Skip to main content
Top
Published in: European Journal of Nuclear Medicine and Molecular Imaging 11/2022

Open Access 26-05-2022 | Pancreatic Cancer | Original Article

Imaging PARP with [18F]rucaparib in pancreatic cancer models

Authors: Chung Ying Chan, Zijun Chen, Gianluca Destro, Mathew Veal, Doreen Lau, Edward O’Neill, Gemma Dias, Michael Mosley, Veerle Kersemans, Florian Guibbal, Véronique Gouverneur, Bart Cornelissen

Published in: European Journal of Nuclear Medicine and Molecular Imaging | Issue 11/2022

Login to get access

Abstract

Purpose

Rucaparib, an FDA-approved PARP inhibitor, is used as a single agent in maintenance therapy to provide promising treatment efficacy with an acceptable safety profile in various types of BRCA-mutated cancers. However, not all patients receive the same benefit from rucaparib-maintenance therapy. A predictive biomarker to help with patient selection for rucaparib treatment and predict clinical benefit is therefore warranted. With this aim, we developed [18F]rucaparib, an 18F-labelled isotopologue of rucaparib, and employed it as a PARP-targeting agent for cancer imaging with PET. Here, we report the in vitro and in vivo evaluation of [18F]rucaparib in human pancreatic cancer models.

Method

We incorporated the positron-emitting 18F isotope into rucaparib, enabling its use as a PET imaging agent. [18F]rucaparib binds to the DNA damage repair enzyme, PARP, allowing direct visualisation and measurement of PARP in cancerous models before and after PARP inhibition or other genotoxic cancer therapies, providing critical information for cancer diagnosis and therapy. Proof-of-concept evaluations were determined in pancreatic cancer models.

Results

Uptake of [18F]rucaparib was found to be mainly dependent on PARP1 expression. Induction of DNA damage increased PARP expression, thereby increasing uptake of [18F]rucaparib. In vivo studies revealed relatively fast blood clearance of [18F]rucaparib in PSN1 tumour-bearing mice, with a tumour uptake of 5.5 ± 0.5%ID/g (1 h after i.v. administration). In vitro and in vivo studies showed significant reduction of [18F]rucaparib uptake by addition of different PARP inhibitors, indicating PARP-selective binding.

