<<

UC San Diego UC San Diego Previously Published Works

Title RRx-001, A novel dinitroazetidine radiosensitizer.

Permalink https://escholarship.org/uc/item/9ng8s05d

Journal Investigational new drugs, 34(3)

ISSN 0167-6997

Authors Oronsky, Bryan Scicinski, Jan Ning, Shoucheng et al.

Publication Date 2016-06-01

DOI 10.1007/s10637-016-0326-y

Peer reviewed

eScholarship.org Powered by the California Digital Library University of California Invest New Drugs (2016) 34:371–377 DOI 10.1007/s10637-016-0326-y

REVIEW

RRx-001, A novel dinitroazetidine radiosensitizer

Bryan Oronsky1 & Jan Scicinski1 & Shoucheng Ning2 & Donna Peehl2 & Arnold Oronsky3 & Pedro Cabrales4 & Mark Bednarski2 & Susan Knox2

Received: 26 October 2015 /Accepted: 26 January 2016 /Published online: 3 February 2016 # The Author(s) 2016. This article is published with open access at Springerlink.com

Summary The ‘holy grail’ in radiation oncology is to improve Introduction the outcome of radiation therapy (RT) with a radiosensitizer—a systemic chemical/biochemical agent that additively or syner- The origin story of the Phase II anticancer agent RRx-001 is gistically sensitizes tumor cells to radiation in the absence of presented in this review. significant toxicity. Similar to the oxygen effect, in which DNA Unlike the pharmaceutical industry’s emphasis on Bme bases modified by reactive oxygen species prevent repair of the too^ drugs that slavishly mimic existing chemical compounds cellular radiation damage, these compounds in general magnify (e.g., statins, antibiotics, H1 and H2 histamine blockers etc.), free radical formation, leading to the permanent Bfixation^ of RRx-001, having been derived from the aerospace and the resultant chemical change in the DNA structure. The defense sector, is the prototype of a pharmacologically purpose of this review is to present the origin story of the unprecedented and decidedly Bnon-me too^ chemical class radiosensitizer, RRx-001, which emerged from the aerospace called dinitroazetidines. industry. The activity of RRx-001 as a chemosensitizer in The strategic decision to move forward a compound hinges multiple tumor types and disease states including malaria, on a risk benefit analysis. In this case the development of hemorrhagic shock and sickle cell anemia, are the subject RRx-001 was predicated on the reasonable anticipation of of future reviews. non-toxicity, even though the benefits initially were unknown. Given that the detonation of nitrogenous combustibles in the atmosphere is a potential threat to the health of humans, live- Keywords Radiosensitizer . Radiosensitization . stock, wildlife, and ecosystems, military agencies in the US Chemosensitizer . Cancer . Clinical trials . RRx-001 have conducted risk assessments [1]; in particular, the safety profile of TNAZ [2], structurally similar to RRx-001 had al- ready been comprehensively characterized, suggesting that the development of dinitroazetidine containing compounds were inherently less risky. The availability of toxicology in- * Bryan Oronsky formation was a significant advantage since GLP-repeated [email protected] dose toxicology studies cost millions of US dollars [3]and take up to or over a year to complete. In addition, since the chemistry of energetic com- 1 EpicentRx Inc, 800 W El Camino Real, Suite 180, pounds is based on free radical-initiated chain reactions, Mountain View, CA 94040, USA synergy with radiation therapy was suggested, given that 2 Stanford University, 875 Blake Wilbur Dr Clinic D, the outcome of the latter depends on the generation of Stanford, CA 94305, USA reactive oxygen species [4]. Free radical production was also 3 InterWest Partners, 2710 Sand Hill Rd #200, Menlo Park, expected because 1,3,3 trinitroazetidine (TNAZ) [5], an ex- CA 94025, USA plosive propellant for guns, artillery, mortars and rockets, and 4 Department of Bioengineering, University of California San Diego the closest chemical analog of RRx-001, yielded free radicals (UCSD), 9500 Gilman Dr, La Jolla, CA 92093, USA during bond cleavage [6]. 372 Invest New Drugs (2016) 34:371–377

