Pamiparib is a potent and selective PARP inhibitor with unique potential for the treatment of brain tumor

Yao Xiong, Yin Guo, Ye Liu, Hexiang Wang, Wenfeng Gong, Yong Liu, Xing Wang, Yajuan Gao, Fenglong Yu, Dan Su, Fan Wang, Yutong Zhu, Yuan Zhao, Yiyuan Wu, Zhen Qin, Xuebing Sun, Bo Ren, Bin Jiang, Wei Jin, Zhirong Shen, Zhiyu Tang, Xiaomin Song, Lai Wang, Xuesong Liu, Changyou Zhou, Beibei Jiang, Yao Xiong, Yin Guo, Ye Liu, Hexiang Wang, Wenfeng Gong, Yong Liu, Xing Wang, Yajuan Gao, Fenglong Yu, Dan Su, Fan Wang, Yutong Zhu, Yuan Zhao, Yiyuan Wu, Zhen Qin, Xuebing Sun, Bo Ren, Bin Jiang, Wei Jin, Zhirong Shen, Zhiyu Tang, Xiaomin Song, Lai Wang, Xuesong Liu, Changyou Zhou, Beibei Jiang

Abstract

Pamiparib, an investigational Poly (ADP-ribose) polymerase (PARP) inhibitor in clinical development, demonstrates excellent selectivity for both PARP1 and PARP2, and superb anti-proliferation activities in tumor cell lines with BRCA1/2 mutations or HR pathway deficiency (HRD). Pamiparib has good bioavailability and is 16-fold more potent than olaparib in an efficacy study using BRCA1 mutated MDA-MB-436 breast cancer xenograft model. Pamiparib also shows strong anti-tumor synergy with temozolomide (TMZ), a DNA alkylating agent used to treat brain tumors. Compared to other PARP inhibitors, pamiparib demonstrated improved penetration across the blood brain barrier (BBB) in mice. Oral administration of pamiparib at a dose as low as 3 mg/kg is sufficient to abrogate PARylation in brain tumor tissues. In SCLC-derived, TMZ-resistant H209 intracranial xenograft model, combination of pamiparib with TMZ overcomes its resistance and shows significant tumor inhibitory effects and prolonged life span. Our data suggests that combination of pamiparib with TMZ has unique potential for treatment of brain tumors. Currently, the combination therapy of pamiparib with TMZ is evaluated in clinical trial [NCT03150862].

Keywords: Brain penetration; PARP; Pamiparib; TMZ.

Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Fig. 1
Fig. 1
The structural of Pamiparib and its activity. (A) The chemical structure of pamiparib. (B) The inhibitory effect of pamiparib on PARP enzymes. (C) The efficacy of pamiparib against intracellular PAR levels in HeLa cells. HeLa cells were incubated with the indicated concentrations of pamiparib for 18 hrs, and then cells were exposed to hydrogen peroxide (H2O2) to induce PAR synthesis. PAR levels without H2O2 induction were set as 100%.
Fig. 2
Fig. 2
Pamiparib shows specific growth inhibitory effects in HR deficient and BRAC mutant cell lines. (A) The anti-proliferative activity of pamiparib in several tumor cell lines. IC50 values indicate that pamiparib potently inhibited the tumor cells with HR-deficiency or BRAC mutants. For all experiments, viability was derived after 7 days of continuous treatment with pamiparib.
Fig. 3
Fig. 3
In vivo efficacy, PK and PD of pamiparib in a human MDA-MB-436 breast cancer xenograft model. (A) In vivo efficacy of pamiparib and olaparib was head-to-head compared in a human MDA-MB-436 breast cancer xenograft model. Doses ranging from 1.6 mg/kg to 12.5 mg/kg of pamiparib and 25 mg/kg, 50 mg/kg olaparib (oral, BID for 28 days) were tested in this model. Pamiparib induced tumor regression on day 29 and resulted in 100% objective responses (PR plus CR). At day 89, two months after treatment was terminated, tumor relapse was observed only in the lowest dose group (1.6 mg/kg); (B) all treatments had no effect on mouse body weight. (C) Dose-dependence of tumor PARylation inhibition and pamiparib plasma/tumor concentrations in the same model. Pamiparib induced a dose-dependent inhibition of PAR levels by 4 h after single oral administration of 0.17–10.9 mg/kg, which was correlated well with its PK in tumors; (D) time-dependence of PAR inhibition in tumors and its correlation with plasma/tumor PK. A single dose of 5.45 mg/kg pamiparib induced a rapid and potent decrease in PAR levels. The PARylation inhibition was 98% at 0.5 h and remained high (≥80%) throughout the first 12 h; at 24 h post treatment, the inhibition remained at 53%. (E) The PARylation inhibited by pamiparib and olaparib were compared head-to-head. Compared to olaparib, pamiparib induced a more sustained PAR inhibition.
Fig. 4
Fig. 4
Pamiparib potentiates temozolomide in both naive and TMZ-resistant H209 cells and in a xenograft mouse model. (A and B) IC50 curves in H209 and H209 TMZ resistance (H209-TR) cell lines. H209-TR cell lines were generated from H209 tumors by treatment with multiple cycles of TMZ in vivo. The derived H209-TR cells remained sensitive to the combination of pamiparib and TMZ in vitro. (C) The combined activity of pamiparib and TMZ in the H209 SCLC xenograft model. TMZ as a single agent was quite effective in this H209 model. One cycle of treatment resulted in all animals becoming tumor-free. However, resistance occurred quickly during the second cycle. Pamiparib and TMZ together significantly prevented resistance, and tumors remained sensitive to the drug combination after three cycles of 5-day treatment. (D) Synergistic activity of pamiparib and TMZ in the H209-TR xenograft model. TMZ single treatment was not effective in this model. In contrast, pamiparib and TMZ combination treatment was efficacious during two cycles of treatment.
Fig. 5
Fig. 5
Brain penetration of pamiparib and its efficacy combined with TMZ in an intracranial H209 xenograft model. (A) Brain penetration of pamiparib in an intracranial H209 xenograft at 1.5 mg/kg p.o. (B) PD activity of pamiparib in the intracranial model, PARylation in brain/tumor was completely inhibited at 4 h after a single dose of 3 mpk p.o. (C and D) The synergistic activity of pamiparib and TMZ in H209 and H209-TR intracranial xenograft. TMZ and the combination treatment of TMZ and pamiparib showed no significant difference in survival in the H209 naïve model. However, the combination treatment of TMZ and pamiparib dramatically prolonged the survival of the H209-TR models. The survival time was defined as the time from the day of tumor cell inoculation to one day before animal death or euthanization. Kaplan–Meier curves were generated using mouse survival duration in days.