Conclusion

Taken together, we demonstrate the potential of [18F]rucaparib as a non-invasive PARP-targeting imaging agent for pancreatic cancers.
Appendix
Available only for authorised users
Literature
1.
go back to reference Montemagno C, Cassim S, De Leiris N, Durivault J, Faraggi M. Pancreatic ductal adenocarcinoma: the dawn of the era of nuclear Medicine? Int J Mol Sci. 2021;22:6413.PubMedPubMedCentralCrossRef Montemagno C, Cassim S, De Leiris N, Durivault J, Faraggi M. Pancreatic ductal adenocarcinoma: the dawn of the era of nuclear Medicine? Int J Mol Sci. 2021;22:6413.PubMedPubMedCentralCrossRef
3.
go back to reference Zhu H, Wei M, Xu J, et al. PARP inhibitors in pancreatic cancer: molecular mechanisms and clinical applications. Mol cancer. 2020;19:1–15. Zhu H, Wei M, Xu J, et al. PARP inhibitors in pancreatic cancer: molecular mechanisms and clinical applications. Mol cancer. 2020;19:1–15.
4.
go back to reference Zhang Z, Song J, Xie C, Pan J, Lu W, Liu M. Pancreatic cancer: recent progress of drugs in clinical trials. AAPS J. 2021;23:1–10.CrossRef Zhang Z, Song J, Xie C, Pan J, Lu W, Liu M. Pancreatic cancer: recent progress of drugs in clinical trials. AAPS J. 2021;23:1–10.CrossRef
5.
go back to reference Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15:333–48.PubMedCrossRef Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15:333–48.PubMedCrossRef
6.
go back to reference Chan CY, Tan KV, Cornelissen B. PARP inhibitors in cancer diagnosis and therapy. Clin Cancer Res. 2021;27:1585–94.PubMedCrossRef Chan CY, Tan KV, Cornelissen B. PARP inhibitors in cancer diagnosis and therapy. Clin Cancer Res. 2021;27:1585–94.PubMedCrossRef
7.
go back to reference Antolin AA, Ameratunga M, Banerji U, Clarke PA, Workman P, Al-Lazikani B. The kinase polypharmacology landscape of clinical PARP inhibitors. Sci Rep. 2020;10:1–14.CrossRef Antolin AA, Ameratunga M, Banerji U, Clarke PA, Workman P, Al-Lazikani B. The kinase polypharmacology landscape of clinical PARP inhibitors. Sci Rep. 2020;10:1–14.CrossRef
8.
go back to reference Knezevic CE, Wright G, Rix LLR, et al. Proteome-wide profiling of clinical PARP inhibitors reveals compound-specific secondary targets. Cell Chem Biol. 2016;23:1490–503.PubMedPubMedCentralCrossRef Knezevic CE, Wright G, Rix LLR, et al. Proteome-wide profiling of clinical PARP inhibitors reveals compound-specific secondary targets. Cell Chem Biol. 2016;23:1490–503.PubMedPubMedCentralCrossRef
9.
go back to reference Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33:244–51.PubMedCrossRef Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33:244–51.PubMedCrossRef
10.
11.
go back to reference Shroff RT, Hendifar A, McWilliams RR, et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. JCO Precis Oncol. 2018;2:1–15.PubMed Shroff RT, Hendifar A, McWilliams RR, et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. JCO Precis Oncol. 2018;2:1–15.PubMed
13.
go back to reference Noordermeer SM, van Attikum H. PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells. Trends Cell Biol. 2019;29:820–34.PubMedCrossRef Noordermeer SM, van Attikum H. PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells. Trends Cell Biol. 2019;29:820–34.PubMedCrossRef
14.
go back to reference Jannetti SA, Zeglis BM, Zalutsky MR, Reiner T. Poly (ADP-Ribose) polymerase (PARP) inhibitors and radiation therapy. Front Pharmacol. 2020;11:1–14.CrossRef Jannetti SA, Zeglis BM, Zalutsky MR, Reiner T. Poly (ADP-Ribose) polymerase (PARP) inhibitors and radiation therapy. Front Pharmacol. 2020;11:1–14.CrossRef
16.
go back to reference Bowden GD, Stotz S, Kinzler J, et al. DoE optimization empowers the automated preparation of enantiomerically pure [18F] talazoparib and its in vivo evaluation as a PARP radiotracer. 2021. 10.26434/chemrxiv.14587401.v1. Bowden GD, Stotz S, Kinzler J, et al. DoE optimization empowers the automated preparation of enantiomerically pure [18F] talazoparib and its in vivo evaluation as a PARP radiotracer. 2021. 10.26434/chemrxiv.14587401.v1.
17.
19.
go back to reference Guibbal F, Hopkins SL, Pacelli A, et al. [18 F] AZD2461, an insight on difference in PARP binding profiles for DNA damage response PET imaging. Mol Imaging Biol. 2020;22:1226–34.PubMedPubMedCentralCrossRef Guibbal F, Hopkins SL, Pacelli A, et al. [18 F] AZD2461, an insight on difference in PARP binding profiles for DNA damage response PET imaging. Mol Imaging Biol. 2020;22:1226–34.PubMedPubMedCentralCrossRef
21.
go back to reference Zhang H, Fu L. The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharm Sin B. 2021;11:1400–11.PubMedPubMedCentralCrossRef Zhang H, Fu L. The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharm Sin B. 2021;11:1400–11.PubMedPubMedCentralCrossRef
22.
go back to reference Murray J, Thomas H, Berry P, et al. Tumour cell retention of rucaparib, sustained PARP inhibition and efficacy of weekly as well as daily schedules. Br J Cancer. 2014;110:1977–84.PubMedPubMedCentralCrossRef Murray J, Thomas H, Berry P, et al. Tumour cell retention of rucaparib, sustained PARP inhibition and efficacy of weekly as well as daily schedules. Br J Cancer. 2014;110:1977–84.PubMedPubMedCentralCrossRef
23.
