Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 67 5 Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation

G-One Ahn and J. Martin Brown

CONTENTS 5.1 Targeting Tumor Hypoxia 5.1 Targeting Tumor Hypoxia 67 5.2 Oxygen-Level Enhancers 68 5.3 Hypoxia-Selective Radiosensitizers 69 Tumor hypoxia was first postulated from histo- 5.3.1 Nitroimidazoles 69 logical studies of human lung adenocarcinomas by 5.3.2 Mixed-Function Radiosensitizers 69 Thomlinson and Gray (1955). They reasoned that, 5.3.3 DNA-Affinic Radiosensitizers 71 because of unrestrained growth, tumor cells are 5.3.4 Limitations of HSR 71 forced away from blood vessels beyond the effec- 5.4 Hypoxic Cytotoxins 72 5.4.1 Introduction 72 tive diffusion distance of oxygen (O2) in respiring 5.4.2 Combination of Hypoxic Cytotoxins with Ionizing tissues, hence becoming hypoxic and eventually Radiation 72 necrotic (Fig. 5.1a). Given the typical values for 5.4.3 Nitroimidazoles 73 intracapillary O2 tensions and consumption rates, 5.4.4 Other Nitroaromatics 73 they calculated that O diffusion distances would 5.4.4.1 CB 1954 73 2 be approximately 150 Pm and this was consistent 5.4.4.2 SN 23862 and PR-104 74 Thomlinson 5.4.4.3 RSU 1069 and RB 6145 74 with their histological observations ( 5.4.5 Quinones 75 and Gray 1955). This type of hypoxia has come to be 5.4.5.1 75 termed “chronic,” or “diffusion-limited,” hypoxia. 5.4.5.2 Porfiromycin 75 Acute hypoxia also develops in tumors through 5.4.5.3 EO9 75 temporal (reversible) cessation or reduction of 5.4.6 Benzotriene di-N-Oxides 76 5.4.7 Tertiary Amine N-Oxides 78 tumor blood flow resulting from highly disorga- 5.4.7.1 Nitracrine N-Oxides 78 nized tumor vasculature (Fig. 5.1b; Brown 1979). 5.4.7.2 AQ4N 79 Definitive evidence for acute hypoxia and fluctu- 5.5 Combination of Radiation-Activated Prodrugs ating blood flow has been demonstrated in trans- with Ionizing Radiation 79 planted tumors in mice injected at some time apart 5.5.1 Concepts of RAP 79 5.5.2 Nitro(Hetero)Cyclic Methylquarternary with two different diffusion limited fluorescent Ammonium Salts 79 dyes showing mismatch of labeled cells (Chaplin et 5.5.3 5- (5-FU)-Releasing Prodrugs 81 al. 1986; Trotter et al. 1989); however, acute and 5.5.4 Transition Metal Complexes 81 chronic hypoxia are in fact the two ends of a con- 5.6 Other Hypoxia-Targeting Strategies 82 tinuum with fluctuations in blood flow without total 5.6.1 GDEPT Targeting Tumor Hypoxia 82 5.6.2 Clostridia-Directed Enzyme Prodrug Therapy 82 occlusion, which are common in both experimental 5.6.3 Targeting HIF-1 83 (Kimura et al. 1996) and human tumors (Hill et 5.7 Conclusion 83 al. 1996), producing a dynamic situation with fluc- References 83 tuating oxygen diffusion distances in many parts of tumors. Tumor hypoxia is a major factor contributing to the failure of radiotherapy (Fig. 5.2). This is largely because DNA damage produced by ionizing radia- tion, which would otherwise become fixed and lethal G. Ahn, PhD to cells by reacting with O under well oxygenated J. M. Brown, P D 2 h conditions, can be restored to its undamaged form Division of Radiation and Cancer Biology, Department of Brown Wilson Radiation Oncology, Stanford School of Medicine, 269 Campus under hypoxic conditions ( and Drive, Center for Clinical Science and Research, Rm 1255, 2004). Clinically hypoxia predicts poor local control Stanford, CA 94305-5152, USA and survival of patients undergoing radiotherapy 68 G. Ahn and J. M. Brown

a b

Fig. 5.1. a A diagram of a tumor capillary and surrounding tumor cells at decreasing oxygen concentrations (in the direction of arrows). Cells become hypoxic (green) and eventually necrotic (blue); chronic hypoxia. Cellular proliferation and chemo- therapeutic drug concentration are also decreasing in the same direction, as a function of distance from the capillary. (From Brown 1999). b A diagram of normal (left) and tumor (right) blood vasculature. The tumor vasculature is highly disorganized resulting in acutely hypoxic regions in the tumor. (From Brown and Giaccia 1998)

teins, such as vascular endothelial growth factor (VEGF), are increased under hypoxic conditions, potentially resulting in increased tumor angiogene- sis (Shweiki et al. 1992; Fang et al. 2001). Thus, there is substantial evidence that hypoxia both interferes with the effective therapy of solid tumors and con- tributes to a more malignant phenotype; however, hypoxia may also prove to be a therapeutic advan- tage: because it is virtually unique to tumor cells, therapies that target hypoxic regions may have the potential to kill malignant cells while leaving non- malignant cells relatively untouched. This chapter discusses some examples of hypoxia targeting com- Fig. 5.2. A Kaplan-Meier plot of overall survival of patients pounds and approaches for combination with ioniz- with head and neck carcinoma undergoing radiotherapy. ing radiation in experimental or clinical settings. Well-oxygenated tumors (pO2 >10 mmHg; dotted line) showed better prognosis than poorly oxygenated (pO2 <10 mmHg; solid line) tumors. (From Brizel et al. 1997) 5.2 Oxygen-Level Enhancers for carcinoma of the head and neck (Nordsmark et al. 1996; Brizel et al. 1997), and cancer of the cervix One of the earliest attempts to overcome the prob- (Hockel et al. 1993; Fyles et al. 1998). lem of the resistance of hypoxic cells in tumors to Hypoxia further complicates cancer manage- radiotherapy was to increase O2 levels in the blood ment by limiting the access of conventional chemo- stream, thereby increasing the diffusion distance rown Wilson therapeutic drugs (Fig. 5.1a; B and of O2. A number of trials were performed with 2004). Hypoxia also increases genomic instability patients breathing 100% O2 at a pressure of 3 atmo- by increasing mutation frequency (Reynolds et al. spheres, but the results were mixed (Watson et al. 1996) or selecting for cells expressing an anti-apop- 1978; Dische et al. 1983; Henk 1986). One potential totic phenotype such as mutated p53 (Graeber et al. reason for such failures is that increasing the diffu- 1996). This leads to a more metastatic phenotype as sion distance of O2 would not be expected to reduce has been observed clinically (reviewed by Rofstad the levels of acute hypoxia. In some systems, the 2000). In addition, expression of proangiogenic pro- use of carbogen (95% O2/5% CO2) appears to have Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 69

greater effect than 100% O2 in increasing O2 level in 5.3.1 the blood stream (Rockwell 1997) possibly by pre- Nitroimidazoles venting the vasoconstriction caused by high partial pressures of O2. Metromidazole and misonidazole (Fig. 5.3) are the Other potential approaches to overcome hypoxia prototype members of this class. They were admin- include the use of nicotinamide (Horsman et al. istered to patients in the 1970s. Disappointingly, they 1987; Chaplin et al. 1991; Kjellen et al. 1991) in showed very little efficacy sensitizing tumors but a conjunction with carbogen (Corry and Rischin high incidence of peripheral neuropathy (Urtasun 2004), agents to increase tumor blood flow such as et al. 1976; Dische et al. 1977). With the limited flunarizine (Jirtle 1988), artificial blood substi- dose of misonidazole that can be administered to Teicher tutes carrying increased levels of O2 ( and patients, almost all of the clinical trials of radio- Rose 1984; Rockwell et al. 1986), drugs to reduce therapy combined with misonidazole were negative Hirst Wood Dische the affinity of hemoglobin for O2 ( and ( 1985); however, a meta-analysis of 50 ran- 1989), blood transfusion (Bush et al. 1978), and domized trials later showed a small but significant hyperthermia (Song et al. 2001). Recently, RSR13, benefit of misonidazole and other hypoxic radio- a drug reducing hemoglobin O2-binding affinity, sensitizers when added to radiotherapy in head and has been claim to benefit non-small cell lung cancer neck cancers (Overgaard 1994). patients receiving radiotherapy in phase-II trials Attempts to produce superior drugs to misoni- (Choy et al. 2005). dazole resulted in the development of etanidazole (Fig. 5.3; Brown et al. 1981), pimonidazole (Fig. 5.3; Smithen et al. 1980), and nimorazole (Fig. 5.3; Dische 1985); of these, nimorazole showed a signifi- 5.3 cant benefit of loco-regional control when given in Hypoxia-Selective Radiosensitizers conjunction with radiotherapy to patients with inva- sive carcinoma of larynx and pharynx (Overgaard In the 1960s Adams and Cooke (1969) proposed that et al. 1998; Overgaard et al. 2005) and is now given electron-affinic drugs might act like O2, a potent as part of the standard of care for radiotherapy of head electron-affinic molecule, to sensitize hypoxic and neck cancer patients in Denmark (Table 5.1). tumor cells. These agents (hypoxia-selective radi- osensitizers (HSR) mimic O2 by reacting with the short-lived DNA free radicals generated by ionizing 5.3.2 radiation; however, unlike O2, HSR are not rapidly Mixed-Function Radiosensitizers metabolized by the cells through which they pen- etrate and are thus able to reach areas beyond the The neurotoxicity of misonidazole stimulated the O2 diffusion distance. Some examples of HSR are search for drugs not only with less toxicity, but also discussed below. with increased efficiency as radiosensitizers on a

