Phase 1 study of ARQ 761, a β-lapachone analogue that promotes NQO1-mediated programmed cancer cell necrosis

David E Gerber, M Shaalan Beg, Farjana Fattah, Arthur E Frankel, Oluwatomilade Fatunde, Yull Arriaga, Jonathan E Dowell, Ajit Bisen, Richard D Leff, Claudia C Meek, William C Putnam, Raja Reddy Kallem, Indhumathy Subramaniyan, Ying Dong, Joyce Bolluyt, Venetia Sarode, Xin Luo, Yang Xie, Brian Schwartz, David A Boothman, David E Gerber, M Shaalan Beg, Farjana Fattah, Arthur E Frankel, Oluwatomilade Fatunde, Yull Arriaga, Jonathan E Dowell, Ajit Bisen, Richard D Leff, Claudia C Meek, William C Putnam, Raja Reddy Kallem, Indhumathy Subramaniyan, Ying Dong, Joyce Bolluyt, Venetia Sarode, Xin Luo, Yang Xie, Brian Schwartz, David A Boothman

Abstract

Background: NAD(P)H:quinone oxidoreductase 1 (NQO1) is a two-electron oxidoreductase expressed in multiple tumour types. ARQ 761 is a β-lapachone (β-lap) analogue that exploits the unique elevation of NQO1 found in solid tumours to cause tumour-specific cell death.

Methods: We performed a 3+3 dose escalation study of 3 schedules (weekly, every other week, 2/3 weeks) of ARQ 761 in patients with refractory advanced solid tumours. Tumour tissue was analysed for NQO1 expression. After 20 patients were analysed, enrolment was restricted to patients with NQO1-high tumours (H-score ≥ 200).

Results: A total of 42 patients were treated. Median number of prior lines of therapy was 4. Maximum tolerated dose was 390 mg/m2 as a 2-h infusion every other week. Dose-limiting toxicity was anaemia. The most common treatment-related adverse events were anaemia (79%), fatigue (45%), hypoxia (33%), nausea (17%), and vomiting (17%). Transient grade 3 hypoxia, reflecting possible methemoglobinaemia, occurred in 26% of patients. Among 32 evaluable patients, best response was stable disease (n = 12); 6 patients had tumour shrinkage. There was a trend towards improved efficacy in NQO1-high tumours (P = 0.06).

Conclusions: ARQ 761 has modest single-agent activity, which appears associated with tumour NQO1 expression. Principal toxicities include anaemia and possible methemoglobinaemia.

Trial registration: ClinicalTrials.gov NCT01502800.

Conflict of interest statement

D.E.G. has received research funding from ArQule, Inc. B.S. is an employee of ArQule, Inc. The other authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Efficacy of ARQ 761 according to tumour NQO1 expression. a Waterfall plot demonstrating best radiographic response in the overall study population. Dark bars indicate NQO1-high cases; light bars indicate NQO1-low cases. b Swimmer’s plot demonstrating time on therapy in the overall study population. Dark bars indicate NQO1-high cases; light bars indicate NQO1-low cases. Asterisks indicate cases for which treatment was discontinued for a reason other than disease progression. c Radiographic response among first 20 patients with both evaluable efficacy and tumour NQO1 expression data. NQO1 expression was higher among patients achieving stable disease compared to those with primary disease progression (P = 0.06). Of note, all six patients with NQO1-negative tumours had progressive disease. Based on these data, subsequent enrolment was limited to patients with NQO1-positive tumours (defined as H-score ≥ 200). PD progressive disease, RECIST, Response Evaluation Criteria in Solid Tumours, SD stable disease. d Progression-free survival according to tumour NQO1 expression. HR 0.68; 95% CI, 0.33–1.39; P = 0.3
Fig. 2
Fig. 2
Example of minor radiographic response from ARQ 761. Patient had heavily pre-treated bladder cancer (five prior lines of therapy). Images show multiple pulmonary metastases that decreased in size after initiation of study treatment
Fig. 3
Fig. 3
NQO1 IHC staining. a Moderately differentiated pancreatic adenocarcinoma, 3+ NQO1 staining, H-score 300. b Moderately differentiated colonic adenocarcinoma, 3+ NQO1 staining, H-score 250. c Lung adenocarcinoma, 3+ NQO1 staining, H-score 290. d High-grade invasive ductal breast carcinoma, 3+ NQO1 staining, H-score 300. e Prostate adenocarcinoma, 0+ NQO1 staining, H-score 0