The only examples of pharmaceutical agents with compa- rable origins to RRx-001 are the antituberculant, iproniazid, and the antidepressant imipramine [7], derived from leftover World War II rocket fuel hydralazine as well as the explosive nitroglycerin (NTG), introduced as a treatment for angina pectoris [8] several years before Alfred Nobel, the inventor of dynamite, developed the condition [9]. TNAZ modified with the removal of a single nitro group (NO2) and substitu- tion of a bromoacetate group produced a non-explosive deriv- ative called ABDNAZ, an acronym for 1-bromoacetyl-3,3- dinitroazetidine, a name later shortened to RRx-001 for Fig. 2 A comparison of the single dose antitumor activity of RRx-001 easier-to-use pronunciation and communication. In vivo (ABDNAZ) to in murine SCCVII tumor model RRx-001 demonstrated single-agent activity as well as hyp- oxic cell radiosensitization [10]. As a common feature of most solid tumors, hypoxia, there- fore, plays a critical role not only in the development of Effects of hypoxia on radiosensitivity radioresistance but also chemoresistance. Unlike tumors, and with the exception of tissues like the retina and the dermis,

When solid tumor growth exceeds a critical diameter of 1– normal cells are normally well-oxygenated (>10 mm/Hg O2) 2mm3 (106 cells) [11], diffusion limitations of oxygen and [17]. A clear therapeutic disadvantage, hypoxia is also poten- nutrients from blood vessels located in the periphery leads to tially an exploitable physiological difference, opening the necrotic centers. The resultant activation of the hypoxia induc- door to the development of hypoxia-selective agents that are ible factor (HIF) system mediates the expression of VEGF, preferentially toxic only to oxygen-deficient tumor cells. In erythropoietin and factors regulating glucose transport and particular, the development of the nitroimidazoles as hypoxic glycolysis such as GLUT-1 and GLUT-3, [12]; the induction cell sensitizers that mimic the effect of oxygen on tumors of these genes drives vascular remodeling and a metabolic resulted from the discovery that 14C-labelled misonidazole switch to aerobic glycolysis, which are integral to malignant bound selectively to macromolecules in hypoxic cells both transformation and progression. [13, 14] Due to a dearth of in vitro and in vivo [18] and were reduced by nitroreductase superoxide, hydrogen peroxide and hydroxyl radicals that ox- enzymes to a radical anion—this reduction only occurs under idatively damage macromolecules including lipid, protein and hypoxic conditions [19]. nucleic acid under low oxygen conditions, the presence of hypoxia predicts for a poor response to radiotherapy.[15]For example, nearly 40 % of breast cancers have hypoxic regions Radiosensitizers defined with oxygen concentrations below the threshold required for half-maximal radiosensitivity (pO2 < 2.5 mmHg), which ad- The term Bradiosensitizer^ refers to an agent that enhances the versely impacts the response to radiotherapy [16]. therapeutic ratio of radiotherapy for similar levels of normal

Fig. 1 The in vitro activity of RRx-001 (ABDNAZ) under normoxia or hypoxia in SCC VII tumor Invest New Drugs (2016) 34:371–377 373