Metronidazole Misonidazole Etanidazole

Fig. 5.3. Examples of nitroimidazole compounds as hypoxia-selective Pimonidazole Nimorazole radiosensitizers (HSR) 70 G. Ahn and J. M. Brown et al. et al. et al. et al. ogaert B et al. (1998) et al. et al. (1989) et al. et al. (1996) et al. et al. (1993) et al. en et al. (2003) et al. et al. (2004) et al. et al. (1995); (1995); et al. Reference Henk Urtasun et al. (1994); (1994); et al. Beard Lawton Lee et al. (1997) et al. Eschwege Dische et al. (1998) et al. Overgaard (1995) et al. et al. Overgaard (1989b) Van D et al. et al. Overgaard Grigsby (1989a); (1999) Chan Simpson t t t t; unusually unusually t; t t t t t cant benefi cant cant benefi cant cant benefi cant cant benefi cant cant benefi cant cant benefi cant cant benefi cant benefi cant cant benefi cant No signifi No previous CHART study CHART previous good response in the control arm in the control good response regional control rate and cancer- rate control regional death) related Outcome 73 signifi No 61 Positive effect compared with 523 signifi No patients No. of No. y 422 effect (better loco- Positive ractionated ractionated accelerated radiation therapy therapy radiation accelerated (CHART) External beam radiation 39 58, signifi No Conventional radiotherapyConventional 374 521, signifi No Conventional radiotherapyConventional 183 signifi No Split-course radiotherapySplit-course 626 fractionation Conventional radiotherapy signifi No Radical radiotherapy Conventional radiotherapyConventional 120 331, signifi No Conventional radiotherapyConventional 239 signifi No sults from the clinical trials from sults Small-cell lung cancerSmall-cell lung radiotherapy Conventional 30 prostate cancer carcinoma uterine cervix Invasive carcinoma of carcinoma Invasive larynx and pharynx head and Advanced neck cancer Carcinoma of the Carcinoma cervix Carcinoma of the Carcinoma uterine cervix small cell lung cancer lung small cell Type of cancerType of radiotherapy Type Phase II and neck Head hyperf Continuous Phase III of the Carcinoma Phase III non- Locally advanced Phase II Locally advanced Phase III and neck Head radiotherap Conventional Phase III and neck Head Type of trial HSR Drug evalua- category tion Pimonidazole HSR Nimorazole HSR Etanidazole HSR Misonidazole HSR Name of Name drug , hypoxia-selective radiosensitizers hypoxia-selective , HSR Examples of radiosensitizers 5.1. Examples and re nitroimidazole Table Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 71 dosage basis. The latter strategy has been achieved by cellular metabolism occurring at 37qC (Roberts by incorporating other functional moieties in the et al. 1987) and poor extravascular penetration drug molecule. through tumor cell layers (Wilson et al. 1986). CB 1954 (2,4-dinitro-5-aziridinylbenzamide; 2-NLP-3 (5-[3-(2-nitro-1-imidazolyl)-propyl]-phen- Fig. 5.4) is an HSR-bearing DNA alkylating moiety anthridinium bromide; Figure 5.5) and 2-NLP-4 (5-[3-(2-nitro-1-imidazolyl)-butyl]-phenanthri- dinium bromide; Fig. 5.5) are 2-nitroimidazoles attached to the DNA intercalator phenanthridine. They were shown to be 10–100 times more effi- cient as HSR than misonidazole both in vitro and in vivo (Cowan et al. 1991). The structurally simi- lar compound, NLA-1 (9-[3-(2-nitro-1-imidazolyl) propylamino]acridine hydrochloride; Fig. 5.5) is CB 1954 RSU 1069 a DNA-targeted acridine-linked 2-nitroimidazole ( Papadopoulou et al. 1992). Although NLA-1 is a Fig. 5.4. Examples of mixed-function compounds as HSR potent HSR in vitro, it lacked in vivo activity (Denny et al. 1992). of , which showed more efficient radiosen- sitization than misonidazole (Chapman et al. 1979). 5.3.4 Similarly, RSU 1069 (1(2-nitro-1-imidazolyl)3- Limitations of HSR aziridinyl-2-propanol; Fig. 5.4), the second genera- tion of CB 1954, showed in vitro radiosensitization The major drawback of HSR is that the radiosensi- potency tenfold greater than misonidazole at equi- tization efficacy of HSR to tumors decreases at the molar doses (Stratford 1982; Adams et al. 1984); radiation doses used in clinically relevant frac- however, it appears that the more effective sensitiz- tionated irradiation. Experimental results showed ing ability of RSU 1069 is due to a greater degree of that 2–3 Gy fractionated irradiation resulted in hypoxic cell cytotoxicity rather than to enhanced lower enhancement ratios than single large doses radiosensitizing ability (Hill et al. 1986). Both CB for misonidazole (Denekamp and Stewart 1954 and RSU 1069 are discussed further as hypoxic 1978; Hill and Bush 1978; Sheldon and Fowler cytotoxins in section 5.4.4. (1978) or etanidazole (Brown and Yu 1984). One explanation for the lack of radiosensitization at fractionated low doses could be reoxygenation 5.3.3 occurring between each fraction (Brown and Yu DNA-Affinic Radiosensitizers 1984); however, an equally likely reason is that it is not the cells at maximum radiation resis- This class of HSR incorporates a DNA-affinic moiety tance, but those at intermediate oxygenation and thereby attracting the drug molecule closer to DNA intermediate radioresistance that dominate the where they exert their radiosensitizing effect. Nitra- response to fractionated irradiation (Wouters crine, 1-nitroacridine derivative (1-nitro-9-(dime and Brown 1997). Though hypoxic cell radiosen- thylaminopropylamino)acridine; Fig. 5.5) showed sitizers at clinically realistic doses can sensitize 1700 times more efficient in vitro radiosensitization the maximally hypoxic cells to radiation killing, than misonidazole (Roberts et al. 1987); however they have little effect on the radiosensitivity of the development of nitracrines as HSR was limited the cells at intermediate hypoxia and radiosen-

2-NLP-3 (n=3) Nitracrine or 2-NLP-4 (n=4) NLA-1

Fig. 5.5. Examples of DNA affi nic compounds as HSR 72 G. Ahn and J. M. Brown sitivity. This is a consequence of the logarithmic Selectivity for hypoxia is usually a result of a futile nature of the curve of radiosensitization vs dose: redox cycling in which the presence of O2 reoxidizes as cell sensitivity increases it takes geometrically the one-electron reduced intermediate thereby more drug to increase their radiosensitivity still regenerating the parent drug (Fig. 5.6a; Mason and oltzman x- further; thus, a hypoxic cell partially sensitized H 1975). Superoxide anion (O2 ), a major by some oxygen takes much more drug to increase by-product of this reaction, is capable of producing its sensitivity by a given amount compared with a DNA strand breaks and other oxidative damage, but fully hypoxic cell. can be detoxified by superoxide dismutase and cata- It thus seems unlikely that hypoxic cell radio- lase (Biaglow et al. 1982). Hypoxia selective cyto- sensitizers – even newer ones with greater potency toxicity therefore occurs when the reduction of the – have a major influence on the outcome of frac- hypoxic cytotoxin produces a more toxic interme- x- Wardman tionated radiotherapy. They may, however, play diate than the O2 radical ( et al. 1995; a role – or probably should play a role – in those Wardman 2001). situations where large, single doses of radiation are given such as with stereotactic radiotherapy of brain tumors. 5.4.2 Combination of Hypoxic Cytotoxins with Ionizing Radiation

5.4 Hypoxic cytotoxins are the ideal drug to combine Hypoxic Cytotoxins with ionizing radiation because they produce a pro- file of cytotoxicity as a function of distance from 5.4.1 blood vessels in tumors that are the opposite to that Introduction produced by ionizing radiation (Fig. 5.6b; Brown 1999). In other words, hypoxic cytotoxins kill the An alternative strategy to overcome the problem of tumor cells that are resistant to ionizing radiation. hypoxic tumor cells is to selectively kill them by In addition, hypoxic cytotoxins may release stable using hypoxic cytotoxins. Typically these are pro- and diffusible cytotoxins capable of killing the sur- drugs of very low cytotoxicity that are reduced by rounding tumor cells at relatively higher O2 con- enzyme(s) in hypoxic tumor cells. This results in centrations, producing a so-called bystander effect conversion to potent cytotoxins killing the activat- (Denny and Wilson 1993). Unlike HSR, the efficacy ing cell and, in some cases, the surrounding cells. of hypoxic cytotoxins is independent of the dose of The development of hypoxic cytotoxins was origi- radiation because there is no interaction between the nally stimulated by the findings that nitroimid- two agents; hence, the benefit of adding a hypoxic azoles were more toxic to hypoxic than oxic tumor cytotoxin to fractionated irradiation increases with cells even without irradiation (Sutherland 1974; the number of times the drug is administered and Hall and Roizon-Towle 1975). can produce greater benefit than if all of the tumor

a

Fig. 5.6. a The concept of hypoxic cytotoxins (indicated as D). Hypoxia selectivity is achieved by the futile cycle by oxygen converting the reduced intermediate (D•-) back to its non- toxic prodrug form (D). b The prediction of tumor cell killing by a hypoxic cytotoxin or radiation and most anticancer drugs as a function of the distance from the capillary. Note the oppo- site profi le in cytotoxicity towards tumor cells; the combina- tion of the two should yield complementary cell killing and is shown as a dashed line. (From Brown 1999) b Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 73 were fully oxygenated as shown in the theoretical Moselen et al. 1995), and NLCQ-1 (Papadopoulou study by Brown and Koong (1991). et al. 2000). NLCQ-1 is a nitroimidazole-linked chlo- roquinoline (4-[3-(2-nitro-1-imidazolyl)-propyl- amino]-7-chloroquinone hydrochloride; Fig. 5.8), a 5.4.3 member of NLP-1/NLA-1/THNLA-1 series, which is Nitroimidazoles also an efficient HSR (Papadopoulou et al. 1994; Papadopoulou et al. 1996). It shows time-dependent A general scheme (Fig. 5.7) for nitroheterocycle increase in hypoxic cytotoxicity in vitro and is cur- reduction is that the addition of an odd number rently under preclinical evaluation (Papadopoulou of electrons (1, 3, 5) leads to radical intermediates, et al. 2000; Squillace et al. 2000). while an even number of electrons (2, 4, 6) leads to In mammalian cells, aldehyde oxidase, DT- the nitroso (R-NO), hydroxylamine (R-NHOH), and diaphorase, xanthine oxidase, NADPH:cytochrome Mason amine reductants (R-NH2), respectively ( and P450 reductase, cytochrome b5 reductase, NADH- Holtzman 1975; Perez-Reyez et al. 1980). Oxygen dehydrogenase, and succinate dehydrogenase is able to reverse or inhibit reduction at the one-elec- (Heimbrook and Sartorelli 1986; Walton and tron radical anion, although it could in principle act Workman 1987; Hodgkiss 1998) have been reported at various stages (Wardman and Clarke 1976). In as nitroreductases. the absence of O2, further reduction occurs primar- x- ily via disproportionation reactions of R-NO2 , ulti- mately leading to the fragmentation of the imidazole 5.4.4 ring (Varghese et al. 1976; Flockhart et al. 1978). Other Nitroaromatics Two- and four-electron reduced derivatives may have different stabilities and reactivities depending 5.4.4.1 upon the nature of the aromatic ring and its sub- CB 1954 stituents (McClelland et al. 1984). Some examples of this class of hypoxic cytotoxins Though not strictly a hypoxic selective cytotoxin, are metronidazole, misonidazole (Fig. 5.8; Rauth et CB1954 has been an important lead for the develop- al. 1984), bis-nitroimidazole (Fig. 5.8; Hay et al. 1994; ment of such agents. CB 1954 (Fig. 5.9) is a mono-

Fig. 5.7. The reduction of nitroimidazole hypoxic cytotoxins. R generalized nitroimidazole ring

Metronidazole Misonidazole

Fig. 5.8. Examples of nitroimidazole Bis-nitroimidazole NLCQ-1 compounds as hypoxic cytotoxins 74 G. Ahn and J. M. Brown