References

    1. Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Li X., Heyer W.D. Homologous recombination in DNA repair and DNA damage tolerance. Cell Res. 2008;18(1):99–113. doi: 10.1038/cr.2008.1.
    1. Narod S.A., Foulkes W.D. BRCA1 and BRCA2: 1994 and beyond. Nat Rev Cancer. 2004;4:665–676. doi: 10.1038/nrc1431.
    1. Rouleau M., Patel A., Hendzel M.J. PARP inhibition: PARP1 and beyond. Nat Rev Cancer. 2010;10(4):293–301. doi: 10.1038/nrc2812.
    1. Lord C.J., Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355(6330):1152–1158. doi: 10.1126/science.aam7344.
    1. Audeh M.W. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet. 2010;376(9737):245–251. doi: 10.1016/S0140-6736(10)60893-8.
    1. Fong P.C., Boss D.S., Yap T.A. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361(2):123–134. doi: 10.1056/NEJMoa0900212.
    1. Fong P.C., Yap T.A., Boss D.S. 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. doi: 10.1200/JCO.2009.26.9589.
    1. Gelmon K.A., Tischkowitz M., Mackay H. 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. doi: 10.1016/S1470-2045(11)70214-5.
    1. Bryant H.E., Schultz N., Thomas H.D. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434(7035):913–917. doi: 10.1038/nature03443.
    1. Farmer H., McCabe N., Lord C.J. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434(7035):917–921. doi: 10.1038/nature03445.
    1. Satoh M.S., Lindahl T. Role of poly(ADP-ribose) formation in DNA repair. Nature. 1992;356(6367):356–358. doi: 10.1038/356356a0.
    1. Litton J.K., Rugo H.S., Ettl J. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379(8):753–763. doi: 10.1056/NEJMoa1802905.
    1. Brown J.S., Kaye S.B., Yap T.A. PARP inhibitors: the race is on. Br J Cancer. 2016;114(7):713–715. doi: 10.1038/bjc.2016.67.
    1. Mateo J., Lord C.J., Serra V. A decade of clinical development of PARP inhibitors in perspective. Ann Oncol. 2019;30(9):1437–1447. doi: 10.1093/annonc/mdz192.
    1. Shapiro G, Kristeleit R, Middleton M, et al. Pharmacokinetics of orally administered rucaparib in patients with advanced solid tumors. Mol Cancer Ther, 2013;12(11 Suppl): Abstract nr A218. DOI: 10.1158/1535–7163.
    1. Michie CO, Sandhu SK, Schelman WR, et al. Final results of the phase I trial of niraparib (MK4827), a poly(ADP)ribose polymerase (PARP) inhibitor incorporating proof of concept biomarker studies and expansion cohorts involving BRCA1/2 mutation carriers, sporadic ovarian, and castration resistant prostate cancer (CRPC). J Clin Oncol, 2013;31(suppl; abstr 2513). DOI:10.1200/jco.2013.31.15_suppl.2513.
    1. Molla S., Hembram K.C., Chatterjee S. PARP inhibitor olaparib enhances the apoptotic potentiality of curcumin by increasing the DNA damage in oral cancer cells through inhibition of BER cascade. Pathol Oncol Res. 2019 doi: 10.1007/s12253-019-00768-0.
    1. Gerson S.L. Clinical relevance of MGMT in the treatment of cancer. J Clin Oncol. 2002;20(9):2388–2399. doi: 10.1200/JCO.2002.06.110.
    1. Esteller M., Herman J.G. Generating mutations but providing chemosensitivity: The role of O6-methylguanine DNA methyltransferase in human cancer. Oncogene. 2004;23(1):1–8. doi: 10.1038/sj.onc.1207316.
    1. Pietanza M.C., Waqar S.N., Krug L.M. Randomized, double-blind, phase II study of temozolomide in combination with either veliparib or placebo in patients with relapsed-sensitive or refractory small-cell lung cancer. J Clin Oncol. 2018;36(23):2386–2394. doi: 10.1200/JCO.2018.77.7672.
    1. Gupta S.K., Kizilbash S.H., Carlson B.L. Delineation of MGMT hypermethylation as a biomarker for veliparib-mediated temozolomide-sensitizing therapy of glioblastoma. J Natl Cancer Inst. 2016;108(5):1–10. doi: 10.1093/jnci/djv369.
    1. Barazzuol L., Jena R., Burnet N.G. Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma. Radiat Oncol. 2013;8:65. doi: 10.1186/1748-717X-8-65.
