1. Academic Validation
  2. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models

ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models

  • Clin Cancer Res. 2007 May 1;13(9):2728-37. doi: 10.1158/1078-0432.CCR-06-3039.
Cherrie K Donawho 1 Yan Luo Yanping Luo Thomas D Penning Joy L Bauch Jennifer J Bouska Velitchka D Bontcheva-Diaz Bryan F Cox Theodore L DeWeese Larry E Dillehay Debra C Ferguson Nayereh S Ghoreishi-Haack David R Grimm Ran Guan Edward K Han Rhonda R Holley-Shanks Boris Hristov Kenneth B Idler Ken Jarvis Eric F Johnson Lawrence R Kleinberg Vered Klinghofer Loren M Lasko Xuesong Liu Kennan C Marsh Thomas P McGonigal Jonathan A Meulbroek Amanda M Olson Joann P Palma Luis E Rodriguez Yan Shi Jason A Stavropoulos Alan C Tsurutani Gui-Dong Zhu Saul H Rosenberg Vincent L Giranda David J Frost
Affiliations

Affiliation

  • 1 Abbott Laboratories, Abbott Park, Illinois 60064-6074, USA.
Abstract

Purpose: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888.

Experimental design: In vitro potency was determined in a PARP-1 and PARP-2 Enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation.

Results: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with K(i)s of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d x 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity.

Conclusions: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.

Figures
Products