RSU 1069

CB 1954 SN 23862

RB 6145

Fig. 5.9. Examples of nitroaromatic hypoxic cytotoxins functional alkylating agent that can be enzymatically et al. 2003). Good bystander effect in both in vitro activated to a potent difunctional alkylating agent and in vivo systems together with substrate speci- which crosslinks DNA (Knox et al. 1988a; Knox et ficity to Escherichia coli B nitroreductase warrants al. 1988b; Brown and Wilson 2004). Reduction of development of SN 23862 for GDEPT (Anlezark et CB 1954 produces the potent cytotoxic metabolites, al. 1995; Wilson et al. 2002). the 4-hydroxylamine, and 2-amine (Helsby et al. PR-104 is a phosphate pre-prodrug of the dini- 2004). The activation of CB 1954 also occurs through trobenzamide mustard PR-104A that is activated a second activation step by a non-enzymatic reac- in hypoxia to become a potent bifunctional alkyl- tion with a thioester (such as acetyl CoA) to form ating agent producing DNA interstrand crosslinks the final DNA reactive species, which is presumably (Douglas et al. 2005). In addition, PR-104 has dem- 4-(N-acetoxy)-5-(aziridin-1-yl)-2-nitrobenzamide onstrated substantial bystander effect killing aero- (Knox et al. 1991). The reduction of CB 1954 has bic as well as hypoxic cells in solid tumors (Wilson been shown to proceed at about equal rates under et al. 2005). Because of this ability to kill both aero- aerobic and hypoxic conditions, presumably because bic and hypoxic cells in tumors PR-104 shows sig- the two-electron reduction by DT-diaphorase is not nificant antitumor activity as a single agent alone. inhibited by O2. Currently, there is interest in CB It is also superior to tirapazamine in combination 1954 in gene-directed enzyme-prodrug therapy with fractionated irradiation in preclinical models (GDEPT) using Escherichia coli B. nitroreductase (Dorie et al., unpublished data). PR-104 entered (Bridgewater et al. 1995; Green et al. 1997), and clinical phase-1 trials in December 2005. is in currently phase-I clinical trials (Chung-Faye et al. 2001; Dachs et al. 2005). 5.4.4.3 RSU 1069 and RB 6145 5.4.4.2 SN 23862 and PR-104 RSU 1069 (Fig. 5.9) is bioreductively activated by NADPH-cytochrome P450 reductase forming a Structurally similar to CB 1954, a nitrogen bifunctional crosslinking agent (Stratford et al. mustard compound, SN 23862 (5-[N,N-bis(2- 1986; Whitmore and Gulyas 1986). In air, RSU 1069 chloroethyl)amino]-2,4-dinitrobenzamide; Fig. 5.9) functions as a monofunctional alkylating agent due exploits the nitro-group as an electronic switch in to the presence of the aziridine group ( Stratford et that nitro-to-amine conversion shifts electron den- al. 1986). It shows in vivo antitumor activity against sity modifying the reactivity of a drug molecule KHT sarcoma and RIF-1 tumor when given before (Siim et al. 1997; Helsby et al. 2003). The chemi- or after irradiation (Hill et al. 1986). cal reduction of each of the 2-nitro and the 4-nitro The bromoethylamine derivative RB 6145 group of SN 23862 showed an increase in cytotoxic- (Fig. 5.9) was developed as a prodrug of RSU 1069 ity by 160- and 9-fold in AA8 cell line, and by 4400- because of irreversible gastrotoxicity observed in and 83-fold in UV 4 cell line, respectively, and that early phase-I trials of RSU 1069 (Horwich et al. the reduction of both nitro groups led to further 1986). RB 6145 showed slightly less hypoxia selec- increase in cytotoxicity (Palmer et al. 1995; Helsby tive cytotoxicity than RSU 1069 in vitro (Jenkins Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 75 et al. 1990) but was active against hypoxic cells in 1986). Mitomycin C can also be reductively activated vivo with lowered toxicity compared with RSU 1069 via two-electron reducing DT-diaphorase generat- Jenkins Cole Iyanagi ( et al. 1990; et al. 1990); however, fur- ing an O2-insensitive hydroquinone ( and ther development of this compound was stopped Yamazaki 1970; Keyes et al. 1984). because preclinical studies showed irreversible Despite variable results in hypoxia selective cyto- cytotoxicity toward retinal cells in mice (Parker et toxicity observed in preclinical studies ( Rockwell al. 1996; Breider et al. 1998). and Kennedy 1979; Rockwell et al. 1982; Keyes et al. 1984), clinical trials have reported that mito- mycin C in combination with radiation shows a sig- 5.4.5 nificant benefit in local regional control rates for Quinones patients with head and neck cancer (Weissberg et al. 1989; Haffty et al. 1993; Haffty et al. 1997), 5.4.5.1 squamous cell carcinoma of the cervix (Roberts et Mitomycin C al. 2000), and laryngeal and hypopharyngeal cancer (Saarilahti et al. 2004) (Table 5.2). Mitomycin C (Fig. 5.10), isolated from Streptomyces caespitosus, was introduced into the clinic in 1958, 5.4.5.2 and was subsequently shown to have a moderate in Porfiromycin vitro hypoxia selective cytotoxicity [hypoxic cyto- toxicity ratio (concentration of drug in air divided Porfiromycin (Fig. 5.10), a second-generation version by concentration of drug in hypoxia to produce the of mitomycin C, showed superior in vitro hypoxic same level of cell killing; Brown 1993) of about 1–5; selectivity over mitomycin C as a result of lowered Rockwell et al. 1982; Fracasso and Sartorelli aerobic cytotoxicity (Fracasso and Sartorelli 1986]. The cytotoxicity of mitomycin C is associ- 1986; Rockwell et al. 1988) and an improved in ated with formation of monofunctional alkylation vivo therapeutic index as a result of higher LD50 (the and more potently with intra- and inter-strand dosage required to kill 50% of the treated popula- crosslinks of DNA, all of which require bioreduc- tion) in mice (Keyes et al. 1985). Although a phase-I tive activation (Iyer and Szybalski 1963; Tomasz trial showed an acceptable toxicity profile, a recent et al. 1987; Volpato et al. 2005). phase-III randomized trial showed that porfiromy- The one-electron reduction of mitomycin C results cin was inferior to mitomycin C as an adjunct to in a semiquinone, which under hypoxic conditions radiotherapy in squamous cell cancer of the head activates the aziridine ring and results in binding and neck (Haffty et al. 2005). For this and other of the drug to DNA (Pan et al. 1984). Following the reasons (Brown (1999), it therefore seems unlikely initial covalent attachment of mitomycin C to DNA, that the clinical activity of mitomycin C is the result the drug can undergo further reductive activation to of its (modest) selectivity as a hypoxic cytotoxin. form a second alkylating site (Pan et al. 1984). The one-electron reduction pathway can be catalysed 5.4.5.3 by any of several enzymes including NADPH:cyto- EO9 chrome P450 reductase (Pan et al. 1984; Keyes et al. 1984) and xanthine oxidase (Pan et al. 1984), in EO9 ([3-hydroxymethyl-5-aziridinyl-2-methyl-2- Bachur a process that can be reversed by O2 ( et al. (H-indole-4,7-indione)-propenol]; Fig. 5.10) was 1978; Bachur et al. 1979; Pritsos and Sartorelli originally developed as a synthetic analog of mito-

Mitomycin C Porfi romycin EO9

Fig. 5.10. Examples of quinones 76 G. Ahn and J. M. Brown mycin C (Oostveen and Speckamp 1987). The triazinyl radical, which can mediate the initial cyto- reductive bioactivation of EO9 occurs in a similar toxic process by abstracting a hydrogen atom from fashion to mitomycin C such that one- and two- the deoxyribose backbone of DNA (Anderson et al. electron reduction processes yield the correspond- 2003). The tirapazamine radical may also lead to the ing semiquinone and hydroquinone, respectively formation of the two-electron reduced product, SR (Maliepaard et al. 1995). The semiquinone is 4317, a major non-toxic metabolite detected in cul- believed to be more cytotoxic than the hydroqui- tured hypoxic cells (Baker et al. 1988; Hicks et al. none based on its ability to produce DNA interstrand 2003), in the mouse (Walton and Workman (1993), crosslinks and strand breaks (Bailey et al. 1993; and in humans (Grahams et al. 1997). Formation of Plumb et al. 1994; Maliepaard et al. 1995). SR 4317 can occur by a number of different routes, Although partial responses were observed including the direct two-electron reduction of tira- in a small number of patients in phase-I trials pazamine by DT-diaphorase (Riley and Workman (Schellens et al. 1994), no apparent antitumor 1992), by radical disproportionation reaction, or by activity by EO9 alone was demonstrated in phase- hydrogen abstraction from macromolecules other II studies in patients with advanced breast, gastric, than DNA (Brown and Wang 1998). Dirix pancreatic, and colorectal carcinoma ( et al. Tirapazamine has a unique O2 concentration 1996); however, some concerns about the design dependency such that its cytotoxicity does not level of the trials were raised in that enzymatic activ- off at high concentrations, but gradually decreases as Koch Hicks ity in patients’ tumors was not measured routinely the O2 concentration increases ( 1993; et (Phillips 1996) and that hypoxic cytotoxins, such al. 2004). Tirapazamine shows hypoxic cytotoxicity as EO9, should be combined with other treatment ratios of up to 200 in murine and 50 in human cell modalities, such as radiotherapy or chemother- lines (Zeman et al. 1986). apy, to demonstrate detectable clinical responses The hypoxic cytotoxicity of tirapazamine is due ( Workman and Stratford 1993). to the formation of DNA strand breaks resulting in chromosome aberrations (Beidermann et al. 1991; Wang et al. 1992; Siim et al. 1996). The chromosome 5.4.6 breaks produced by tirapazamine were shown to be Benzotriene di-N-Oxides less easily repaired than those produced by X-rays, and this has been suggested to be a result of prob- Tirapazamine (SR 4233; Fig. 5.11a) is the prototype able metabolism by reductases located close to DNA of this class of hypoxic cytotoxins. Under hypoxic (Wang et al. 1992). Although a large proportion of conditions, tirapazamine is reduced to an O2-sensi- tirapazamine is metabolized in the cytoplasm by tive tirapazamine radical (Lloyd et al. 1991). The enzymes, such as cytochrome P450 (Walton et al. tirapazamine radical then eliminates a water mol- 1992; Wang et al. 1993; Riley et al. 1993), NADPH ecule to form a nitrogen-centered oxidant, benzo- cytochrome P450 reductase (Cahill and White

a

Fig. 5.11. a Chemical structure of tirapazamine. b Synergistic activity of tirapazamine in killing SCCVII tumors in vivo when combined with fractionated irradiation (fi lled circles), deter- Relative clonogenic cells/tumor Relative mined by clonogenic cell survival. Tirapazamine alone and radiation alone (2.5 Gy per fraction) are shown as open circles and dots, respectively. From Brown and Lemmon (1990) Number of fractions b Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 77 et al. (2004) et al. et al. (1989) et al. (2005) et al. (2000) et al. et al. (1991) et al. et al. (2004) et al. et al. (2001) et al. Saarilahti Widder Roberts Weissberg et al. (1993) Haffty et al. (1995) McKeown et al. Teicher (1990) Cole et al. Rischin Rischin et al. (1990) Cole et al. (2005) et al. Papadopoulou Reference t cant benefi cant ve effect ve Additive effect Additive Positive effect Positive effect Positive effect (4-year disease-free effect (4-year Positive survival) disease-free effect (5-year Positive survival free and local recurrence survival) Positive effect loco- 3-year free survival rates; rates) failure-free regional Outcome u3 Gy) Additive effect u3 Gy) Additive effect u1 Gy) No signifi nitive RT nitive failure- effect (3-year Positive 21 patients with accelerated with21 patients accelerated RT hyperfractionated 160 patients with160 patients radical RT with120 patients RT conventional 182 patients with182 patients RT conventional single-dose RT12-Gy effect Additive Fractionated RT (5 RT Fractionated Fractionated RT (5 RT Fractionated 122 patients, defi 122 patients, 5-Gy single-dose RT5-Gy with convention- 16 patients effect Additive RT fractionated ally Fractionated RT (4 RT Fractionated No. of patients and/or type of and/or patients No. of RT brosarcoma Advanced squa- Advanced FSaIIC murine FSaIIC murine KHT sarcoma single-dose RT 10-Gy Advanced laryngeal Advanced T50/80 murine T50/80 murine and hypopharyngeal cancer Advanced head and Advanced neck cancer mous-cell carcinoma carcinoma mous-cell of the cervix cell Squamous of the carcinoma head and neck mammary carci- noma fi KHT sarcoma single-dose RT 10-Gy Additi xenograft head and neck cancer Locally advanced advanced Locally U251 human glioma U251 human Type of cancer/ Type evaluated tumor ecent clinical/preclinical evaluation data evaluation clinical/preclinical ecent Phase III Phase-II trialrandomized evaluation system Clinical Phase II Clinical Phase I Preclinical vivo In Evaluation of trial/ Type BEP category , radiotherapy , AQ4N BEP RSU-1069 and BEP HSR Preclinical vivo In RB-6145 and BEP HSR Preclinical vivo In NLCQ-1 and BEP HSR Preclinical vivo In Tirapaza- mine Mitomycin C Mitomycin BEP RT Name of drugName Drug evaluation Potential candidates of hypoxic cytotoxins and their r of hypoxic candidates 5.2. Potential Table 78 G. Ahn and J. M. Brown

1990; Lloyd et al. 1991; Walton et al. 1992; Wang 5.4.7 et al. 1993; Patterson et al. 1995; Patterson et al. Tertiary Amine N-Oxides 1997), xanthine oxidase (Laderoute et al. 1988), and DT-diaphorase (Riley and Workman 1992; 5.4.7.1 Wang et al. 1993), it is the nuclear metabolism of Nitracrine N-Oxides tirapazamine (ca. 20% of the overall cellular metab- olism) that accounts for essentially all of the tira- Nitracrine N-oxide (Fig. 5.12a) was developed as a pazamine-induced DNA damage under hypoxic prodrug of nitracrine, a hypoxia cytotoxin whose conditions (Evans et al. 1998; Delahoussaye et al. cytotoxicity is due to nitroreduction that is inhibited Wilson 2001). Recently, tirapazamine in hypoxic conditions by O2 ( et al. 1986). While nitracrine showed has also shown to markedly inhibit DNA replication the degree of hypoxic selectivity similar to misoni- (Peters et al. 2001) and to poison topoisomerase-II dazole (ca. tenfold) in vitro, its metabolism was too activity (Peters and Brown 2002) in vitro leading rapid to provide selective killing of hypoxic cells in to the suggestion that the DNA double-strand breaks vivo (Wilson et al. 1986). Derivatization with ter- produced by tirapazamine result from its poisoning tiary amine N-oxide lowered DNA binding (15-fold), topoisomerase II (Peters and Brown 2002). cell uptake, and aerobic cytotoxicity compared with Tirapazamine potentiates cell killing with frac- nitracrine, while its hypoxic selectivity was greatly tionated irradiation in mouse tumors (Fig. 5.11b; increased (1000- to 1500-fold; Wilson et al. 1992). Brown and Lemmon 1990), and with in The very high hypoxia selectivity was suggested to a highly schedule-dependent manner (Dorie and be due to a requirement for both the nitro and N- Brown 1993). Tirapazamine is currently in phase- oxide moieties for full activation in an O2 inhibitable III evaluation and appears particularly effective in manner (Wilson et al. 1992). Despite some improve- combination with cisplatin (Rodriguez et al. 1996; ment of extravascular diffusion properties observed Miller 1997; Treat 1998; von Powel 2000) or in vitro (Wilson et al. 1992), in vivo activity against as an adjunct to cisplatin/radiotherapy treatment KHT tumors was only observed at doses lethal to the (Rischin et al. 2001; Rischin et al. 2005). host (Lee et al. 1996).