go back to reference Staszewski O, Nikolova T, Kaina B. Kinetics of γ-H2AX focus formation upon treatment of cells with UV light and alkylating agents. Environ Mol Mutagen. 2008;49:734–40.PubMedCrossRef Staszewski O, Nikolova T, Kaina B. Kinetics of γ-H2AX focus formation upon treatment of cells with UV light and alkylating agents. Environ Mol Mutagen. 2008;49:734–40.PubMedCrossRef
24.
go back to reference Fuchs RP, Isogawa A, Paulo JA, et al. Crosstalk between repair pathways elicits double-strand breaks in alkylated DNA and implications for the action of temozolomide. Elife. 2021;10:e69544.PubMedPubMedCentralCrossRef Fuchs RP, Isogawa A, Paulo JA, et al. Crosstalk between repair pathways elicits double-strand breaks in alkylated DNA and implications for the action of temozolomide. Elife. 2021;10:e69544.PubMedPubMedCentralCrossRef
25.
go back to reference Michel LS, Dyroff S, Brooks FJ, et al. PET of poly (ADP-ribose) polymerase activity in cancer: preclinical assessment and first in-human studies. Radiology. 2017;282:453–63.PubMedCrossRef Michel LS, Dyroff S, Brooks FJ, et al. PET of poly (ADP-ribose) polymerase activity in cancer: preclinical assessment and first in-human studies. Radiology. 2017;282:453–63.PubMedCrossRef
26.
27.
go back to reference McDonald ES, Doot RK, Pantel AR, et al. Positron emission tomography imaging of poly–(adenosine diphosphate–ribose) polymerase 1 expression in breast cancer: a nonrandomized clinical trial. JAMA Oncol. 2020;6:921–3.PubMedPubMedCentralCrossRef McDonald ES, Doot RK, Pantel AR, et al. Positron emission tomography imaging of poly–(adenosine diphosphate–ribose) polymerase 1 expression in breast cancer: a nonrandomized clinical trial. JAMA Oncol. 2020;6:921–3.PubMedPubMedCentralCrossRef
28.
go back to reference Sparidans RW, Durmus S, Schinkel AH, Schellens JH, Beijnen JH. Liquid chromatography–tandem mass spectrometric assay for the PARP inhibitor rucaparib in plasma. J Pharm Biomed. 2014;88:626–9.CrossRef Sparidans RW, Durmus S, Schinkel AH, Schellens JH, Beijnen JH. Liquid chromatography–tandem mass spectrometric assay for the PARP inhibitor rucaparib in plasma. J Pharm Biomed. 2014;88:626–9.CrossRef
29.
go back to reference Parrish KE, Cen L, Murray J, et al. Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther. 2015;14:2735–43.PubMedPubMedCentralCrossRef Parrish KE, Cen L, Murray J, et al. Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther. 2015;14:2735–43.PubMedPubMedCentralCrossRef
30.
go back to reference Edmonds CE, Makvandi M, Lieberman BP, et al. [18F] FluorThanatrace uptake as a marker of PARP1 expression and activity in breast cancer. Am J Nucl Med. 2016;6:94–101. Edmonds CE, Makvandi M, Lieberman BP, et al. [18F] FluorThanatrace uptake as a marker of PARP1 expression and activity in breast cancer. Am J Nucl Med. 2016;6:94–101.
31.
go back to reference Curtin NJ, Szabo C. Poly (ADP-ribose) polymerase inhibition: past, present and future. Nat Rev Drug Discov. 2020;19:711–36.PubMedCrossRef Curtin NJ, Szabo C. Poly (ADP-ribose) polymerase inhibition: past, present and future. Nat Rev Drug Discov. 2020;19:711–36.PubMedCrossRef
33.
go back to reference Salinas B, Irwin CP, Kossatz S, et al. Radioiodinated PARP1 tracers for glioblastoma imaging. EJNMMI Res. 2015;5:1–14.CrossRef Salinas B, Irwin CP, Kossatz S, et al. Radioiodinated PARP1 tracers for glioblastoma imaging. EJNMMI Res. 2015;5:1–14.CrossRef
34.
go back to reference Anderson R-C, Makvandi M, Xu K, et al. Iodinated benzimidazole PARP radiotracer for evaluating PARP1/2 expression in vitro and in vivo. Nucl Med Biol. 2016;43:752–8.PubMedCrossRef Anderson R-C, Makvandi M, Xu K, et al. Iodinated benzimidazole PARP radiotracer for evaluating PARP1/2 expression in vitro and in vivo. Nucl Med Biol. 2016;43:752–8.PubMedCrossRef
35.
go back to reference Liao M, Watkins S, Nash E, et al. Evaluation of absorption, distribution, metabolism, and excretion of [14 C]-rucaparib, a poly (ADP-ribose) polymerase inhibitor, in patients with advanced solid tumors. Invest New Drugs. 2020;38:765–75.PubMedCrossRef Liao M, Watkins S, Nash E, et al. Evaluation of absorption, distribution, metabolism, and excretion of [14 C]-rucaparib, a poly (ADP-ribose) polymerase inhibitor, in patients with advanced solid tumors. Invest New Drugs. 2020;38:765–75.PubMedCrossRef
36.
go back to reference Kossatz S, Carney B, Farley C, Weber WA, Drain CM, Reiner T. Direct imaging of drug distribution and target engagement of the PARP inhibitor rucaparib. J Nucl Med. 2018;59:1316–20.PubMedPubMedCentralCrossRef Kossatz S, Carney B, Farley C, Weber WA, Drain CM, Reiner T. Direct imaging of drug distribution and target engagement of the PARP inhibitor rucaparib. J Nucl Med. 2018;59:1316–20.PubMedPubMedCentralCrossRef
37.
Metadata
Title
Imaging PARP with [18F]rucaparib in pancreatic cancer models
Authors
Chung Ying Chan
Zijun Chen
Gianluca Destro
Mathew Veal
Doreen Lau
Edward O’Neill
Gemma Dias
Michael Mosley
Veerle Kersemans
Florian Guibbal
Véronique Gouverneur
Bart Cornelissen
Publication date
26-05-2022
Publisher
Springer Berlin Heidelberg
Published in
European Journal of Nuclear Medicine and Molecular Imaging / Issue 11/2022
Print ISSN: 1619-7070
Electronic ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-022-05835-4

Other articles of this Issue 11/2022

European Journal of Nuclear Medicine and Molecular Imaging 11/2022 Go to the issue