Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of the Poly(ADP-ribose) Polymerase (PARP) Inhibitor Veliparib (ABT-888) in Combination with Irinotecan in Patients with Advanced Solid Tumors

Purpose: PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor–mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. Experimental Design: Patients with advanced solid tumors were treated with 100 mg/m2 irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10–50 mg) occurred on days 3 to 14 (cycle 1) and days −1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). Results: Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m2 irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days −1–14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. Conclusions: Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227–37. ©2016 AACR.

[1]  L. Dawson,et al.  A final report of a phase I study of veliparib (ABT-888) in combination with low-dose fractionated whole abdominal radiation therapy (LDFWAR) in patients with advanced solid malignancies and peritoneal carcinomatosis with a dose escalation in ovarian and fallopian tube cancers. , 2016, Gynecologic oncology.

[2]  Ayala Hubert,et al.  Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[3]  M. Mulcahy,et al.  The Safety and Tolerability of Veliparib (V) plus Capecitabine (C) and Radiation (RT) in Subjects with Locally Advanced Rectal Cancer (LARC): Results of a Phase 1b Study , 2014 .

[4]  E. Winer,et al.  Phase I trial of olaparib in combination with cisplatin for the treatment of patients with advanced breast, ovarian and other solid tumors. , 2014, Annals of oncology : official journal of the European Society for Medical Oncology.

[5]  N. Curtin,et al.  Preferential potentiation of topoisomerase I poison cytotoxicity by PARP inhibition in S phase , 2014, British Journal of Cancer.

[6]  James H. Doroshow,et al.  Rationale for Poly(ADP-ribose) Polymerase (PARP) Inhibitors in Combination Therapy with Camptothecins or Temozolomide Based on PARP Trapping versus Catalytic Inhibition , 2014, The Journal of Pharmacology and Experimental Therapeutics.

[7]  R. Kurzrock,et al.  A phase I study of niraparib in combination with temozolomide (TMZ) in patients with advanced cancer. , 2014 .

[8]  L. Dawson,et al.  A Phase I Study of Veliparib (ABT-888) in Combination with Low-Dose Fractionated Whole Abdominal Radiation Therapy in Patients with Advanced Solid Malignancies and Peritoneal Carcinomatosis , 2014, Clinical Cancer Research.

[9]  V. Giranda,et al.  Targeting DNA repair with combination veliparib (ABT-888) and temozolomide in patients with metastatic castration-resistant prostate cancer , 2014, Investigational New Drugs.

[10]  Y. Pommier,et al.  PARP1–TDP1 coupling for the repair of topoisomerase I–induced DNA damage , 2014, Nucleic acids research.

[11]  G. Batist,et al.  Use of statins and the risk of death in patients with prostate cancer. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[12]  Y. Pommier,et al.  Stereospecific PARP Trapping by BMN 673 and Comparison with Olaparib and Rucaparib , 2013, Molecular Cancer Therapeutics.

[13]  Y. Pommier Drugging topoisomerases: lessons and challenges. , 2013, ACS chemical biology.

[14]  David Davidson,et al.  The PARP inhibitor ABT-888 synergizes irinotecan treatment of colon cancer cell lines , 2013, Investigational New Drugs.

[15]  Y. Pommier,et al.  Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. , 2012, Cancer research.

[16]  J. Doroshow,et al.  Advances in using PARP inhibitors to treat cancer , 2012, BMC Medicine.

[17]  Raquel Herrador,et al.  Topoisomerase I poisoning results in PARP-mediated replication fork reversal , 2012, Nature Structural &Molecular Biology.

[18]  Y. Pommier,et al.  Tyrosyl-DNA Phosphodiesterase 1 (TDP1) Repairs DNA Damage Induced by Topoisomerases I and II and Base Alkylation in Vertebrate Cells* , 2012, The Journal of Biological Chemistry.

[19]  Larry Rubinstein,et al.  A Phase I Study of Veliparib in Combination with Metronomic Cyclophosphamide in Adults with Refractory Solid Tumors and Lymphomas , 2012, Clinical Cancer Research.

[20]  K. Flatten,et al.  Enhanced Killing of Cancer Cells by Poly(ADP-ribose) Polymerase Inhibitors and Topoisomerase I Inhibitors Reflects Poisoning of Both Enzymes* , 2011, The Journal of Biological Chemistry.

[21]  Y. Pommier,et al.  Phase I study of PARP inhibitor ABT-888 in combination with topotecan in adults with refractory solid tumors and lymphomas. , 2011, Cancer research.