Nitracrine N-oxide AQ4N a

Fig. 5.12. a Examples of tertiary amine N-oxide compounds. b Tumor growth delay of T50/80 in BDF mice treated with AQ4N. Combination with a single dose radiation (fi lled squares) pro- duced a signifi cantly greater antitumor activity compared with b AQ4N alone (open squares). (From McKeown et al. 1996) Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 79

- 5.4.7.2 prodrugs (RAP) are reduced by eaq (aquated/ AQ4N hydrated electron) generated from the radiolysis of water, offering two distinct mechanisms strongly AQ4N (1,4-bis-{[2-(dimethylamino-N-oxide)ethyl] inhibited by O2. These mechanisms involve either amino}5,8-dihydroxyanthracene-9,10-dione; back-oxidization of the one-electron reduction x- - Fig. 5.12a) is a prodrug designed to be prevented intermediate of RAP (RAP ) or to scavenge eaq at from binding to DNA (Patterson 1993; Smith et diffusion controlled rate (Fig. 5.13; Other primary al. 1997b) until metabolized in hypoxic cells to give radicals formed by radiolysis of water (OHx and Hx) AQ4, a stable, oxygen insensitive metabolite (Smith are expected to be scavenged at very high rates by et al. 1997a). AQ4 is a DNA intercalator and potent other biomolecules. Some of the resulting organic inhibitor of DNA topoisomerase II ( Patterson radicals also have reducing properties so might 1993; Patterson et al. 1994; Smith et al. 1997b). make a further contribution to prodrug reduction. Early development of AQ4N has been confounded The RAP approach offers a number of potential by the lack of hypoxia selective activity in several advantages such as selective activation within the cell lines; however, a large (>100-fold) increase radiation field (tumor bearing volume); exploita- in cytotoxicity of AQ4N was observed when cells tion of necrotic hypoxic regions lacking reductase were incubated under hypoxic conditions with liver activity through release of a stable cytotoxin with microsomes (Patterson 1993). It was later shown a good bystander effect, no requirement for expres- that AQ4N is not activated by NAD(P)H-dependent sion of specific reductase(s); lack of two-electron cytochrome P450 reductase per se (Patterson et activation; and improved extravascular transport al. 1999), but by cytochrome P450 (CYP) isoforms. by allowing design of prodrugs that are not sub- Several studies have shown that the metabolism of strates for metabolism in tumors (Wilson et al. AQ4N correlates with levels of CYP1A1, 2B6, and 1998). Some of the main classes of the compounds 3A in humans (Patterson and McKeown 2000), that have been considered as candidates for RAP are 3A in mice (Patterson et al. 2000), and 2B and 2E discussed below. in rats (Raleigh et al. 1999). Reduction of the two N-oxide functionalities of AQ4N has been suggested to be involved in an oxygen atom transfer from the N-oxide side chains in a process that is O2 sensitive (Patterson and McKeown 2000). Moreover, heme- containing systems other than CYP, such as nitric oxide synthase, have also shown to mediate AQ4N reduction (Raleigh et al. 1998). AQ4N has shown in vivo antitumor activity when combined with radia- tion (Fig. 5.12b; McKeown et al. 1995), McKeown et Fig. 5.13. The concept of radiation-activated prodrugs (RAP). Hypoxia selectivity occurs by the futile cycling of oxygen al. 1996) or when combined with a range of methods reverting the reduced intermediate of RAP (RAP•-) or scav- inducing additional tumor hypoxia, e.g., hydralazine - enging hydrated electron (eaq ) generated from the radiolysis (Patterson et al. 2000), dimethylxanthenone acetic of water acid (Wilson et al. 1996), or clamping (Patterson et al. 2000). It is currently in phase-I clinical trials. 5.5.2 Nitro(Hetero)Cyclic Methylquarternary Ammonium Salts 5.5 Combination of Radiation-Activated NMQ ammonium salts such as 4-nitroimidazole Prodrugs with Ionizing Radiation (SN 25341; N,N-bis(2-chloroethyl)-N-methyl-N-[(1- methyl-4-nitro-5-imidazolyl)methyl]ammonium 5.5.1 chloride; 4-NIQ-HN2; Fig. 5.14a) and 5-nitropyrrole Concepts of RAP (N,N-bis(2-chloroethyl)-N-methyl-N-[(1-methyl- 5-nitro-1-pyrrolyl)methyl]ammonium chloride; 5- Another way to target hypoxic tumor cells is to use NPQ-HN2; Fig. 5.14a) that were originally developed ionizing radiation, rather than enzymes, to effect as hypoxic cytotoxins (Tercel et al. 2001), have the reduction of the prodrug. Radiation-activated also been reported as RAP that are reduced with 80 G. Ahn and J. M. Brown

SN 25341 5-NPQ-HN2

a 4-NBQ-AMAC 4-NIQ-AMAC 5-NPQ-AMAC

Fig. 5.14. a Examples of NMQ compounds as RAP. b Radiolytic acti- vation of 4-NIQ-HN2 in human plasma determined as apparent IC50 against UV4 cells. Oxic (95% O2 and 5% CO2) and anoxic (95% N2 and 5% CO2) are shown in open and fi lled circles, respectively. The IC50 for b authentic HN2 is also shown (fi lled square) with an arrow

one-electron stoichiometry by pulse and steady- reduction potential of the prodrug (therefore sus- state radiolysis (Anderson et al. 1997; Wilson et ceptible for enzymatic reduction) and only modest al. 1998). Irradiating 4-NIQ-HN2 in anoxic human cytotoxicity of HN2 limited their utility to that of plasma, followed by exposure to UV4 cells showed only model compounds. that IC50 (concentration of a compound giving 50% In order to incorporate a more potent cyto- growth inhibition relative to the control cells) was toxin within the prodrug system, 4-NBQ-AMAC markedly decreased with radiation, approaching a (Fig. 5.14a), 4-NIQ-AMAC (Fig. 5.14a), and 5-NPQ- value equal to that of the cytotoxin, mechloretha- AMAC (Fig. 5.14a) were synthesized. These prodrugs mine (HN2), itself after ca. 20 Gy (Fig. 5.14b; Wilson were two orders of magnitude less cytotoxic than the et al. 1998); however, a relatively high one-electron released cytotoxin (AMAC) against a panel of tumor Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 81 cell lines (Wilson et al. 1998). Irradiation in anoxic OFU001 (1-(2’-oxopropyl)-5-fluorouracil; Fig. 5.15) culture medium followed by exposure to UV4 cells and OFU101 (Fig. 5.15) have been reported to showed significant activation of 4-NIQ-AMAC and release 5-FU and 5-fluoro-2’deoxyuridine (FdUrd), 5-NPQ-AMAC at 2 Gy, although the yield of AMAC respectively, with higher efficiency (two-electron was lower than that for the HN2 analogs (Wilson stoichiometry) in anoxic buffer upon irradiation et al. 1998). The AMAC prodrugs showed appre- (Shibamoto et al. 2000; Shibamoto et al. 2004). - ciable enzymatic reduction in hypoxic cell cultures, The mechanism proposed is that attachment of eaq , and convulsion in mice by a mechanism unrelated generated by radiolysis of water, weakens the N(1)- to the cytotoxin release (Wilson et al. 1998), sug- C(1’) bond that links 5-FU or FdUrd to the oxoalkyl gesting that they are not likely to be useful as RAP side chain. Although these compounds showed cell prodrugs. killing after hypoxic irradiation, they failed to dem- onstrate any significant antitumor activity in vivo (Shibamoto et al. 2000, 2001, 2004). 5.5.3 5-Fluorouracil (5-FU)-Releasing Prodrugs 5.5.4 Nishimoto et al. (1992) first reported that the Transition Metal Complexes N(1)-C(5)-linked dimer of 5-FU (1-(5’-fluoro-6’- hydroxy-5’,6’-dihydrouracil-5’-yl)-5-fluorouracil) A number of transition metal complexes, such as releases 5-FU with ca. three-electron stoichiometry those of cobalt (III) (Co(III) (Fig. 5.16; Wilson et by irradiation in anoxic aqueous buffer and some al. 1994; Ahn et al. 2004b) and copper (II) (Cu(II) indication of antitumor effect in combination with (Fig. 5.16; Parker et al. 2004; Torre et al. 2005), radiation in mice bearing SCCVII tumors. Recently, have been reported as hypoxia-selective prodrugs. In particular, Co(III) complexes are kinetically inert when not reduced; however, when reduced radio- - 6 lytically by eaq , the inert d electron spin state of Co(III) becomes labile d7, which dramatically weak- ens the coordination bond hence releasing the cyto- toxin (Fig. 5.17; Ware et al. 1993). A Co(III) complex bearing a very potent DNA minor groove alkylator cyclopropylindoline (Fig. 5.17) has demonstrated a number of attractive features as a RAP such as: a good masking of the cytotoxicity of cyclopropyl- OFU001 OFU001 indoline in the intact Co(III) complex; an efficient release of the cytotoxin with clinically relevant Fig. 5.15. Chemical structures of 5-FU prodrugs of RAP radiation dose of 2 Gy in anoxic human plasma

Co(III) complex Cu(II) complex

Fig. 5.16. Chemical structures of Co(III) and Cu(II) complexes of hypoxic prodrugs 82 G. Ahn and J. M. Brown

Inactive IR

– eaq +

DNA alkylation Fig. 5.17. A Co(III) complex of cyclopropylindoline releasing its cytotoxin upon ionizing radiation (IR)

or buffer; cell exclusion property of the complex ing an HRE sequence into an appropriate expres- enhancing extravascular transport through multi- sion cassette (Dachs et al. 1997). With this system, cellular tumor layers; a HCR of up to 20 in human studies have shown that in transfected tumor cells, colorectal HT29 cells (Ahn et al. 2002; Ahn 2003); NADPH:cytochrome P450 reductase enhances the however, further development of this Co(III) com- antitumor efficacy of RSU 1069 or tirapazamine in plex as a RAP has failed due to the lack of efficacy combination with ionizing radiation (Patterson et in RIF-1 bearing mice when combined with ionizing al. 2002; Cowen et al. 2004). Another approach has radiation (Ahn 2003). shown antitumor activity with HRE driven nitrore- ductase enhancing the activity of CB1954 (Shibata et al. 2002). Dual responsive promoters have also been devel- 5.6 oped whose expression is driven not only by HRE Other Hypoxia-Targeting Strategies but also by radiation-responsive promoter sequence such as the early growth response-1 (Egr-1) to 5.6.1 enhance the reporter gene expression in response to GDEPT Targeting Tumor Hypoxia hypoxia in conjunction with low dose of radiation (Greco et al. 2002; Chadderton et al. 2005). As discussed in section 5.1, many genes supporting anaerobic metabolism and angiogenesis are upregu- lated under hypoxic conditions by the transcription 5.6.2 factor, hypoxia-inducible factor-1 (HIF-1). Specifi- Clostridia-Directed Enzyme Prodrug Therapy cally, HIF-1, once it heterodimerizes with D and E subunits, binds to a common hypoxia-responsive The above-mentioned GDEPT systems have the major element (HRE) found in the enhancer region of drawback of utilizing ex vivo manipulated (trans- all HIF-1 responsive genes (Semenza 2001). One fected) tumor cell lines implying that their clini- GDEPT approach targeting tumor hypoxia aims to cal applicability may be difficult. To overcome this express an exogenous therapeutic gene by introduc- limitation Brown and colleagues have exploited the Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 83 necrotic regions as a target for cancer therapy using Interestingly, ionizing radiation has been recently a non-pathogenic strain of the bacterial genus Clos- shown to upregulate HIF-1 activity in tumors tridium, an obligate anaerobe (Lemmon et al. 1994; ( Moeller et al. 2004); however, the effect of HIF-1 Fox et al. 1996). This Gram-positive, spore-forming on the radiosensitivity of the tumor is controversial. bacteria becomes vegetative and grows only in the Moeller et al. (2005) showed that functional HIF-1 absence or at very low levels of oxygen (Brown and enhances tumor radiosensitivity by promoting ATP Wilson 2004). Clinical studies in 1970s already have metabolism, cell proliferation, and p53 activation, proven the safety of Clostridium and its tumor-spe- whereas others have reported that it is the defi- cific germination (Carey et al. 1967; Heppner and ciency in HIF-1 that increases tumor radiosensitiv- Mose 1978; Heppner et al. 1983). Recently, geneti- ity, independent of p53 function (Unruh et al. 2003; cally engineered Clostridium expressing E. coli Williams et al. 2005). The reason for this discrep- enzyme cytosine deaminase showed tumor-specific ancy is yet to be determined, but the difference in delivery of the enzyme and the consequent antitu- experiment systems (RNA interference vs cell lines mor activity upon 5-fluorocytosine (5-FC) prodrug deficient for either HIF-1D or -E) could have contrib- administration (Liu et al. 2002; Ahn (2004a). The uted. combination of CDEPT with ionizing radiation is expected to result in enhanced antitumor activity because of ionizing radiation killing well-oxygen- ated tumor cells and/or the radiosensitizing abilities 5.7 of 5-FC and 5-FU. Conclusion

