The Circuitous Path of PARP Inhibitor Development in Epithelial Ovarian Cancer

Publication
Article
OncologyONCOLOGY Vol 26 No 2
Volume 26
Issue 2

Ovarian cancers account for more than 50% of gynecologic cancer deaths. This is attributable to the late stage of the disease at diagnosis.

Ovarian cancers account for more than 50% of gynecologic cancer deaths. This is attributable to the late stage of the disease at diagnosis. Overall, 22,280 new cases of epithelial ovarian cancer and 15,500 deaths are estimated for 2012.[1] Although ovarian cancer is highly sensitive to initial platinum-based chemotherapy, with response rates of 70% to 90% in previously untreated patients, only 30% are cured by upfront surgery and chemotherapy.[2,3] The remaining cases will relapse, and cure after relapse is unlikely. Rational therapies directed toward specific molecular targets, and a better understanding of the mechanisms of action and resistance of novel agents, are urgently needed to improve survival of patients with recurrent ovarian carcinoma. Recently, PARP inhibitors, which impair the base excision DNA repair (BER) pathway, have been emerging as promising targeted therapies.

PARP Inhibitors: Preclinical Data and Rationale

Poly(ADP-ribose) polymerase (PARP) proteins are involved in DNA replication, transcriptional regulation, and DNA damage repair.[4-6] Inhibition of PARP converts common single-strand DNA breaks (SSB) into double-strand breaks (DSB) during DNA replication. In the absence of homologous recombination mechanisms (HR), inhibition of PARP results in poor repair of these lesions and apoptosis. Therefore, PARP inhibitors currently entering clinical trials have been of interest in combination with radiation and chemotherapy, and as single agents in cases of patients with deficient HR (for example, in the presence of BRCA mutations), especially in patients with breast and ovarian cancers. The concept of “synthetic lethality,” which exploits the vulnerability of cancer cells that have lost one mechanism of DNA repair (ie, deficiency in HR) by targeting a second pathway (ie, BER repair with PARP inhibitors), may be a particularly attractive therapeutic approach.

Xenograft models of other types of solid tumors (eg, cervix, colon, lung, glioblastoma, and squamous cell carcinoma of the head and neck) have shown that PARP inhibitors administered hours prior to radiation delayed tumor growth or reduced tumor volumes.[7-12] Additive or synergistic antitumor effects have been shown with cisplatin, doxorubicin, and topotecan.[13,14]

Beyond BRCA Mutation Carriers

Approximately 10% of all ovarian cancers are associated with deleterious BRCA1 or BRCA2 mutations, and up to 35% of sporadic serous ovarian cancers exhibit BRCA1 inactivation via epigenetic silencing.[15-17] Finally, other proteins involved in the homologous recombination pathway, such as ATM, MRE-11, EMSY, and the Fanconi anemia proteins are affected in 17% to 60% of patients.[18-20] Given these other mechanisms of BRCA dysfunction that profoundly sensitize cells to PARP inhibition, it may be rational to study patients with sporadic ovarian cancer displaying BRCAness, as well those with BRCA mutations. A molecular characterization study suggested that more than 50% of patients with high-grade EOC had loss of BRCA function, either by genetic or epigenetic events.[21] In addition, 60% of endometrioid ovarian tumors exhibited loss of heterozygosity at the PTEN locus.[22] PTEN-deficient tumors also have deficiency of homologous recombination repair, and preclinical data suggest profound sensitivity of PTEN-deficient tumor cells to PARP inhibitors.[23]

PARP Inhibitors: Clinical Trials

PARP inhibitors have demonstrated activity when used as single agents for patients with BRCA-associated cancers who have a deficient homologous recombination DNA repair pathway. Olaparib (AZD 2281), a PARP-1 inhibitor, has demonstrated promising results as a single agent in phase I and expansion trials.[24,25] The agent showed a high response rate in a phase II trial of 57 BRCA-positive patients. The drug was well tolerated, and the results confirmed phase I data.[26] Another phase II study of 90 patients reported a RR of 24% in high-grade serous (HGS) ovarian cancers, and a RR of 41% in BRCA-mutation carriers.[27] Lederman et al presented a randomized placebo-controlled trial of olaparib in 265 patients with platinum-sensitive relapsed ovarian cancer, and noted a statistically significant improvement in progression-free survival (PFS) in the maintenance setting.[28] Given that clinical trials of olaparib have shown no improvement in overall survival, however, the manufacturer, AstraZeneca, recently announced that there would be no further investigation of the agent in the maintenance setting for ovarian cancer. As Zorn discusses in her article, in another randomized phase II study of pegylated liposomal doxorubicin (PLD) vs olaparib there was no difference in PFS, but this may have been due to the improved sensitivity of BRCA-mutation carriers to PLD.[29]

Veliparib, a PARP 1 and 2 inhibitor, in a phase I study confirmed potentiation of topotecan's effects on DNA, with increased levels of γH2AX; however, it was noted to be too toxic and required dose de-escalation.[30] A phase I study of irinotecan and veliparib in solid tumors, including seven patients with ovarian cancer, demonstrated that the combination of irinotecan at 100 mg/m2 on days 1 and 8 with veliparib was associated with a clinical benefit.[31] Although olaparib and veliparib are the most widely studied in ovarian cancer, additional PARP inhibitors, including BSI-201, AG014699, CEP9722, MK4827, E7016, LT673, and others, are in development as noted in Zorn's review.