Fig. 3 RRx-001 (ABDNAZ) in human colon cancer HT29 and murine carcinoma SCC VII cell lines: radiosensitization effects tissue toxicity, which is tantamount to the Holy Grail in radi- 3) Sequence-dependent synergy with radiation with no over- ation oncology and cancer therapy in general because selec- lapping toxicity tive cytotoxicity predicts improved patient tolerance and over- 4) Activation under hypoxia all quality of life. Like the reaction of oxygen, which leads to 5) Broad therapeutic index the formation of DNA hydroxyl and peroxyl radicals that di- 6) Normal tissue radioprotection and tumor radiosensitization rectly attack DNA, radiosensitizers increase the pool of oxi- dizing species, resulting in enhanced Bfixation^ of free-radical Given its novel redox-based mechanism of radiosensitization, DNA damage [20]. favorable toxicity profile, and inherent cytotoxicity, RRx-001 Unfortunately, however, the history of radiosensitization is fits the definition of a promising carcinoma radiosensitizer, associated with the limited clinical efficacy and substantial nor- based on the criteria listed above. mal tissue toxicity observed with potential radiosensitizers in- cluding the halogenated pyrimidines [21] and other antimetab- olites, cisplatin and 5- (5-FU), the nitroimidazoles, RRx-001 radiosensitization properties and the hypoxic cytotoxins such as tirapazamine and the mitomycin-related quinones EO9 and porfiromycin [22, 23]. Inherently selective cytotoxicity The lessons learned from the failure of these compounds is that a radiosensitizer should ideally possess or exhibit: RRx-001 is an optimized derivative of TNAZ, a compound chosen from a collection of energetic polynitro propellant ma- 1) Systemic single agent activity terials on the basis of a greater increase in IC50 hypoxia com- 2) Tumor specificity pared to normoxia (Fig. 1)[10].

Fig. 4 RRx-001 (ABDNAZ) in murine SCCVII tumor model: potentiation of the effect of radiation. RRx-001 (ABDNAZ) was given at a dose of 5 mg/ kg QD for 5 days. Radiotherapy: 250 cGy QD for 5 days 374 Invest New Drugs (2016) 34:371–377

and synergism revealed a synergistic interaction between RRx-001 and radiation. Potentiation of radiation-induced growth delay in murine tumors was both dose and schedule dependent. Maximum tumor growth delay occurred when RRx-001 was adminis- tered minutes prior to or concomitant with (during) radiation (Fig. 5). The mechanistic basis of radiosensitization is mediated by an intricate interaction of RRx-001-modified RBCs (on administration RRx-001 penetrates the red blood cell Fig. 5 Tumor growth delay (TGD) and time between RRx-001 membrane and binds irreversibly to a particular residue (ABDNAZ) dosage and radiation in murine SCCVII tumor model. hemoglobin, beta Cysteine 93) with the tumor vasculature. p = 0.05 RRx-001 (ABDNAZ) t = 0 and t =24h;p =0.09RRx-001 [25, 26] The preferential adhesion of RRx-001 RBCs to the t t (ABDNAZ) = 0 and =2h vascular endothelium is followed by tumor internalization and catabolization in a Trojan Horse manner, releasing redox active In vivo activity of RRx-001 in the SCCVII syngeneic RRx-001 and RBC metabolites (i.e. nitric oxide [26, 27] iron mouse tumor model demonstrated equivalent activity to and heme) EpicentRx unpublished data. Cisplatin, with no apparent induced side effects, as shown The beneficial pleiotropic effects of this oxidative damage below (Fig. 2), indicative of promising antitumor activity include: nitric oxide generation, [28] increased tumor per- and a favorably low acute toxicity profile [10]. fusion, arrest, apoptosis and inhibition of cell division, inhibition of epigenetic enzymes responsible for DNA methylation and various histone modifications [29]and Synergistic effects with radiation effects on DNA damage and repair pathways. This underlying mechanism of action is suggested by an expanding body of In vitro experiments showed that RRx-001 synergistically preclinical evidence: 1) in a dose dependent manner, RRx-001 enhances XRT-induced inhibition of proliferation of both enhanced radiation-induced pro-oxidant production (Fig. 6). 2) radiosensitive SCCVII cells and relatively radioresistant RRx-001 significantly improved tumor blood flow/perfusion HT-29 tumor cells (Fig. 3), potentiates the survival of from baseline values compared to control in a murine SCCVII SCCVII tumor-bearing mice, and significantly improves tumor model. The enhanced blood flow and, by extension, the therapeutic ratio of radiotherapy (Fig. 4). Analysis with oxygenation may be, at least, in part, related to the over- Jin’sformula(Q = Ea + b/(Ea + Eb − Ea × Eb)[24]ofbothin production of nitric oxide (NO) via RRx-001-modified vitro and in vivo experiments for antagonism, additive effects, deoxyhemoglobin under hypoxic conditions that are specific