The potential clinical benefit of exploiting tumor 5.6.3 hypoxia by combining a hypoxia activated drug with Targeting HIF-1 conventional cancer therapy has yet to be realized in routine clinical practice. Despite this, the positive In addition to its role in hypoxic conditions, HIF-1 clinical results with the combination of the hypoxic can be upregulated in the tumor in an O2-inde- cytotoxin tirapazamine with cisplatin in advanced pendent manner. Oncogenic mutations and loss- non-small cell lung cancer and with chemoradio- of-function mutations in tumor-suppressor genes therapy with advanced head and neck cancer demon- have been shown to be associated with the increased strate the potential of this approach. There is a good activity of HIF-1 (Semenza 2003; Li et al. 2005). reason to expect that future drugs or strategies will Evidence for HIF-1 serving as a potential target do better: indeed advances made in experimental for cancer therapy is substantial and extensively models identifying the determinants of the efficacy reviewed elsewhere (Semenza 2003; Giaccia et al. of these hypoxia-targeting compounds, together 2003). For example, HIF-1D (the O2 sensitive HIF-1 with other strategies to exploit tumor hypoxia, auger subunit) expression has been shown to be a poor well for the future of this field. prognostic factor in a number of cancers including bladder cancer (Nakanishi et al. 2005; Theodo- ropoulos et al. 2004), pancreatic cancer (Shibaji et al. 2003), and breast carcinoma (Bos et al. 2003). References HIF-1 is also reported to promote tumor growth in many studies (Ryan et al. 1998; Carmeliet et al. Adams GE, Cooke MS (1969) Electron-affinic sensitization. I. 1998; Chen et al. 2003; Stoeltzing et al. 2004). A structural basis for chemical radiosensitizers in bacteria. Int J Radiat Biol Relat Stud Phys Chem Med 15:457–471 Some recent approaches targeting HIF-1 include Adams GE, Ahmed I, Sheldon PW, Stratford IJ (1984) Radia- using small molecule inhibitors blocking transcrip- tion sensitization and chemopotentiation: RSU 1069, a tional activity of HIF-1 (Kung 2004; Kong et al. compound more efficient than misonidazole in vitro and 2005) or those blocking HIF-1D protein synthesis in vivo. Br J Cancer 49:571–577 (Tan et al. 2005), inhibitors of HIF-1 downstream Ahn G (2003) Investigation of an aza-chloromethylbenzindo- Majumder Zhong line cobalt(III) complex as a hypoxia-activated prodrug for target ( et al. 2004; et al. 2004; cancer therapy. Pathology Auckland, University of Auck- Fang et al. 2005), enhancement of HIF-1D protein land, New Zealand degradation (Li et al. 2004), and antisense (Sun et Ahn G, Ware DC, Botting KJ, Kriste AG, Denny WA, Wilson WR al. 2005) or RNA interference against HIF-1D (Li (2002) A novel Co(III) complex of the DNA minor-groove et al. 2005). binder aza-seco-CBI-TMI as a hypoxia-selective prodrug 84 G. Ahn and J. M. Brown

that is activated by enzymatic or radiolytic reduction. Proc carcinoma of the head and neck. Int J Radiat Oncol Biol Am Assoc Cancer Res 43:656 Phys 38:285–289 Ahn G, Liu SC, Menke D, Dorie MJ, Brown JM (2004a) Enhanced Brown JM (1979) Evidence for acutely hypoxic cells in mouse antitumor activity of 5-FC by genetically engineered anaer- tumours, and a possible mechanism of reoxygenation. Br obic bacteria C. sporogenes expressing cytosine deaminase J Radiol 52:650–656 when combined with a vascular targeting agent, ZD6126 Brown JM (1993) SR 4233 (tirapazamine): a new anticancer or DMXAA. Second International Conference on Vascular drug exploiting hypoxia in solid tumours. Br J Cancer Targeting, Miami, Florida 67:1163–1170 Ahn GO, Ware DC, Denny WA, Wilson WR (2004b) Optimiza- Brown JM (1999) The hypoxic cell: a target for selective cancer tion of the auxiliary ligand shell of cobalt(III)(8-hydroxy- therapy. Eighteenth Bruce F. Cain Memorial Award Lecture. quinoline) complexes as model hypoxia-selective radia- Cancer Res 59:5863–5870 tion-activated prodrugs. Radiat Res 162:315–325 Brown JM, Giaccia AJ (1998) The unique physiology of solid Anderson RF, Denny WA, LI W, Packer JE, Tercel M, Wilson tumors: opportunities (and problems) for cancer therapy. WR (1997) Pulse radiolysis studies on the fragmentation Cancer Res 58:1408–1416 of arylmethyl quaternary nitrogen mustards by one-elec- Brown JM, Koong A (1991) Therapeutic advantage of hypoxic tron reduction in aqueous solution. J Phys Chem 101:9704– cells in tumors: a theoretical study. J Natl Cancer Inst 9709 83:178–185 Anderson RF, Shinde SS, Hay MP, Gamage SA, Denny WA Brown JM, Lemmon MJ (1990) Potentiation by the hypoxic (2003) Activation of 3-amino-1,2,4-benzotriazine 1,4-diox- cytotoxin SR 4233 of cell killing produced by fractionated ide antitumor agents to oxidizing species following their irradiation of mouse tumors. Cancer Res 50:7745–7749 one-electron reduction. J Am Chem Soc 125 748–756 Brown JM, Wang LH (1998) Tirapazamine: laboratory data rel- Anlezark GM, Melton RG, Sherwood RF, Wilson WR, Denny evant to clinical activity. Anticancer Drug Des 13:529–539 WA, Palmer BD, Knox RJ, Friedlos F, Williams A (1995) Bio- Brown JM, Wilson WR (2004) Exploiting tumour hypoxia in activation of dinitrobenzamide mustards by an E. coli B cancer treatment. Nat Rev Cancer 4:437–447 nitroreductase. Biochem Pharmacol 50:609–618 Brown JM, Yu NY (1984) Radiosensitization of hypoxic cells Bachur NR, Gordon SL, Gee MV (1978) A general mechanism in vivo by SR 2508 at low radiation doses: a preliminary for microsomal activation of quinone anticancer agents to report. Int J Radiat Oncol Biol Phys 10:1207–1212 free radicals. Cancer Res 38:1745–1750 Brown JM, Yu NY, Brown DM, Lee WW (1981) SR-2508: A 2- Bachur NR, Gordon SL, Gee MV, Kon H (1979) NADPH-cyto- nitroimidazole amide which should be superior to miso- chrome P-450 reductase activation of quinone anticancer nidazole as a radiosensitizer for clinical use. Int J Radiat agents to free radicals. Proc Natl Acad Sci USA 76:954–957 Oncol Biol Phys 7:695–703 Bailey SM, Lewis AD, Patterson LH, Fischer GR, Workman P Bush RS, Jenkin RDT, Allt WEC, Beale FA, Bean H, Dembo AJ, (1993) Free radical generation following reduction of EO9: Pringle JF (1978) Definitive evidence for hypoxic cells influ- involvement in cytotoxicity. Br J Cancer 67:8 encing cure in cancer therapy. Br J Cancer 37:302–306 Baker MA, Zeman EM, Hirst VK, Brown JM (1988) Metabolism Cahill A, White IN (1990) Reductive metabolism of 3-amino- of SR 4233 by Chinese hamster ovary cells: basis of selective 1,2,4-benzotriazine-1,4-dioxide (SR 4233) and the induc- hypoxic cytotoxicity. Cancer Res 48:5947–5952 tion of unscheduled DNA synthesis in rat and human Beard C, Buswell L, Rose MA, Noql L, Johnson D, Coleman derived cell lines. Carcinogenesis 11:1407–1411 CN (1994) Phase-II trial of external beam radiation with Carey RW, Holland JF, Whang HY, Neter E, Bryant B (1967) etanidazole (SR 2508) for the treatment of locally advanced Clostridial oncolysis in man. Eur J Cancer 3:37–46 prostate cancer. Int J Radiat Oncol Biol Phys 29:611–616 Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Beidermann KA, Wang J, Graham RP, Brown JM (1991) SR 4233 Dewerchin M, Neeman M, Bono F, Abramovitch R, Maxwell cytotoxicity and metabolism in DNA repair-competent and P, Koch CJ, Ratcliffe P, Moons L, Jain RK, Collen D, Keshert repair-deficient cell cultures. Br J Cancer 63:358–362 E (1998) Role of HIF-1 alpha in hypoxia-mediated apop- Biaglow JE, Varnes ME, Koch CJ, Sridhar R (1982) Metabolic tosis, cell proliferation and tumour angiogenesis. Nature activation of carcinogenic nitro-compounds to oxygen 394:485–490 reactive intermediates. In: Floyd RA (ed) Free radicals and Chadderton N, Cowen RL, Sheppard FCD, Robinson S, Greco cancer. Marcel Dekker, New York O, Scott SD, Stratford IJ, Patterson AV, Williams KJ (2005) Bos R, Van der Groep P, Greijer AE, Shvarts A, Meijer S, Pinedo Dual responsive promoters to target therapeutic gene HM, Semenza GL, Van Diest PJ, Van der Wall E (2003) expression to radiation-resistant hypoxic tumor cells. Int Levels of hypoxia-inducible factor-1D independently pre- J Radiat Oncol Biol Phys 62:213–222 dict prognosis in patients with lymph node negative breast Chan P, Milosevic M, Fyles AW, Carson J, Pintilie M, Rauth M, carcinoma. Cancer 97:1573–1581 Thomas G (2004) A phase III randomized study of misoni- Breider MA, Pilcher GD, Graziano MJ, Gough AW (1998) Reti- dazole plus radiation vs radiation alone for cervix cancer. nal degeneration in rats induced by CI-1010, a 2-nitroimi- Radiother Oncol 70:295–299 dazole radiosensitizer. Toxicol Pathol 26:234–239 Chaplin DJ, Durand RE, Olive PL (1986) Acute hypoxia in Bridgewater JA, Springer CJ, Knox RJ, Minton NP, Michael NP, tumors: implications for modifiers of radiation effects. Int Collins MK (1995) Expression of the bacterial nitroreduct- J Radiat Oncol Biol Phys 12:1279–1282 ase enzyme in mammalian cells renders them selectively Chaplin DJ, Horsman MR, Aoki DS (1991) Nicotinamide, Fluo- sensitive to killing by the prodrug CB1954. Eur J Cancer sol DA and carbogen: a strategy to oxygenate acutely and 31A:2362–2370 chronically hypoxic cells in vivo. Br J Cancer 63:109–113 Brizel DM, Sibley GS, Prosnitz LR, Scher RL, Dewhirst MW Chapman JD, Raleigh JA, Pederson L, Ngan J, Shum FY, Meeker (1997) Tumor hypoxia adversely affects the prognosis of BE, Urtasun RC (1979) Potentially three distinct roles for Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 85

