed less PARP. [<sup>123</sup>I]CC1 did not cause significant toxicity to normal tissues. Conclusion: Taken together, these results show the potential of [<sup>123</sup>I]CC1 as a radioligand therapy for PARP-expressing cancers.
URI: https://bura.brunel.ac.uk/handle/2438/29407
DOI: https://doi.org/10.2967/jnumed.123.265429
ISSN: 0161-5505
Other Identifiers: ORCiD: Doreen Lau https://orcid.org/0000-0002-7623-2401
Appears in Collections:Dept of Life Sciences Research Papers

Files in This Item:
File Description SizeFormat 
FullText.pdfCopyright & Usage © 2023 by the Society of Nuclear Medicine and Molecular Imaging. Immediate Open Access: Creative Commons Attribution 4.0 International License (CC BY) allows users to share and adapt with attribution, excluding materials credited to previous publications. License: https://creativecommons.org/licenses/by/4.0/. Details: https://jnm.snmjournals.org/site/misc/permission.xhtml.1.32 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons