PET of Poly (ADP-Ribose) Polymerase Activity in Cancer: Preclinical Assessment and First In-Human Studies

Published Online:https://doi.org/10.1148/radiol.2016161929

Our data support the feasibility of evaluating fluorine 18 fluorthanatrace as a quantifiable biomarker of poly (adenosine diphosphate–ribose) polymerase expression in clinical trials.

Purpose

To demonstrate that positron emission tomography (PET) with fluorine 18 (18F) fluorthanatrace (FTT) depicts activated poly (adenosine diphosphate–ribose)polymerase (PARP) expression and is feasible for clinical trial evaluation.

Materials and Methods

All studies were conducted prospectively from February 2012 through July 2015 under protocols approved by the local animal studies committee and institutional review board. The area under the receiver operating characteristic curve (AUC, in g/mL· min) for 18F-FTT was assessed in normal mouse organs before and after treatment with olaparib (n = 14), a PARP inhibitor, or iniparib (n = 11), which has no PARP inhibitory activity. Murine biodistribution studies were performed to support human translational studies. Eight human subjects with cancer and eight healthy volunteers underwent imaging to verify the human radiation dosimetry of 18F-FTT. The Wilcoxon signed rank test was used to assess for differences among treatment groups for the mouse studies.

Results

In mice, olaparib, but not iniparib, significantly reduced the 18F-FTT AUC in the spine (median difference before and after treatment and interquartile range [IQR]: −17 g/mL· min and 10 g/mL · min, respectively [P = .0001], for olaparib and −3 g/mL · min and 13 g/mL · min [P = .70] for iniparib) and in nodes (median difference and interquartile range [IQR] before and after treatment: −23 g/mL · min and 13 g/mL · min [P = .0001] for olaparib; −9 g/mL · min and 17 g/mL · min [P = .05] for iniparib). The effective dose was estimated at 6.9 mSv for a 370-MBq 18F-FTT dose in humans. In humans, the organs with the highest uptake on images were the spleen and pancreas. Among five subjects with measurable tumors, increased 18F-FTT uptake was seen in one subject with pancreatic adenocarcinoma and another with liver cancer.

Conclusion

The results suggest that 18F-FTT uptake reflects PARP expression and that its radiation dosimetry profile is compatible with those of agents currently in clinical use.

© RSNA, 2016

Online supplemental material is available for this article.