hypoxic cell radiosensitizers in the clinic. Proc 6th Int Dische S, Anderson PJ, Sealy R, Watson ER (1983) Carcinoma of Congr Radiat Res, Tokyo the cervix: anaemia, radiotherapy and hyperbaric oxygen. Chen J, Zhao S, Nakada K, Kuge Y, Tamaki N, Okada F, Wang J, Br J Radiol 56:251–255 Shindo M, Higashiro F, Takeda K, Asaka M, Katoh H, Sugi- Dische S, Chassagne D, Hope-Stone HF, Dawes PJDK, Roberts yama T, Hosokawa M, Kobayashi M (2003) Dominant-nega- JT, Yosef H, Bey P, Horiot J-C, Jacobson A, Frankendal B, tive hypoxia-inducible factor-1D reduces tumorigenicity of Gonzales Gonzales D, Nguyen TD, Daly NJ, Le Floch O, pancreatic cancer cells through the suppression of glucose Newman H, Vieiro E, Bennett MH, Bichel P, Duvillard P, metabolism. Am J Pathol 162:1283–1291 Jacobson A, Cook PA, Everett V, Machin D (1993) A trial Choy H, Nabid A, Stea B, Scott C, Roa W, Kleinberg L, Ayoub of Ro 03-8799 (pimonidazole) in carcinoma of the uterine J, Smith C, Souhami L, Hamburg S, Spanos W, Kreisman cervix: an interim report from the Medical Research Coun- H, Boyd AP, Cagnoni PJ, Curran WJ (2005) Phase II multi- cil Working Party on advanced carcinoma of the cervix. center study of induction followed by con- Radiother Oncol 26:93–103 current efaproxiral (RSR13) and thoracic radiotherapy for Dische S, Saunders MI, Lee ME, Adams GE, Flockhart IR (1977) patients with locally advanced non-small-cell lung cancer. Clinical testing of the radiosensitizer Ro 07-0582: experi- J Clin Oncol 23:5918–5928 ence with multiple doses. Br J Cancer 35:567–579 Chung-Faye G, Palmer D, Anderson D, Clark J, Downes M, Bad- Dorie MJ, Brown JM (1993) Tumor-specific, schedule-depend- deley J, Hussain S, Murray PI, Searle P, Seymour L, Harris ent interaction between tirapazamine (SR 4233) and cispla- PA, Ferry D, Kerr DJ (2001) Virus-directed, enzyme pro- tin. Personal communication drug therapy with nitroimidazole reductase: a phase I and Douglas RS, Patterson AV, Yang S, Wilson WR, Siim BG (2005) pharmacokinetic study of its prodrug, CB1954. Clin Cancer Induction of DNA damage and repair responses by the Res 7:2662–2668 hypoxia-activated prodrug PR-104. AACR-NCI-EORTC Cole S, Stratford IJ, Adams GE, Fielden EM, Jenkins TC (1990) Conference on Molecular Targets and Cancer Therapeutics: Dual-function 2-nitroimidazoles as hypoxic cell radiosen- Discovery, Biology, and Clinical Applications. Philadelphia sitizers and bioreductive cytotoxins: in vivo evaluation in Eschwege F, Sancho-Garnier H, Chassagne D, Brisgand D, KHT murine sarcomas. Radiat Res 124:S38–S43 Guerra M, Malaise EP, Bey P, Busutti L, Cionini L, N’guyen Corry J, Rischin D (2004) Strategies to overcome accelerated T, Romanini A, Chavaudra J, Hill C (1997) Results of an repopulation and hypoxia: What have we learned from European randomized trial of etanidazole combined with clinical trials? Semin Oncol 31:802–808 radiotherapy in head and neck carcinomas. Int J Radiat Cowan DS, Panicucci R, Mcclelland RA, Rauth AM (1991) Tar- Oncol Biol Phys 39:275–281 geting radiosensitizers to DNA by attachment of an inter- Evans JW, Yudoh K, DeLahoussaye YM, Brown JM (1998) Tira- calating group: nitroimidazole-linked phenanthridines. pazamine is metabolized to its DNA-damaging radical by Radiat Res 127:81–89 intranuclear enzymes. Cancer Res 58:2098–2101 Cowen RL, Williams KJ, Chinje EC, Jaffar M, Sheppard FCD, Fang J, Yan L, Shing Y, Moses MA (2001) HIF-1 alpha-mediated Telfer BA, Wind NS, Stratford IJ (2004) Hypoxia targeted up-regulation of vascular endothelial growth factor, inde- gene therapy to increase the efficacy of tirapazamine as an pendent of basic fibroblast growth factor, is important in adjuvant to radiotherapy: reversing tumor resistance and the switch to the angiogenic phenotype during early tum- effecting cure. Cancer Res 64:1396–1402 origenesis. Cancer Res 61:5731–5735 Dachs GU, Patterson AV, Firth JD, Ratcliffe PJ, Townsend KMS, Fang J, Xia C, Cao Z, Zheng JZ, Reed E, Jiang B-H (2005) Api- Stratford IJ, Harris AL (1997) Targeting gene expression to genin inhibits VEGF and HIF-1 expression via PI3K/AKT/ hypoxic tumor cells. Nat Med 3:515–520 p70S6K1 and HDM2/p53 pathways. FASEB J 19:342–353 Dachs GU, Tupper J, Tozer GM (2005) From bench to bedside Flockhart IR, Large P, Troup D, Malcolm SH, Marten TR (1978) for gene-directed enzyme prodrug therapy of cancer. Anti Pharmacokinetic and metabolic studies of the hypoxic cell Cancer Drugs 16:349–359 radiosensitizer misonidazole. Xenobiotica 8:97–105 Delahoussaye YM, Evans JW, Brown JM (2001) Metabolism of Fox ME, Lemmon MJ, Mauchline ML, Davis TO, Giaccia AJ, tirapazamine by multiple reductases in the nucleus. Bio- Minton NP, Brown JM (1996) Anaerobic bacteria as a chem Pharmacol 62:1201–1209 delivery system for cancer gene therapy: activation of 5- Denekamp J, Stewart FA (1978) Sensitization of mouse tumours fluorocytosine by genetically engineered clostridia. Gene using fractionated X-irradiation. Br J Cancer 37:259–263 Therapy 3:173–178 Denny WA, Wilson WR (1993) Bioreducible mustards: a para- Fracasso PM, Sartorelli AC (1986) Cytotoxicity and DNA digm for hypoxia-selective prodrugs of diffusible cytotox- lesions produced by mitomycin C and porfiromycin in ins (HPDCs). Cancer Met Rev 12:135–151 hypoxic and aerobic EMT6 and Chinese hamster ovary Denny WA, Roberts PB, Anderson RF, Brown JM, Phil D, Wilson cells. Cancer Res 46:3939–3944 WR (1992) NLA-1: A 2-nitroimidazole radiosensitizer tar- Fyles AW, Milosevic M, Pintilie M, Hill RP (1998) Cervix cancer geted to DNA by intercalation. Int J Radiat Oncol Biol Phys oxygenation measured following external radiation ther- 22:553–556 apy. Int J Radiat Oncol Biol Phys 42:751–753 Dirix LY, Tonnesen F, Cassidy J, Epelbaum R, Bokkel Huinink Giaccia A, Siim BG, Johnson RS (2003) HIF-1 as a target for WW, Pavlidis N, Sorio R, Gamucci T, Wolff I, Te VA, Lan J, drug development. Nat Rev Drug Discovery 2:1–9 Verweij J (1996) EO9 Phase II study in advanced breast, Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe gastric, pancreatic and colorectal carcinoma by the EORTC SW, Giaccia AJ (1996) Hypoxia-mediated selection of cells Early Clinical Studies Group. Eur J Cancer 32A:2019–2022 with diminished apoptotic potential in solid tumours. Dische S (1985) Chemical sensitisers for hypoxic cells: a Nature 379:88–91 decade of experience in clinical radiotherapy. Radiother Grahams MA, Senan S, Robin HJ, Eckhardt N, Lendrem D, Oncol 3:97–115 Hinks J, Greenslade D, Rampling R, Kaye SB, Roemeling R 86 G. Ahn and J. M. Brown

von, Workman P (1997) of the hypoxic Henk JM, Bishop K and Shepherd SF (2003) Treatment of head cell cytotoxic agent tirapazamine and its major biore- and neck cancer with CHART and nimorazole: phase II ductive metabolites in mice and humans: retrospective study. Radiother Oncol 66:65–70 analysis of a pharmacokinetically guided dose-escalation Heppner F, Mose JR (1978) The liquefaction (oncolysis) of strategy in a phase I trial. Cancer Chemother Pharmacol malignant gliomas by a non pathogenic clostridium. Acta 40:1–10 Neuro 12 123–125 Greco O, Marples B, Dachs GU, Williams KJ, Patterson AV, Scott Heppner F, Mose J, Ascher PW, Walter G (1983) Oncolysis of SD (2002) Novel chimeric gene promoters responsive to malignant gliomas of the brain. Thirteenth Int Cong Che- hypoxia and ionizing radiation. Gene Ther 9:1403–1411 mother 226:38–45 Green NK, Youngs DJ, Neoptolemos JP, Friedlos F, Knox RJ, Hicks KO, Pruijn FB, Sturman JR, Denny WA, Wilson WR (2003) Springer CJ, Anlezark GM, Michael NP, Melton RG, Ford Multicellular resistance to tirapazamine is due to restricted MJ, Young LS, Kerr DJ, Searle PF (1997) Sensitization of extravascular transport: a pharmacokinetic/pharmacody- colorectal and pancreatic cancer cell lines to the prodrug namic study in HT29 multicellular layer cultures. Cancer 5-(aziridin-1-yl)-2,4-dinitrobenzamide (CB1954) by retro- Res 63:5970–5977 viral transduction and expression of the E. coli nitroreduct- Hicks KO, Siim BG, Pruijn FB, Wilson WR (2004) Oxygen ase gene. Cancer Gene Ther 4:229–238 dependence of the metabolic activation and cytotoxicity Grigsby PW, Winter K, Wasserman TH, Marcial VA, Rotman M, of tirapazamine: implications for extravascular transport Cooper JS, Keys H, Asbell SO, Phillips TL (1999) Irradiation and activity in tumors. Radiat Res 161:656–666 with or without misonidazole for patients with stages IIIB Hill RP, Bush RS (1978) The effect of misonidazole in com- and IVA carcinoma of the cervix: final results of RTOG 80- bination with radiation dose fractionation. Br J Cancer 05. Int J Radiat Oncol Biol Phys 44:513–517 37:255–258 Haffty BG, Son YH, Sasaki CT, Papac R, Fischer D, Rockwell S, Hill RP, Gulyas S, Whitmore GF (1986) Studies of the in vivo Sartorelli AC, Fischer JJ (1993) Mitomycin C as an adjunct and in vitro cytotoxicity of the drug RSU-1069. Br J Cancer to postoperative radiation therapy in squamous cell carci- 53:743–751 noma of the head and neck: results from two randomized Hill SA, Pigott KH, Saunders MI, Powell ME, Arnold S, Obeid clinical trials. Int J Radiat Oncol Biol Phys 27:241–250 A, Ward G, Leahy M, Hoskin PJ, Chaplin DJ (1996) Microre- Haffty BG, Son YH, Papac R, Sasaki CT, Weissberg JB, Fischer gional blood flow in murine and human tumours assessed D, Rockwell S, Sartorelli AC, Fischer JJ (1997) Chemo- using laser Doppler microprobes. Br J Cancer 27 (Suppl): therapy as an adjunct to radiation in the treatment of S260–S263 squamous cell carcinoma of the head and neck: results Hirst DG, Wood PJ (1989) Altered radiosensitivity in a mouse of the Yale Mitomycin Randomized Trials. J Clin Oncol carcinoma after administration of clofibrate and benzafi- 15:268–276 brate. Radiother Oncol 15:55–61 Haffty BG, Wilson LD, Son YH, Cho EI, Papac RJ, Fischer DB, Hockel M, Knoop C, Schlenger K, Vorndran B, Baussmann E, Rockwell S, Sartorelli AC, Ross DA, Sasaki CT, Fischer JJ Mitze M, Knapstein PG, Vaupel P (1993) Intratumoral pO2 (2005) Concurrent chemo-radiotherapy with mitomycin C predicts survival in advanced cancer of the uterine cervix. compared with porfiromycin in squamous cell cancer of Radiother Oncol 26:45–50 the head and neck: final results of a randomized clinical Hodgkiss RJ (1998) Use of 2-nitroimidazoles as bioreduc- trial. Int J Radiat Oncol Biol Phys 61:119–128 tive markers for tumour hypoxia. Anticancer Drug Des Hall EJ, Roizon-Towle L (1975) Hypoxic radiosensitisers: 13:687–702 radiobiological studies at the cellular level. Radiology Horsman MR, Chaplin DJ, Brown JM (1987) Radiosensitiza- 117:453–457 tion by nicotinamide in vivo: a greater enhancement of Hay MP, Wilson WR, Moselen JW, Palmer BD and Denny WA tumor damage compared to that of normal tissues. Radiat (1994) Hypoxia-selective antitumor agents. 8. Bis(nitroimi Res 109:479–489 dazolyl)alkanecarboxamides: a new class of hypoxia-selec- Horwich A, Holliday SB, Deacon JM, Peckham MJ (1986) A tox- tive cytotoxins and hypoxic cell radiosensitisers. J Med icity and pharmacokinetic study in man of the hypoxic-cell Chem 37 381–391 radiosensitizer RSU-1069. Br J Radiol 59:1238–1240 Heimbrook DC, Sartorelli AC (1986) Biochemistry of miso- Iyanagi T, Yamazaki I (1970) One-electron reactions in bio- nidazole by NADPH-cytochrome c (P-450) reductase. Mol chemical systems. V. Differences in the mechanism of Pharmacol 29:168–172 quinone reduction by the NADP dehydrogenase and the Helsby NA, Wheeler SJ, Pruijn FB, Palmer BD, Yang S, Denny HAD(P)H dehydrogenase (DT-diaphorase). Biochim Bio- WA, Wilson WR (2003) Effect of nitroreduction on the phys Acta 216:282–294 alkylating reactivity and cytotoxicity of the 2,4-dinitroben- Iyer NV, Szybalski W (1963) and porfiromycin: zamide-5-aziridine CB1954 and the corresponding nitro- chemical mechanisms of activation and cross-linking of gen mustard SN 23862: distinct mechanisms of bioreduc- DNA. Microbiology 50:355–362 tive activation. Chem Res Toxicol 16:469–478 Jenkins TC, Naylor MA, O’Neill P, Threadgill MD, Cole S, Stratford Helsby NA, Ferry DM, Patterson AV, Pullen SM, Wilson WR IJ, Adams GE, Fielden EM, Suto MJ, Stier MA (1990) Synthe- (2004) 2-Amino metabolites are key mediators of CB 1954 sis and evaluation of alpha-[[(2-haloethyl)amino]methyl]- and SN 23862 bystander effects in nitroreductase GDEPT. 2-nitro-1H-imidazole-1-ethanols as prodrugs of alpha-[[(2- Br J Cancer 90:1084–1092 aziridinyl)methyl]2-nitro-1H-imidazole-1-ethanol (RSU- Henk JM (1986) Late results of a trial of hyperbaric oxygen 1069) and its analogues which are radiosensitizers and bio- and radiotherapy in head and neck cancer: a rationale for reductively activated cytotoxins. J Med Chem 33:2603–2610 hypoxic cell radiosensitizers. Int J Radiat Oncol Biol Phys Jirtle R (1988) Chemical modification of tumour blood flow. 12:1339–1341 Int J Hyperthermia 4:355–371 Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 87