Fig. 6 a Generation of ROS over time in HT29 tumor cells by RRx- 001 (ABDNAZ) with and without radiation. b Generation of ROS in SCVII tumor cells by RRx-001 (ABDNAZ) with and without radiation Invest New Drugs (2016) 34:371–377 375 to cancer cells (Fig. 7). Exposure of HT-29 cells RRx-001 results in the formation of a dose-dependent increase in DNA double strand breaks assessed by the measurement of gamma-H2AX, a biomarker of DNA damage (Fig. 8)[10].

Conclusion and future directions

Despite the demonstration of activity as a chemosensitizer, Fig. 8 RRx-001 (ABDNAZ) induced DNA damage measured by degree chemo-resensitizer [29, 30] and immunosensitizer in multiple of induction of γH2AX positive cells tumor types, RRx-001 has continued along the development path of radiosensitization: currently two Phase I/II clinical trials are underway in brain metastases with whole brain ra- previously failed radiosensitizers such as misonidazole, diotherapy (WBRT) and in primary GBM with radiation and motexafin gadolinium (Xcytrin), Efaproxyn (efaproxiral or . The emerging data is highly positive, albeit RSR-13) and bortozemib (Velcade), tirapazamine, RSR-13, limited and preliminary. These caveats notwithstanding, the eniposide, , , cisplatin and IUDR, activity profile of RRx-001 + radiation therapy suggests syn- tumor-targeted radiosensitization remains an attractive, if ergistic cancer cell cytotoxicity in the absence of any neuro- utopian, strategy to improve local control or cure rates. logical toxicity, which would likely support multicenter Phase However, the feasibility of radiosensitization as a therapeu- III clinical trials with concurrent radiation and chemoradiation tic strategy ultimately depends on the optimization of the del- in these indications. icate balance between efficacy and normal tissue toxicity. The The percentage of cancer patients that will receive radiation use of the quinone , for example, despite a pref- therapy (RT) at some time during their course of their disease erential toxicity for hypoxic tumor cells, is limited due to is approximately 50–60 %.[31] A well-characterized dose- cumulative myelosuppression. [33] Likewise, tirapazamine, response relationship between malignant and normal tissue the first hypoxic cytotoxin to enter clinical trials [34], may has been described: higher exposures of radiation lead to prematurely ‘preactivate’ in tissues with a modest degree of better responses; at sufficiently high doses RT sterilizes hypoxia such as the retina and dermis (1.5 % oxygen) before even Bradioresistant^ tumors. Likewise for normal tissues, reaching the true hypoxic core of the tumor (0.5 % oxygen), where higher doses lead to greater damage, [32], treatment leading to relatively poor selectivity for neoplastic cells and a related toxicity is a major cause for the failure of radio- narrow therapeutic index. therapy. A potential solution to this insuperable problem is Given its solid-tumor efficacy profile, minimal toxicity, radiosensitization; therefore, despite the laundry list of hypoxic cell preference, unique mechanism of action and