References

    1. Pink JJ, et al. NAD(P)H:Quinone oxidoreductase activity is the principal determinant of beta-lapachone cytotoxicity. J. Biol. Chem. 2000;275:5416–5424. doi: 10.1074/jbc.275.8.5416.
    1. Bentle MS, Bey EA, Dong Y, Reinicke KE, Boothman DA. New tricks for old drugs: the anticarcinogenic potential of DNA repair inhibitors. J. Mol. Histol. 2006;37:203–218. doi: 10.1007/s10735-006-9043-8.
    1. Marin A, et al. DT-diaphorase and cytochrome B5 reductase in human lung and breast tumours. Br. J. Cancer. 1997;76:923–929. doi: 10.1038/bjc.1997.485.
    1. Belinsky M, Jaiswal AK. NAD(P)H:quinone oxidoreductase1 (DT-diaphorase) expression in normal and tumor tissues. Cancer Metastasis Rev. 1993;12:103–117. doi: 10.1007/BF00689804.
    1. Ough M, et al. Efficacy of beta-lapachone in pancreatic cancer treatment: exploiting the novel, therapeutic target NQO1. Cancer Biol. Ther. 2005;4:95–102. doi: 10.4161/cbt.4.1.1382.
    1. Dong Y, et al. Intratumoral delivery of beta-lapachone via polymer implants for prostate cancer therapy. Clin. Cancer Res. 2009;15:131–139. doi: 10.1158/1078-0432.CCR-08-1691.
    1. Bey EA, et al. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc. Natl. Acad. Sci. USA. 2007;104:11832–11837. doi: 10.1073/pnas.0702176104.
    1. Blanco E, et al. Beta-lapachone micellar nanotherapeutics for non-small cell lung cancer therapy. Cancer Res. 2010;70:3896–3904. doi: 10.1158/0008-5472.CAN-09-3995.
    1. Li CJ, Li YZ, Pinto AV, Pardee AB. Potent inhibition of tumor survival in vivo by beta-lapachone plus taxol: combining drugs imposes different artificial checkpoints. Proc. Natl. Acad. Sci. USA. 1999;96:13369–13374. doi: 10.1073/pnas.96.23.13369.
    1. Park HJ, et al. Susceptibility of cancer cells to beta-lapachone is enhanced by ionizing radiation. Int. J. Radiat. Oncol. Biol. Phys. 2005;61:212–219. doi: 10.1016/j.ijrobp.2004.09.018.
    1. Doll DC, Weiss RB, Issell BF. Mitomycin: ten years after approval for marketing. J. Clin. Oncol. 1985;3:276–286. doi: 10.1200/JCO.1985.3.2.276.
    1. Schellens JH, et al. Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09. J. Natl. Cancer Inst. 1994;86:906–912. doi: 10.1093/jnci/86.12.906.
    1. Khong HT, et al. A phase 2 study of ARQ 501 in combination with gemcitabine in adult patients with treatment naive, unresectable pancreatic adenocarcinoma. J. Clin. Oncol. 2007;25(18S (June 20 Supplement)):15017.
    1. Kawecki A, et al. A phase II study of ARQ 501 in patients with advanced squamous cell carcinoma of the head and neck. J. Clin. Oncol. 2007;25(18S (June 20 Supplement)):16509.
    1. Nasongkla N, et al. Enhancement of solubility and bioavailability of beta-lapachone using cyclodextrin inclusion complexes. Pharm. Res. 2003;20:1626–1633. doi: 10.1023/A:1026143519395.
    1. Blanco E, et al. Beta-lapachone-containing PEG-PLA polymer micelles as novel nanotherapeutics against NQO1-overexpressing tumor cells. J. Control. Release. 2007;122:365–374. doi: 10.1016/j.jconrel.2007.04.014.
    1. Li Y, Sun X, LaMont JT, Pardee AB, Li CJ. Selective killing of cancer cells by beta -lapachone: direct checkpoint activation as a strategy against cancer. Proc. Natl. Acad. Sci. USA. 2003;100:2674–2678. doi: 10.1073/pnas.0538044100.
    1. Phillips RM, Burger AM, Fiebig HH, Double JA. Genotyping of NAD(P)H:quinone oxidoreductase (NQO1) in a panel of human tumor xenografts: relationship between genotype status, NQO1 activity and the response of xenografts to Mitomycin C chemotherapy in vivo(1) Biochem. Pharmacol. 2001;62:1371–1377. doi: 10.1016/S0006-2952(01)00769-9.
    1. Kim S, et al. Pharmacokinetics and tolerability of MB12066, a beta-lapachone derivative targeting NAD(P)H: quinone oxidoreductase 1: two independent, double-blind, placebo-controlled, combined single and multiple ascending dose first-in-human clinical trials. Drug Des. Dev. Ther. 2017;11:3187–3195. doi: 10.2147/DDDT.S151269.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Fraley C, Raftery AE. Model-based clustering, discriminant analysis, and desnity estimation. J. Am. Stat. Assoc. 2002;97:611–631. doi: 10.1198/016214502760047131.
    1. Borinstein SC, Xu M, Hawkins DS. Methemoglobinemia and hemolytic anemia caused by rasburicase administration in a newly diagnosed child with Burkitt lymphoma/leukemia. Pediatr. Blood Cancer. 2008;50:189. doi: 10.1002/pbc.21193.