Keyes SR, Fracasso PM, Heimbrook DC, Rockwell S, Sligar Lee HH, Wilson WR, Ferry DM, Van Zijl P, Pullen SM, Denny SG, Sartorelli AC (1984) Role of NADPH:cytochrome c WA (1996) Hypoxia-selective antitumour agents. 13. Effects reductase and DT-diaphorase in the biotransformation of of acridine substitution on the hypoxia-selective cytotoxic- mitomycin C. Cancer Res 44:5638–5643 ity and metabolic reduction of the bis-bioreductive agent Keyes SR, Rockwell S, Sartorelli AC (1985) Porfiromycin as nitracrine N-oxide. J Med Chem 39:2508–2517 a bioreductive alkylating agent with selective toxicity to Lemmon MJ, Elwell JH, Brehm JK, Mauchline ML, Minton hypoxic EMT6 tumor cells in vivo and in vitro. Cancer Res NP, Giaccia AJ, Brown JM (1994) Anaerobic bacteria as a 45:3642–3645 gene delivery system to tumors. Proc Am Assoc Cancer Res Kimura H, Braun RD, Ong ET, Hsu R, Secomb TW, Papahad- 35:374 jopoulos D, Hong K, Dewhirst MW (1996) Fluctuations in Li M-H, Miao Z-H, Tan W-F, Yue J-M, Zhang C, Lin L-P, Zhang red cell flux in tumor microvessels can lead to transient X-W, Ding J (2004) Pseudolaric acid B inhibits angiogen- hypoxia and reoxygenation in tumor parenchyma. Cancer esis and reduces hypoxia-inducible factor 1D by promot- Res 56:5522–5528 ing proteasome-mediated degradation. Clin Cancer Res Kjellen E, Joiner MC, Collier JM, Johns J and Rojas A (1991) A 10:8266–8274 therapeutic benefit from combining normobaric carbogen Li YM, Zhou BP, Deng J, Pan Y, Hay N, Hung M-C (2005) A or oxygen with nicotinamide in fractionated X-ray treat- hypoxia-independent hypoxia-inducible factor-1 activa- ments. Radiother Oncol 22:81–91 tion pathway induced by phophatidylinositol-3 kinase/Akt Knox RJ, Boland MP, Friedlos F, Coles B, Southan C, Roberts in HER2 overexpressing cells. Cancer Res 65:3257–3263 JJ (1988a) The nitroreductase enzyme in Walker cells that Liu SC, Minton NP, Giaccia AJ, Brown JM (2002) Anticancer activates 5-(aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) efficacy of systemically delivered anaerobic bacteria as to 5-(aziridin-1-yl)-4-hydroxylamino-2-nitrobenzamide is gene therapy vectors targeting tumor hypoxia/necrosis. a form of NAD(P)H dehydrogenase (quinone) (EC 1.6.99.2). Gene Ther 9:291–296 Biochem Pharmacol 37:4671–4677 Lloyd RV, Duling DR, Rumyantseva GV, Mason RP, Bridson Knox RJ, Friedlos F, Jarman M, Roberts JJ (1988b) A new PK (1991) Microsomal reduction of 3-amino-1,2,4-ben- cytotoxic, DNA interstrand crosslinking agent, 5-(aziri- zotriazine-1,4-dioxide to a free radical. Mol Pharmacol din-1-yl)-4-hydroxylamino-2-nitrobenzamide, is formed 40:440–445 from 5-(aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) by Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q, Mcmahon nitroreductase enzyme in Walker carcinoma cells. Biochem LM, Manola J, Brugarolas J, Mcdonnell TJ, Golub TR, Loda Pharmacol 37:4661 M, Lane HA, Sellers WR (2004) mTOR inhibition reverses Knox RJ, Friedlos F, Marchbank T, Roberts JJ (1991) Bioacti- Akt-dependent prostate intraepithelial neoplasia through vation of CB 1954: reaction of the active 4-hydroxylamino regulation of apoptotic and HIF-1-dependent pathways. derivative with thioesters to form the ultimate DNA-DNA Nat Med 10:594–601 interstrand crosslinking species. Biochem Pharmacol Maliepaard M, Wolfs A, Groot SE, De Mol NJ, Janssen LHM 42:1691–1697 (1995) Indoloquinone EO9: DNA interstrand cross-linking Koch CJ (1993) Unusual oxygen concentration dependence upon reduction by DT-diaphorase or xanthine oxidase. Br of toxicity of SR-4233, a hypoxic cell toxin. Cancer Res J Cancer 71:836–839 53:3992–3997 Mason RP, Holtzman JL (1975) The mechanism of microsomal Kong D, Park EJ, Stephen AG, Calvani M, Cardellina JH, Monks and mitochondrial nitroreductase. Biochemistry 14:1626– A, Fisher RJ, Shoemaker RH, Melillo G (2005) Echinomycin, 1632 a small-molecule inhibitor of hypoxia-inducible factor-1 McClelland RA, Fuller JR, Seaman NE, Rauth AM, Battistella DNA-binding activity. Cancer Res 65:9047–9055 R (1984) 2-hydroxylaminoimidazoles: unstable intermedi- Kung AL, Zabludoff SD, France DS, Freedman SJ, Tanner EA, ates in the reduction of 2-nitroimidazoles. Biochem Phar- Vieira A, Cornell-Kennon S, Lee J, Wang B, Wang J, Mem- macol 33:303–309 mert K, Naegeli H-U, Petersen F, Eck MJ, Bair KW, Wood McKeown SR, Friery OP, Mcintyre IA, Hejmadi MV, Patterson AW, Livingston DM (2004) Small molecule blockade of LH, Hirst DG (1996) Evidence for a therapeutic gain when transcriptional coactivation of the hypoxia-inducible AQ4N or tirapazamine is combined with radiation. Br J factor pathway. Cancer Cell 6:33–43 Cancer 27 (Suppl):S39–S42 Laderoute K, Wardman P, Rauth AM (1988) Molecular mecha- McKeown SR, Hejmadi MV, Mcintyre IA, Mcaleer JJ, Patterson nisms for the hypoxia-dependent activation of 3-amino- LH (1995) AQ4N: an alkylaminoanthraquinone N-oxide 1,2,4-benzotriazine-1,4-dioxide (SR 4233). Biochem Phar- showing bioreductive potential and positive interaction macol 37:1487–1495 with radiation in vivo. Br J Cancer 72:76–81 Lawton CA, Coleman CN, Buzydlowski JW, Forman JD, Marcial Miller VA, Ng KK, Grant SC, Kindler H, Pizzo B, Heelan RT, VA, Delrowe JD, Rotman M (1996) Results of a Phase-II trial Roemeling R von, Kris MG (1997) Phase II study of the of external beam radiation with etanidazole (SR 2508) for combination of the novel bioreductive agent, tirapazamine, the treatment of locally advanced prostate cancer (RTOG with cisplatin in patients with advanced non-small-cell protocol 90-20). Int J Radiat Oncol Biol Phys 36:673–680 lung cancer. Ann Oncol 8:1269–1271 Lee D-J, Cosmatos D, Marcial VA, Fu KK, Rotman M, Cooper Moeller BJ, Cao Y, Li CY, Dewhirst MW (2004) Radiation acti- JS, Ortiz HG, Beitler JJ, Abrams RA, Curran WJ, Coleman vates HIF-1 to regulate vascular radiosensitivity in tumors: CN, Wasserman TH (1995) Results of an RTOG phase role of reoxygenation, free radicals, and stress granules. III trial (RTOG 85-27) comparing radiotherapy plus eta- Cancer Cell 5:429–441 nidazole with radiotherapy alone for locally advanced Moeller BJ, Dreher MR, Rabbani ZN, Schroeder T, Cao Y, Li CY, head and neck carcinomas. Int J Radiat Oncol Biol Phys Dewhirst MW (2005) Pleiotropic effects of HIF-1 blockade 32:567–576 on tumor radiosensitivity. Cancer Cell 8:99–110 88 G. Ahn and J. M. Brown