Fig. 7 RRx-001 (ABDNAZ) causes and increase in blood perfusion and blood volume in murine SCCVII tumor model. The slope represents relative rate of tumor blood perfusion and the level of the plateau represents relative blood volume 376 Invest New Drugs (2016) 34:371–377 synergy with radiation, RRx-001 has the potential to fill this 12. Bel Aiba RS, Dimova EY,Görlach A, Kietzmann T (2006) The role – treatment vacuum in the therapeutic arsenal as a radiosensitizer. of hypoxia inducible factor-1 in cell metabolism a possible target in cancer therapy. Expert Opin Ther Targets 10(4):583–599 13. Boyle RG, Travers S (2006) Hypoxia: targeting the tumour. Anti Cancer Agents Med Chem 6(4):281–286 Acknowledgments The authors gratefully acknowledge the contribu- tions of Robert B. Wardle and Louis F. Cannizzo to the discovery and 14. Maxwell PH (2005) The HIF pathway in cancer. Semin Cell Dev – – development of ABDNAZ. Biol 16(4 5):523 530 15. Williams KJ, Cowen RL, Stratford IJ (2001) Hypoxia and oxidative stress. tumour hypoxia–therapeutic considerations. Breast Cancer – Compliance with ethical standards Res 3(5):328 331 16. Vaupel P, Schlenger K, Knoop C, Höckel M (1991) Oxygenation of Disclosure statement for conflicts Of interest The authors declare that human tumors: evaluation of tissue oxygen distribution in breast EpicentRx funds research of RRx-001. Authors Bryan. cancers by computerized O2 tension measurements. Cancer Res Oronsky and Jan. Scicinski are employees at EpicentRx. 51(12):3316–3322 17. Hockel M, Vaupel P (2001) Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93(4): Open Access This article is distributed under the terms of the Creative 266–276 Commons Attribution 4.0 International License (http:// 18. Varghese AJ, Gulyas S, Mohindra JK (1976) Hypoxia-dependent creativecommons.org/licenses/by/4.0/), which permits unrestricted use, reduction of 1-(2-nitro-1-imidazolyl)-3-methoxy-2-propanol by distribution, and reproduction in any medium, provided you give appro- Chinese hamster ovary cells and KHT tumor cells in vitro and in priate credit to the original author(s) and the source, provide a link to the vivo. Cancer Res 36(10):3761–3765 Creative Commons license, and indicate if changes were made. 19. Chacon E, Morrow CJ, Leon AA, Born JL, Smith BR (1988) Regioselective formation of a misonidazole-glutathione conjugate as a function of pH during chemical reduction. Biochem Pharmacol 37(2):361–363 References 20. Chapman JD, Reuvers AP, Borsa J, Greenstock CL (2012) Chemical radioprotection and radiosensitization of mammalian cells growing in Vitro. Radiat Res 178(2):214–222 1. Pichtel J (2012) Distribution and fate of military explosives and 21. Russell KJ, Rice GC, Brown JM (1986) In vitro and in vivo radi- propellants in soil: a review. Applied and Environmental Soil ation sensitization by the halogenated pyrimidine 5-chloro-2'- Science 2012 Article ID 617236:33 deoxycytidine. Cancer Res 46(6):2883–2887 2. Prezioso JA, Desai L, Fitzgerald G, Paika IJ, Digiulio N, Dodd D 22. Belcourt MF, Hodnick WF, Rockwell S, Sartorelli AC (1996) (1994) Genetic Toxicity Evaluation of 1,3,3-Trinitroazetidine. Volume Differential toxicity of mitomycin C and porfiromycin to aerobic 4. Summary report on the Genotoxicity of TNAZ. TOXIKON CORP, and hypoxic Chinese hamster ovary cells overexpressing human WOBURN NADPH:cytochrome c (P-450) reductase. Proc Natl Acad Sci U S 3. Chong CR, Sullivan DJ Jr (2007) New uses for old drugs. Nature A 93(1):456–460 8(448):645–646 23. Oronsky BT, Knox SJ, Scicinski J (2011) Six degrees of separation: 4. Azzam EI, Jay-Gerin JP, Pain D (2012) Ionizing radiation-induced the oxygen effect in the development of radiosensitizers. Transl metabolic oxidative stress and prolonged cell injury. Cancer Lett Oncol 4(4):189–198 327(1–2):48–60 24. Jin ZJ (1980) Addition in drug combination (author's transl). 