    1. de Gooijer M.C., Buil L.C.M., Çitirikkaya C.H. ABCB1 attenuates the brain penetration of the PARP inhibitor AZD2461. Mol Pharm. 2018;15(11):5236–5243. doi: 10.1021/acs.molpharmaceut.8b00742.
    1. Durmus S., Sparidans R.W., van Esch A. Breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (P-GP/ABCB1) restrict oral availability and brain accumulation of the PARP inhibitor rucaparib (AG-014699) Pharm Res. 2015;32(1):37–46. doi: 10.1007/s11095-014-1442-z.
    1. Kizilbash S.H., Gupta S.K., Chang K. Restricted delivery of talazoparib across the blood-brain barrier limits the sensitizing effects of PARP inhibition on Temozolomide therapy in glioblastoma. Mol. Cancer Ther. 2017;16:2735–2746. doi: 10.1158/1535-7163.
    1. Mendes-Pereira A.M., Martin S.A., Brough R. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med. 2009;1(6–7):315–322. doi: 10.1002/emmm.200900041.
    1. Stupp R., Mason W.P., van den Bent M.J. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. doi: 10.1056/NEJMoa043330.
    1. Pietanza M.C., Kadota K., Huberman K. Phase II trial of temozolomide in patients with relapsed sensitive or refractory small cell lung cancer, with assessment of methylguanine-DNA methyltransferase as a potential biomarker. Clin Cancer Res. 2012;18(4):1138–1145. doi: 10.1158/1078-0432.CCR-11-2059.
    1. Bansal M., Yang J., Karan C. A community computational challenge to predict the activity of pairs of compounds. Nat Biotechnol. 2014;32(12):1213–1222. doi: 10.1038/nbt.3052.
    1. Quan A.L., Videtic G.M.M., Suh J.H. Brain metastases in small cell lung cancer. Oncology (Williston Park) 2004;18(8):961–972.
    1. Walbert T., Gilbert M.R. The role of chemotherapy in the treatment of patients with brain metastases from solid tumors. Int J Clin Oncol. 2009;14(4):299–306. doi: 10.1007/s10147-009-0916-1.
    1. Kortmann U., McAlpine J.N., Xue H. Tumor growth inhibition by Olaparib in BRCA2 germline-mutated patient derived ovarian cancer tissue xenografts. Clin Cancer Res. 2010;4(17):783–791. doi: 10.1158/1078-0432.
    1. Sztupinszki Z., Diossy M., Krzystanek M. Detection of molecular signatures of homologous recombination deficiency in prostate cancer with or without BRCA1/2 mutations. Clin Cancer Res. 2020 doi: 10.1158/1078-0432.CCR-19-2135.
    1. Pommier Y., O’Connor M.J., de Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016;8(362):362ps17. doi: 10.1126/scitranslmed.aaf9246.
    1. Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol Oncol, 2011;594: 387–93. DOI: 10.1016/j.molonc.2011.07.001.
    1. O’Connor M.J. Targeting the DNA damage response in cancer. Mol Cell. 2015;60(4):547–560. doi: 10.1016/j.molcel.2015.10.040.
    1. Yung W.K., Albright R.E., Olson J. A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer. 2000;83(5):588–593. doi: 10.1054/bjoc.2000.1316.
    1. Abbott N.J., Patabendige A.A.K., Dolman D.E.M. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010;37(1):13–25. doi: 10.1016/j.nbd.2009.07.030.
    1. Wu X., Wang J., Zhou Q. Pamiparib, a novel PARP 1/2 inhibitor, monotherapy for gBRCAm patients with recurrent ovarian, fallopian, and primary peritoneal cancer: An open-label, multicenter, phase II trial in China. Ann Oncol. 2018;29(8):viii356. doi: 10.1093/annonc/mdy285.205.
    1. Henneman L., van Miltenburg M.H., Michalak E.M. Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer. Proc Natl Acad Sci U S A. 2015;112(27):8409–8414. doi: 10.1073/pnas.1500223112.
    1. Dufour R., Daumar P., Mounetou E. BCRP and P-gp relay overexpression in triple negative basal-like breast cancer cell line: a prospective role in resistance to Olaparib. Sci Rep. 2015;5:12670. doi: 10.1038/srep12670.
    1. Kubalanza K., Konecny G.E. Mechanisms of PARP inhibitor resistance in ovarian cancer. Curr Opin Obstet Gynecol. 2020;32(1):36–41. doi: 10.1097/GCO.0000000000000600.

Source: PubMed

3
Se inscrever