References

  • 1. Gibson BA, Kraus WL. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat Rev Mol Cell Biol 2012;13(7):411–424.
  • 2. Schreiber V, Amé JC, Dollé P, et al. Poly(ADP-ribose) polymerase-2 (PARP-2) is required for efficient base excision DNA repair in association with PARP-1 and XRCC1. J Biol Chem 2002;277(25):23028–23036.
  • 3. Beck C, Robert I, Reina-San-Martin B, Schreiber V, Dantzer F. Poly(ADP-ribose) polymerases in double-strand break repair: focus on PARP1, PARP2 and PARP3. Exp Cell Res 2014;329(1):18–25.
  • 4. Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005;434(7035):913–917.
  • 5. Senra JM, Telfer BA, Cherry KE, et al. Inhibition of PARP-1 by olaparib (AZD2281) increases the radiosensitivity of a lung tumor xenograft. Mol Cancer Ther 2011;10(10):1949–1958.
  • 6. Turner NC, Ashworth A. Biomarkers of PARP inhibitor sensitivity. Breast Cancer Res Treat 2011;127(1):283–286.
  • 7. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly(ADP-ribose) polymerase (PARP) inhibitor lethality in homologous recombination-deficient cells. Proc Natl Acad Sci U S A 2011;108(8):3406–3411.
  • 8. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009;361(2):123–134.
  • 9. Fong PC, Yap TA, Boss DS, et al. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol 2010;28(15):2512–2519.
  • 10. Pyndiah S, Tanida S, Ahmed KM, Cassimere EK, Choe C, Sakamuro D. c-MYC suppresses BIN1 to release poly(ADP-ribose) polymerase 1: a mechanism by which cancer cells acquire cisplatin resistance. Sci Signal 2011;4(166):ra19.
  • 11. Drew Y. The development of PARP inhibitors in ovarian cancer: from bench to bedside. Br J Cancer 2015;113(Suppl 1):S3–S9.
  • 12. Bennet N. No survival benefit of olaparib in ovarian cancer patients. Lancet Oncol 2012;13(5):e192.
  • 13. Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med 2012;366(15):1382–1392.
  • 14. Gelmon KA, Tischkowitz M, Mackay H, et al. Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study. Lancet Oncol 2011;12(9):852–861.
  • 15. Plummer ER, Middleton MR, Jones C, et al. Temozolomide pharmacodynamics in patients with metastatic melanoma: DNA damage and activity of repair enzymes O6-alkylguanine alkyltransferase and poly(ADP-ribose) polymerase-1. Clin Cancer Res 2005;11(9):3402–3409.
  • 16. Bundred N, Gardovskis J, Jaskiewicz J, et al. Evaluation of the pharmacodynamics and pharmacokinetics of the PARP inhibitor olaparib: a phase I multicentre trial in patients scheduled for elective breast cancer surgery. Invest New Drugs 2013;31(4):949–958.
  • 17. Yang SX, Kummar S, Steinberg SM, et al. Immunohistochemical detection of poly(ADP-ribose) polymerase inhibition by ABT-888 in patients with refractory solid tumors and lymphomas. Cancer Biol Ther 2009;8(21):2004–2009.
  • 18. Reiner T, Lacy J, Keliher EJ, et al. Imaging therapeutic PARP inhibition in vivo through bioorthogonally developed companion imaging agents. Neoplasia 2012;14(3):169–177.
  • 19. Zhou D, Chu W, Xu J, et al. Synthesis, [18F] radiolabeling, and evaluation of poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors for in vivo imaging of PARP-1 using positron emission tomography. Bioorg Med Chem 2014;22(5):1700–1707.
  • 20. Edmonds CE, Makvandi M, Lieberman BP, et al. [(18)F]Fluorthanatrace uptake as a marker of PARP1 expression and activity in breast cancer. Am J Nucl Med Mol Imaging 2016;6(1):94–101.
  • 21. Liu X, Shi Y, Maag DX, et al. Iniparib nonselectively modifies cysteine-containing proteins in tumor cells and is not a bona fide PARP inhibitor. Clin Cancer Res 2012;18(2):510–523.
  • 22. Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, Kaufmann SH. Failure of iniparib to inhibit poly(ADP-ribose) polymerase in vitro. Clin Cancer Res 2012;18(6):1655–1662.
  • 23. Michels J, Vitale I, Galluzzi L, et al. Cisplatin resistance associated with PARP hyperactivation. Cancer Res 2013;73(7):2271–2280.
  • 24. Herrero P, Laforest R, Shoghi K, et al. Feasibility and dosimetry studies for 18F-NOS as a potential PET radiopharmaceutical for inducible nitric oxide synthase in humans. J Nucl Med 2012;53(6):994–1001.
  • 25. Nomura F, Yaguchi M, Togawa A, et al. Enhancement of poly-adenosine diphosphate-ribosylation in human hepatocellular carcinoma. J Gastroenterol Hepatol 2000;15(5):529–535.
  • 26. Ogino H, Nozaki T, Gunji A, et al. Loss of PARP-1 affects gene expression profile in a genome-wide manner in ES cells and liver cells. BMC Genomics 2007;8:41.
  • 27. Makvandi M, Xu K, Lieberman BP, et al. A radiotracer strategy to quantify PARP-1 expression in vivo provides a biomarker that can enable patient selection for PARP inhibitor therapy. Cancer Res 2016;76(15):4516–4524.
  • 28. Rosado MM, Bennici E, Novelli F, Pioli C. Beyond DNA repair, the immunological role of PARP-1 and its siblings. Immunology 2013;139(4):428–437.
  • 29. Hegedu˝s C, Robaszkiewicz A, Lakatos P, Szabó É, Virág L. Poly(ADP-ribose) in the bone: from oxidative stress signal to structural element. Free Radic Biol Med 2015;82:179–186.
  • 30. 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(3):244–250.
  • 31. Quiles-Perez R, Muñoz-Gámez JA, Ruiz-Extremera A, et al. Inhibition of poly adenosine diphosphate-ribose polymerase decreases hepatocellular carcinoma growth by modulation of tumor-related gene expression. Hepatology 2010;51(1):255–266.
  • 32. Ferraris DV. Evolution of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors: from concept to clinic. J Med Chem 2010;53(12):4561–4584.
  • 33. Ruf A, de Murcia G, Schulz GE. Inhibitor and NAD+ binding to poly(ADP-ribose) polymerase as derived from crystal structures and homology modeling. Biochemistry 1998;37(11):3893–3900.
  • 34. Gottipati P, Vischioni B, Schultz N, et al. Poly(ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res 2010;70(13):5389–5398.
  • 35. D’Amours D, Desnoyers S, D’Silva I, Poirier GG. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem J 1999;342(Pt 2):249–268.

Article History

Received August 16, 2016; revision requested September 13; revision received September 27; accepted October 4; final version accepted October 20.
Published online: Nov 14 2016
Published in print: Feb 2017