5. Sikder N, Sikder AK, Bulakh NR, Gandhe BR (2004) 1,3,3- Zhongguo Yao Li Xue Bao 1(2):70–76 trinitroazetidine (TNAZ), a melt-cast explosive: synthesis, charac- 25. Scicinski J, Oronsky B, Taylor M, Luo G, Musick T, Marini J, terization and thermal behaviour. J Hazard Mater 113(1–3):35–43 Adams CM, Fitch WL (2012) Preclinical evaluation of the metab- 6. Fried L, Manaa M, Pagoria P, Simpson R (2001) Design and syn- olism and disposition of RRx-001, a novel investigative anticancer thesis of energetic materials. Annu Rev Mater Res 31:291–321 agent. Drug Metab Dispos 40(9):1810–1816 7. Lopez-Munoz F, Alamo C (2009) Monoaminergic neurotransmis- 26. Scicinski J, Oronsky B, Ning S, Knox S, Peehl D, Kim MM, sion: the history of the discovery of antidepressants from 1950s Langecker P, Fanger G (2015) NO to cancer: the complex and until today. Curr Pharm Des 15(14):1563–1586 multifaceted role of nitric oxide and the epigenetic nitric oxide 8. Ignarro LJ (2002) After 130 years, the molecular mechanism of donor, RRx-001. Redox Biol 6:1–8 action of nitroglycerin is revealed. Proc Natl Acad Sci U S A 27. Oronsky BT, Knox SJ, Scicinski JJ (2012) Is nitric oxide (NO) the 99(12):7816–7817 last word in radiosensitization? A review. Transl Oncol 5(2):66–71 9. Marsh N, Marsh A (2000) A short history of nitroglycerine and nitric 28. Sharma Das D, Tian Z, Ray A, Ravillah D, Song Y, Richardson P, oxide in pharmacology and physiology. Clin Exp Pharmacol Physiol Oronsky B, Scicinski J, Chauhan D, Anderson K (2014) Anti- 27(4):313–319 myeloma activity of a novel free radical inducer rrx-001. Blood 10. Ning S, Bednarski M, Oronsky B, Scicinski J, Saul G, Knox SJ 124(21):4712 (2012) Dinitroazetidines are a novel class of anticancer agents and 29. Oronsky BT, Oronsky AL, Lybeck M, Oronsky NC, Scicinski JJ, hypoxia-activated radiation sensitizers developed from highly ener- Carter C, Day RM, Rodriguez Orengo JF, Rodriguez-Torres M, getic materials. Cancer Res 72(10):2600–2608 Fanger GF, Reid TR (2015) Episensitization: defying Time's Arrow. 11. Baronzio G, Gramaglia A, Baronzio A, Freitas I (2000) Influence of Front Oncol 5:134 Tumor Microenvironment on Thermoresponse: Biologic and 30. Reid T, Dad S, Korn R, Oronsky B, Knox S, Scicinski J (2014) Two Clinical Implications, In: Madame Curie Bioscience Database case reports of resensitization to previous with [Internet]. Landes Bioscience, Austin the novel hypoxia-activated hypomethylating anticancer agent Invest New Drugs (2016) 34:371–377 377

RRx-001 in metastatic colorectal cancer patients. Case Rep 33. Seiwert TY, Salama JK, Vokes EE (2007) The chemoradia- Oncol 7(1):79–85 tion paradigm in head and neck cancer. Nat Clin Pract Oncol 31. Halperin BL, Perez C (2008) Perez and Brady's: Principles and 4(3):156–171 Practice of Radiation Oncology. Lippincott Williams and Wilkins, 34. Zeman EM, Brown JM, Lemmon MJ, Hirst VK, Lee WW (1986) Philadelphia SR-4233: a new bioreductive agent with high selective toxicity for 32. Denardo SJ, Williams LE, Leigh BR, Wahl RL (2002) Choosing an hypoxic mammalian cells. Int J Radiat Oncol Biol Phys 12:1239– optimal radioimmunotherapy dose for clinical response. Cancer 1242 94(4 Suppl):1275–1286