Difficulties in Development

Despite promising data on olaparib, it has been 3 years since the publication of the initial phase I data, and there has yet to be a large randomized phase III trial in ovarian cancer. A trial of this nature is required to obtain registration and FDA approval. As a research community, however, we have been sidetracked by the complexities of targeted therapy with PARP inhibitors. First, the notion that PARP inhibitors may potentiate chemotherapy and radiation therapy has led to combination studies, instead of a focus on single-agent trials. Second, trials have expanded patient cohorts to include HGS tumors without an approved, confirmed biomaker test to predict response to PARP inhibition. Both of these are important areas of study, but it is essential to conduct a large phase III trial in ovarian cancer patients with BRCA mutations, to definitively establish initial efficacy in the recurrent setting. This would enable many patients seeking PARP inhibitors for treatment of their ovarian cancer to gain improved access to this highly coveted class of drugs.

Further delays in drug development include the reformulation of olaparib, the most widely studied PARP inhibitor to date. Additionally, recent negative phase III data on iniparib (BSI-201) in breast cancer have decreased the enthusiasm for PARP inhibitors as a class of agents. However, it must be noted that iniparib may not be a true PARP inhibitor and does not inhibit PARP 1 and 2 like olaparib and veliparib.[32] It is hoped that, as a scientific community, we will overcome these roadblocks, learn from the complex issues encountered in their development, and regain focus to properly investigate this class of drugs to definitively establish their role in the treatment of ovarian cancer. It is a responsibility that we must take seriously, for the lives of our patients suffering from ovarian cancer are at stake. These issues are most pressing for ovarian cancer patients with BRCA mutations, a clinical setting in which the rationale for use of PARP inhibitors, and their single-agent activity, are compelling.

Financial Disclosure:The author has no significant financial interest or other relationship with the manufacturers of any products or providers of any service mentioned in this article.

References:

Financial Disclosure:The author has no significant financial interest or other relationship with the manufacturers of any products or providers of any service mentioned in this article.

References

1. American Cancer Society: Cancer Facts & Figures 2012. Atlanta, GA, 2012. Available at http://www.cancer.org/Research/CancerFactsFigures/index.

2. Covens A, Carey M, Bryson P, et al. Systematic review of first-line chemotherapy for newly diagnosed postoperative patients with stage II, III, or IV epithelial ovarian cancer. Gynecol Oncol. 2002;85:71-80.

3. Gadducci A, Sartori E, Maggino T, et al. Analysis of failures after negative second-look in patients with advanced ovarian cancer: an Italian multicenter study. Gynecol Oncol. 1998;68:150-5.

4. Powell C, Mikropoulos C, Kaye SB, et al. Pre-clinical and clinical evaluation of PARP inhibitors as tumour-specific radiosensitisers. Cancer Treat Rev. 2010;36:566-75.

5. D'Amours D, Desnoyers S, D'Silva I, et al. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem J. 1999;342:249-68.

6. Pleschke JM, Kleczkowska HE, Strohm M, et al. Poly(ADP-ribose) binds to specific domains in DNA damage checkpoint proteins. J Biol Chem. 2000;275:40974-80.

7. Kelland LR, Tonkin KS. The effect of 3-aminobenzamide in the radiation response of three human cervix carcinoma xenografts. Radiother Oncol. 1989;15:363-9.

8. Calabrese CR, Almassy R, Barton S, et al. Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361. J Natl Cancer Inst. 2004;96:56-67.

9. Albert JM, Cao C, Kim KW, et al. Inhibition of poly(ADP-ribose) polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models. Clin Cancer Res. 2007;13:3033-42.

10. Khan K, Araki K, Wang D, et al. Head and neck cancer radiosensitization by the novel poly(ADP-ribose) polymerase inhibitor GPI-15427. Head Neck. 2010;32:381-91.

11. Russo AL, Kwon HC, Burgan WE, et al. In vitro and in vivo radiosensitization of glioblastoma cells by the poly (ADP-ribose) polymerase inhibitor E7016. Clin Cancer Res. 2009;15:607-12.

12. Donawho CK, Luo Y, Luo Y, et al. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA damaging agents in preclinical tumor models. Clin Cancer Res. 2007;13:2728-37.

13. Penning TD, Zhu GD, Gandhi VB, 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. J Med Chem. 2009;52:514-23.

14. Muñoz-Gámez JA, Martín-Oliva D, Aguilar-Quesada R, et al. PARP inhibition sensitizes p53-deficient breast cancer cells to doxorubicin-induced apoptosis. Biochem J. 2005;386:119-25.

15. Pal T, Permuth-Wey J, Betts JA, et al. BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases. Cancer. 2005;104:2807-16.