    1. Cortazzo JA, Lichtman AD. Methemoglobinemia: a review and recommendations for management. J. Cardiothorac. Vasc. Anesth. 2014;28:1043–1047. doi: 10.1053/j.jvca.2013.02.005.
    1. Huang X, et al. An NQO1 substrate with potent antitumor activity that selectively kills by PARP1-induced programmed necrosis. Cancer Res. 2012;72:3038–3047. doi: 10.1158/0008-5472.CAN-11-3135.
    1. Chakrabarti G, et al. Tumor-selective use of DNA base excision repair inhibition in pancreatic cancer using the NQO1 bioactivatable drug, beta-lapachone. Sci. Rep. 2015;5:17066. doi: 10.1038/srep17066.
    1. Ma X, et al. Esterase-activatable beta-lapachone prodrug micelles for NQO1-targeted lung cancer therapy. J. Control. Release. 2015;200:201–211. doi: 10.1016/j.jconrel.2014.12.027.
    1. Moore Z, et al. NAMPT inhibition sensitizes pancreatic adenocarcinoma cells to tumor-selective, PAR-independent metabolic catastrophe and cell death induced by beta-lapachone. Cell Death Dis. 2015;6:e1599. doi: 10.1038/cddis.2014.564.
    1. Chakrabarti G, et al. Targeting glutamine metabolism sensitizes pancreatic cancer to PARP-driven metabolic catastrophe induced by ss-lapachone. Cancer Metab. 2015;3:12. doi: 10.1186/s40170-015-0137-1.
    1. Madajewski B, Boatman MA, Chakrabarti G, Boothman DA, Bey EA. Depleting tumor-NQO1 potentiates anoikis and inhibits growth of NSCLC. Mol. Cancer Res. 2016;14:14–25. doi: 10.1158/1541-7786.MCR-15-0207-T.
    1. Silvers MA, et al. The NQO1 bioactivatable drug, beta-lapachone, alters the redox state of NQO1+ pancreatic cancer cells, causing perturbation in central carbon metabolism. J. Biol. Chem. 2017;292:18203–18216. doi: 10.1074/jbc.M117.813923.
    1. Kepa JK, Ross D. DT-diaphorase activity in NSCLC and SCLC cell lines: a role for fos/jun regulation. Br. J. Cancer. 1999;79:1679–1684. doi: 10.1038/sj.bjc.6690268.
    1. Kepa JK, Ross D. Differential expression of the antioxidant response element within the hNQO1 promoter in NSCLC versus SCLC. Biochem. Biophys. Res. Commun. 2003;311:446–453. doi: 10.1016/j.bbrc.2003.10.021.
    1. Gaedigk A, et al. NAD(P)H:quinone oxidoreductase: polymorphisms and allele frequencies in Caucasian, Chinese and Canadian Native Indian and Inuit populations. Pharmacogenetics. 1998;8:305–313. doi: 10.1097/00008571-199808000-00004.
    1. Boothman DA, Meyers M, Fukunaga N, Lee SW. Isolation of X-ray-inducible transcripts from radioresistant human melanoma cells. Proc. Natl. Acad. Sci. USA. 1993;90:7200–7204. doi: 10.1073/pnas.90.15.7200.
    1. Valerio LG, Jr., Kepa JK, Pickwell GV, Quattrochi LC. Induction of human NAD(P)H:quinone oxidoreductase (NQO1) gene expression by the flavonol quercetin. Toxicol. Lett. 2001;119:49–57. doi: 10.1016/S0378-4274(00)00302-7.
    1. Bey EA, et al. Catalase abrogates beta-lapachone-induced PARP1 hyperactivation-directed programmed necrosis in NQO1-positive breast cancers. Mol. Cancer Ther. 2013;12:2110–2120. doi: 10.1158/1535-7163.MCT-12-0962.
    1. Huang X, et al. Leveraging an NQO1 bioactivatable drug for tumor-selective use of poly(ADP-ribose) polymerase inhibitors. Cancer Cell. 2016;30:940–952. doi: 10.1016/j.ccell.2016.11.006.
    1. Yang Y, et al. Clinical implications of high NQO1 expression in breast cancers. J. Exp. Clin. Cancer Res. 2014;33:14. doi: 10.1186/1756-9966-33-14.
    1. Li Z, et al. NQO1 protein expression predicts poor prognosis of non-small cell lung cancers. BMC Cancer. 2015;15:207. doi: 10.1186/s12885-015-1227-8.
    1. Cui X, et al. High expression of NQO1 is associated with poor prognosis in serous ovarian carcinoma. BMC Cancer. 2015;15:244. doi: 10.1186/s12885-015-1271-4.
    1. Lin L, et al. Prognostic implication of NQO1 overexpression in hepatocellular carcinoma. Hum. Pathol. 2017;69:31–37. doi: 10.1016/j.humpath.2017.09.002.
    1. Park MT, et al. beta-lapachone significantly increases the effect of ionizing radiation to cause mitochondrial apoptosis via JNK activation in cancer cells. PLoS ONE. 2011;6:e25976. doi: 10.1371/journal.pone.0025976.
    1. Zhang L, et al. beta-Lapachone and paclitaxel combination micelles with improved drug encapsulation and therapeutic synergy as novel nanotherapeutics for NQO1-targeted cancer therapy. Mol. Pharm. 2015;12:3999–4010. doi: 10.1021/acs.molpharmaceut.5b00448.

Source: PubMed

3
Suscribir