[22]  R. DiPaola,et al.  Phase I trial of veliparib, (ABT-888), a poly(ADP-ribose) polymerase (PARP) inhibitor, in combination with doxorubicin and cyclophosphamide in breast cancer and other solid tumors. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[23]  M. Knopp,et al.  A phase I dose-escalation study of ABT-888 (veliparib) in combination with carboplatin in HER2-negative metastatic breast cancer (MBC). , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[24]  J. Larkin,et al.  A phase I study of the safety and tolerability of olaparib (AZD2281, KU0059436) and dacarbazine in patients with advanced solid tumours , 2011, British Journal of Cancer.

[25]  Y. Pommier,et al.  Poly(ADP-ribose) polymerase and XPF–ERCC1 participate in distinct pathways for the repair of topoisomerase I-induced DNA damage in mammalian cells , 2011, Nucleic acids research.

[26]  Yvonne A. Evrard,et al.  Development of a Validated Immunofluorescence Assay for γH2AX as a Pharmacodynamic Marker of Topoisomerase I Inhibitor Activity , 2010, Clinical Cancer Research.

[27]  James H. Doroshow,et al.  Histone γH2AX and Poly(ADP-Ribose) as Clinical Pharmacodynamic Biomarkers , 2010, Clinical Cancer Research.

[28]  P. LoRusso,et al.  Poly(ADP-Ribose) Polymerase Inhibitors: A Novel Drug Class With a Promising Future , 2010, Cancer journal.

[29]  P. LoRusso,et al.  Simultaneous determination of ABT-888, a poly (ADP-ribose) polymerase inhibitor, and its metabolite in human plasma by liquid chromatography/tandem mass spectrometry. , 2010, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[30]  A. Ashworth,et al.  Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. , 2009, The New England journal of medicine.

[31]  Yin-Feng Xu,et al.  The canadian traveller problem and its competitive analysis , 2009, J. Comb. Optim..

[32]  Larry Rubinstein,et al.  Phase 0 clinical trial of the poly (ADP-ribose) polymerase inhibitor ABT-888 in patients with advanced malignancies. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[33]  Eric F. Johnson,et al.  Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer. , 2009, Journal of medicinal chemistry.

[34]  L. Rubinstein,et al.  Preclinical Modeling of a Phase 0 Clinical Trial: Qualification of a Pharmacodynamic Assay of Poly (ADP-Ribose) Polymerase in Tumor Biopsies of Mouse Xenografts , 2008, Clinical Cancer Research.

[35]  Allison L. Zulli,et al.  The selective poly(ADP-ribose) polymerase-1(2) inhibitor, CEP-8983, increases the sensitivity of chemoresistant tumor cells to temozolomide and irinotecan but does not potentiate myelotoxicity , 2007, Molecular Cancer Therapeutics.

[36]  Eric F. Johnson,et al.  ABT-888, an Orally Active Poly(ADP-Ribose) Polymerase Inhibitor that Potentiates DNA-Damaging Agents in Preclinical Tumor Models , 2007, Clinical Cancer Research.

[37]  E. Mazzon,et al.  Inhibition of poly(ADP‐ribose) polymerase prevents irinotecan‐induced intestinal damage and enhances irinotecan/temozolomide efficacy against colon carcinoma , 2006, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[38]  Y. Pommier,et al.  Repair of topoisomerase I-mediated DNA damage. , 2006, Progress in nucleic acid research and molecular biology.

[39]  N. Curtin,et al.  The Novel Poly(ADP-Ribose) Polymerase Inhibitor, AG14361, Sensitizes Cells to Topoisomerase I Poisons by Increasing the Persistence of DNA Strand Breaks , 2005, Clinical Cancer Research.

[40]  Soma Das,et al.  Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[41]  Robert Almassy,et al.  Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361. , 2004, Journal of the National Cancer Institute.

[42]  S. Culine,et al.  Sensitive HPLC-fluorescence method for irinotecan and four major metabolites in human plasma and saliva: application to pharmacokinetic studies. , 2003, Clinical chemistry.

[43]  R. Bonner,et al.  Histone H2AX phosphorylation is dispensable for the initial recognition of DNA breaks , 2003, Nature Cell Biology.

[44]  B. Ruggeri,et al.  Chemopotentiation of temozolomide, irinotecan, and cisplatin activity by CEP-6800, a poly(ADP-ribose) polymerase inhibitor. , 2003, Molecular cancer therapeutics.

[45]  E. Chu A Novel Methionine-Based Signaling Mechanism Regulating the Expression of Thymidylate Synthase , 2003, Cancer biology & therapy.

[46]  R. Schilsky,et al.  UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity , 2002, The Pharmacogenomics Journal.

[47]  J. Verweij,et al.  Pharmacokinetic, metabolic, and pharmacodynamic profiles in a dose-escalating study of irinotecan and cisplatin. , 2000, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.