16. Baldwin RL, Nemeth E, Tran H, et al. BRCA1promoter region hypermethylation in ovarian carcinoma: a population-based study. Cancer Res. 2000;60:5329-33.

17. Geisler JP, Hatterman-Zogg MA, Rathe JA, et al. Frequency of BRCA1 dysfunction in ovarian cancer. J Natl Cancer Inst. 2002;94:61-7.

18. Taniguchi T, Tischkowitz M, Ameziane N, et al. Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med. 2003;9:568-74.

19. Hughes-Davies L, Huntsman D, Ruas M, et al. EMSY links the BRCA2 pathway to sporadic breast and ovarian cancer. Cell. 2003;115:523-35.

20. Parikh RA, White JS, Huang X, et al. Loss of distal 11q is associated with DNA repair deficiency and reduced sensitivity to ionizing radiation in head and neck squamous cell carcinoma. Genes Chromosomes Cancer. 2007;46:761-75.

21. Press JZ, De Luca A, Boyd N, et al. Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities. BMC Cancer. 2008; 8:17.

22. Kolasa IK, Rembiszewska A, Janiec-Jankowska A, et al. PTEN mutation, expression and LOH at its locus in ovarian carcinomas. Relation to TP-53, K-RAS, and BRCA-1 mutations. Gynecol Oncol. 2006;103:692-7.

23. Mendes-Pereira AM, Martin SA, Brough R, et al. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med. 2009;1:315-22.

24. 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;361:123-34.

25. 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;28:2512-9.

26. Audeh MW, Carmichael J, Penson RT, et al. 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;376:245-51.

27. 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. 2011;12:852-61.

28. Ledermann JA, Harter P, Gourley C, et al. Phase II randomized placebo-controlled study of olaparib (AZD2281) in patients with platinum-sensitive relapsed serous ovarian cancer (PSR SOC). J Clin Oncol. 2011;29(suppl): abstract 5003.

29. Kaye S, Kaufman B, Lubinski J, et al. Phase II study of the oral PARP inhibitor olaparib (AZD2281) versus liposomal doxorubicin in ovarian cancer patients with BRCA1 and/or BRCA2 mutations. European Society for Medical Oncology (ESMO) Congress 2010. Oct 8-12, 2010. Milan, Italy. Abstract 3725.

30. Ji JJ, Kummar S, Chen AP, et al. Pharmacodynamic response in phase I combination study of ABT-888 and topotecan in adults with refractory solid tumors and lymphomas. J Clin Oncol. 2010;28(suppl 15s): abstract 2514.

31. Lorusso P, Ji JJ, Li J, et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the poly(ADP-ribose) polymerase (PARP) inhibitor veliparib (ABT-888; V) in combination with irinotecan (CPT-11; Ir) in patients (pts) with advanced solid tumors. J Clin Oncol. 2011;29(suppl): abstract 3000.

32. Ji J, Lee MP, Kadota M, et al. Pharmacodynamic and pathway analysis of three presumed inhibitors of poly(ADP-ribose) polymerase: ABT-888, AZD2281, and BSI201. American Association for Cancer Research 102nd Annual Meeting. Apr 2-6, 2011. Orlando, FL. Abstract 4527.

Related Videos
Brian Slomovitz, MD, MS, FACOG discusses the use of new antibody drug conjugates for treating patients with various gynecologic cancers.
Developing novel regimens may continue to improve survival outcomes of patients with advanced cervical cancer following the FDA approval of pembrolizumab and chemoradiation, says Jyoti S. Mayadev, MD.
Treatment with pembrolizumab plus chemoradiation appears to be well tolerated with no detriment to quality of life among those with advanced cervical cancer.
Jyoti S. Mayadev, MD, says that pembrolizumab in combination with chemoradiation will be seamlessly incorporated into her institution’s treatment of those with FIGO 2014 stage III to IVA cervical cancer following the regimen’s FDA approval.
Domenica Lorusso, MD, PhD, says that paying attention to the quality of chemoradiotherapy is imperative to feeling confident about the potential addition of pembrolizumab for locally advanced cervical cancer.
Guidelines from the Society of Gynecologic Oncology may help with managing the ongoing chemotherapy shortage in the treatment of patients with gynecologic cancers, according to Brian Slomovitz, MD, MS, FACOG.
Interim data reveal favorable responses in patients with low-grade serous ovarian cancer treated with avutometinib plus defactinib, according to Susana N. Banerjee, MD.
Brian Slomovitz, MD, MS, FACOG, notes that sometimes there is a need to substitute cisplatin for carboplatin, and vice versa, to best manage gynecologic cancers during the chemotherapy shortage.
Findings from the phase 3 MIRASOL trial support mirvetuximab soravtansine as a standard treatment option for platinum-resistant ovarian cancer, according to Ritu Salani, MD.
Trastuzumab deruxtecan appears to elicit ‘impressive’ responses among patients with HER2-positive gynecologic cancers regardless of immunohistochemistry in the phase 2 DESTINY-PanTumor02 trial.
Related Content