Moselen JW, Hay MP, Denny WA, Wilson WR (1995) N-[2-(2- tetrahydroacridine hydrochloride. A novel DNA-affinic methyl-5-nitroimidazolyl)ethyl]-4-(2-nitroimidazolyl)but hypoxic cell cytotoxin and radiosensitizer. Comparison anamide (NSC 639862), a bisnitroimidazole with enhanced with NLA-1. Oncol Res 6:439–448 selectivity as a bioreductive drug. Cancer Res 55:574–580 Papadopoulou MV, JI M, Bloomer WD (1996) THNLA-1 as Nakanishi K, Hiroi S, Tominaga S, Aida S, Kasamatsu H, Mat- radio/chemosensitiser of EMT-6 tumours in mice. Br J suyama S, Matsuyama T, Kawai T (2005) Expression of Cancer 74:S267–S270 hypoxia-inducible factor-1D protein predicts survival in Papadopoulou MV, Ji M, Rao MK, Bloomer WD (2000) 4-[3- patients with transitional cell carcinoma of the upper uri- (2-nitro-1-imidazolyl)propylamino]-7-chloroquinoline nary tract. Clin Cancer Res 11:2583–2590 hydrochloride (NLCQ-1), a novel bioreductive agent as Nishimoto SI, Hatta H, Ureshima H, Kagiya T (1992) 1-(5’- radiosensitizer in vitro and in vivo: comparison with tira- fluoro-6’-hydroxy-5’,6’-dihydrouracil-5’-yl)-5-fluorouracil, pazamine. Oncol Res 12:325–333 a novel N(1)-C(5)-linked dimer that releases 5-fluorouracil Papadopoulou MV, Bloomer WD, Hollingshead MG (2005) by radiation activation under hypoxic conditions. J Med NLCQ-1 (NSC 709257) in combination with radiation Chem 35:2711–2712 against human glioma U251 xenografts. Anticancer Res Nordsmark M, Overgaard M, Overgaard J (1996) Pretreat- 25:1865–1869 ment oxygenation predicts radiation response in advanced Parker RF, Vincent PW, Elliot WL (1996) Alkylating property of squamous cell carcinoma of the head and neck. Radiother nitro-imidazole radiosensitizers is required for induction Oncol 41:31–39 of murine retinal degeneration. Vet Pathol 33:625 Oostveen EA, Speckamp WN (1987) Mitomycin analogs. I. Parker LL, Lacy SM, Farrugia LJ, Evans C, Robins DJ, O’Hare Indoloquinones as (potential) bisalkylating agents. Tetra- CC, Hartley JA, Jaffar M, Stratford IJ (2004) A novel design hedron 43:255–262 strategy for stable metal complexes of nitrogen mustards as Overgaard J (1994) Clinical evaluation of nitroimidazoles as bioreductive prodrugs. J Med Chem 47:5683–5689 modifiers of hypoxia in solid tumors. Oncol Res 6:509– Patterson AV, Barham HM, Chinje EC, Adams GE, Harris AL, 518 Stratford IJ (1995) Importance of P450 reductase activity Overgaard J, Bentzen SM, Kolstad P, Kjoerstad K, Davy M, Ber- in determining sensitivity of breast tumour cells to the telsen K, Mantyla M, Frankendal B, Skryten A, Loftquist bioreductive drug, tirapazamine (SR 4233). Br J Cancer I, Pedersen M, Sell A, Hammer R (1989a) Misonidazole 72:1144–1150 combined with radiotherapy in the treatment of carci- Patterson AV, Saunders MP, Chinje EC, Talbot DA, Harris AL, noma of the uterine cervix. Int J Radiat Oncol Biol Phys Stratford IJ (1997) Overexpression of human NADPH:cyto- 16:1069–1072 chrome c (P450) reductase confers enhanced sensitivity to Overgaard J, Hansen HS, Anderson AP, Hjelm-Hansen M, Jor- both tirapazamine (SR 4233) and RSU 1069. Br J Cancer gensen K, Sandberg E, Berthelsen A, Hammer R, Pedersen 76:1338–1347 M (1989b) Misonidazole combined with split-course radio- Patterson AV, Williams KJ, Cowen RL, Jaffar M, Telfer BA, therapy in the treatment of invasive carcinoma of larynx Saunders M, Airley R, Honess D, Van der Kogel AJ, Wolf and pharynx: report from the DAHANCA 2 study. Int J CR, Stratford IJ (2002) Oxygen-sensitive enzyme-prodrug Radiat Oncol Biol Phys 16:1065–1068 gene therapy for the eradication of radiation-resistant solid Overgaard J, Hansen HS, Overgaard M, Bastholt L, Berthelsen tumours. Gene Ther 9:946–954 A, Specht L, Lindelov B, Jorgensen KA (1998) A randomized Patterson LH (1993) Rationale for the use of aliphatic N-oxides double-blind phase III study of nimorazole as a hypoxic of cytotoxic as prodrug DNA binding radiosensitizer of primary radiotherapy in supraglottic agents: a new class of bioreductive agent. Cancer Met Rev larynx and pharynx carcinoma. Results of Danish Head 12:119–134 and Neck Cancer Study (DAHANCA) Protocol 5-85. Radio- Patterson LH, McKeown SR (2000) AQ4N: a new approach ther Oncol 46:135–146 to hypoxia-activated cancer chemotherapy. Br J Cancer Overgaard J, Eriksen JG, Nordsmark M, Alsner J, Horsman 83:1589–1593 MR (2005) Plasma osteopontin, hypoxia, and response Patterson LH, Craven MR, Fisher GR, Teesdale-Spittle P (1994) to the hypoxia sensitiser nimorazole in radiotherapy Aliphatic amine N-oxides of DNA binding agents as biore- of head and neck cancer: results from the DAHANCA 5 ductive drugs. Oncol Res 6:533–538 randomised double-blind placebo-controlled trial. Lancet Patterson LH, McKeown SR, Robson T, Gallagher R, Raleigh Oncol 6:757–764 SM, Orr S (1999) Antitumour prodrug development using Palmer BD, Van Zijl P, Denny WA, Wilson WR (1995) Reduc- cytochrome P450 (CYP) mediated activation. Anticancer tive chemistry of the novel hypoxia-selective cytotoxin Drug Des 14:473–486 5-[N,N-bis(2-chloroethyl)amino]-2,4-dinitrobenzamide. J Patterson LH, McKeown SR, Ruparelia K, Double JA, Bibby Med Chem 38 1229–1241 MC, Cole S, Stratford IJ (2000) Enhancement of chemo- Pan SS, Andrews PA, Glover CJ, Bachur NR (1984) Reductive therapy and radiotherapy of murine tumours by AQ4N, activation of mitomycin C and mitomycin C metabolites a bioreductively activated anti-tumour agent. Br J Cancer catalysed by NADPH-cytochrome P-450 reductase and 82:1984–1990 xanthine oxidase. J Biol Chem 259:959–966 Perez-Reyez E, Kalyanaraman B, Mason RP (1980) The reduc- Papadopoulou MV, Epperly MW, Shields DS, Bloomer WD tive metabolism of metronidazole and ronidazole by aero- (1992) Radiosensitization and hypoxic cell toxicity of NLA- bic liver microsomes. Mol Pharmacol 17:239–244 1 and NLA-2, two bioreductive compounds. Jpn J Cancer Peters KB, Brown JM (2002) Tirapazamine: a hypoxia-acti- Res 83:410–414 vated topoisomerase II poison. Cancer Res 62:5248–5253 Papadopoulou MV, Rosenzweig HS, Doddi M, Bloomer WD Peters KB, Wang H, Brown JM, Iliakis G (2001) Inhibition of (1994) 9-[3-)2-nitro-1-imidazolyl)propylamino]-1,2,3,4- DNA replication by tirapazamine. Cancer Res 61:5425–5431 Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 89

Phillips RM (1996) Bioreductive activation of a series of ana- DB, Fischer JJ (2000) Interim results of a randomized trial logues of 5-aziridinyl-3-hydroxymethyl-1-methyl-2-[1H- of mitomycin C as an adjunct to radical radiotherapy in the indole-4,7-dione]prop-beta-en-alpha-ol (EO9) by human treatment of locally advanced squamous-cell carcinoma of DT-diaphorase. Biochem Pharmacol 52:1711–1718 the cervix. Int J Cancer 90:206–223 Plumb JA, Gerritsen M, Workman P (1994) DT-diaphorase Rockwell S (1997) Oxygen delivery: implications for the biol- protects cells from the hypoxic cytotoxicity of indoloqui- ogy and therapy of solid tumors. Oncol Res 9:383–390 none EO9. Br J Cancer 70:1136–1143 Rockwell S, Kennedy KA (1979) Combination therapy with Powel J von, Roemeling R von, Gatzemeier U, Boyer M, Elisson radiation and mitomycin C: preliminary results with EMT6 LO, Clark P, Talbot S, Rey A, Butler TW, Hirsh V, Olver I, tumor cells in vitro and in vivo. Int J Radiat Oncol Biol Bergman B, Ayoub J, Ricahrdson G, Dunlop D, Arcenas A, Phys 5:1673–1676 Vescio R, Viallet J, Treat J (2000) Tirapazamine plus cispla- Rockwell S, Kennedy KA, Sartorelli AC (1982) Mitomycin C as tin versus cisplatin in advanced non-small-cell lung cancer: a prototype bioreductive alkylating agent: in vitro stud- a report of the international CATAPULT I study group. J ies of metabolism and cytotoxicity. Int J Radiat Oncol Biol Clin Oncol 18:1351–1359 Phys 8:753–755 Pritsos CA, Sartorelli AC (1986) Generation of reactive oxygen Rockwell S, Mate TP, Irvin CG, Nierenburg M (1986) Reactions radicals through bioactivation of mitomycin antibiotics. of tumors and normal tissues in mice to irradiation in the Cancer Res 46:3528–3532 presence and absence of a perfluorochemical emulsion. Int Raleigh SM, Wanogho E, Burke MD, McKeown SR, Patterson J Radiat Oncol Biol Phys 12:1315–1318 LH (1998) Involvement of human cytochromes P450 (CYP) Rockwell S, Keyes SR, Sartorelli AC (1988) Preclinical stud- in the reductive metabolism of AQ4N, a hypoxia activated ies of porfiromycin as adjunct to radiotherapy. Radiat Res di-N-oxide prodrug. Int J Radiat Oncol Biol 116:100–113 Phys 42:763–767 Rodriguez GI, Valdivieso M, Hoff DD von, Kraut M, Burris Raleigh SM, Wanogho E, Burke MD and Patterson LH (1999) HA, Eckardt JR, Lockwood G, Kennedy H, Roemeling R Rat cytochromes P450 (CYP) specifically contribute to the von (1996) A phase I/II trial of the combination of tirapa- reductive bioactivation of AQ4N, an alkylaminoanthraqui- zamine and cisplatin in patients with non-small cell lung none-di-N-oxide anticancer drug. Xenobiotica 29:1115– cancer (NSCLC). Proc Am Assoc Cancer Res 15:382 1122 Rofstad EK (2000) Microenvironment-induced cancer metas- Rauth AM, McClelland RA, Michaels HB, Battistella R (1984) tasis. Int J Radiat Biol:76 589–605 The oxygen dependence of the reduction of nitroimida- Ryan HE, Lo J, Johnson RS (1998) HIF-1 alpha is required for zoles in a radiolytic model system. Int J Radiat Oncol Biol solid tumor formation and embryonic vascularization. Phys 10:1323–1326 EMBO J 17:3005–3015 Reynolds TY, Rockwell S, Glazer PM (1996) Genetic instabil- Saarilahti K, Kajanti M, Atula T, Makitie A, Aaltonen L-M, ity induced by the tumor microenvironment. Cancer Res Kouri M, Mantyla M (2004) Biweekly escalated, accelerated 56:5754–5757 hyperfractionated radiotherapy with concomitant single- Riley RJ, Workman P (1992) Enzymology of the reduction of dose mitomycin C results in a high rate of local control in the potent benzotriazine-di-N-oxide hypoxic cell cytotoxin advanced laryngeal and hypopharyngeal cancer. Am J Clin SR 4233 (WIN 59075) by NAD(P)H: (quinone acceptor) Oncol 27:589–594 oxidoreductase (EC 1.6.99.2) purified from Walker 256 rat Schellens JHM, Planting AST, Van Acker BAC, Loos WJ, De tumour cells. Biochem Pharmacol 43:167–174 Boer-Denert M, Van der Burg MEL, Koier I, Krediet RT, Riley RJ, Hemingway SA, Graham MA, Workman P (1993) Stoter G, Verweij J (1994) Phase I and pharmacologic study Initial characterization of the major mouse cytochrome of the novel indoloquinone bioreductive alkylating cyto- P450 enzymes involved in the reductive metabolism of the toxic drug EO9. J Natl Cancer Inst 86:906–912 hypoxic cytotoxin 3-amino-1,2,4-benzotriazine-1,4-di-N- Semenza GL (2001) Hypoxia-inducible factor 1: oxygen oxide (tirapazamine, SR 4233, WIN 59075). Biochem Phar- homeostasis and disease pathophysiology. Trends Mol Med macol 45:1065–1077 7:345–350 Rischin D, Peters L, Hicks R, Hughes P, Fisher R, Hart R, Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat Sexton M, D’Costa I, Roemeling R von (2001) Phase I trial Rev Cancer 3:721–732 of concurrent tirapazamine, cisplatin, and radiotherapy in Sheldon PW, Fowler JF (1978) Radiosensitization by misonida- patients with advanced head and neck cancer. J Clin Oncol zole (Ro-07-0582) of fractionated X-rays in murine tumour. 19:535–542 Br J Cancer 37:243–245 Rischin D, Peters L, Fisher R, Macann A, Danham J, Poulsen Shibaji T, Nagao M, Ikeda N, Kanehiro H, Hisanaga M, Ko S, M, Jackson M, Kenny L, Penniment M, Corry J, Lamb D, Fukumoto A, Nakajima Y (2003) Prognostic significance of McClure B (2005) Tirapazamine, cisplatin, and radiation HIF-1 alpha overexpression in human pancreatic cancer. versus fluorouracil, cisplatin, and radiation in patients Anticancer Res 23:4721–4727 with locally advanced head and neck cancer: a randomized Shibamoto Y, Zhou L, Hatta H, Mayuko M, Nishimoto S (2000) Phase-II trial of the trans-Tasman radiation oncology A novel class of antitumor prodrug, 1-(2’-oxopropyl)-5- group (TROG 98.02). J Clin Oncol 23:79–87 fluorouracil (OFU001), that releases 5-fluorouracil upon Roberts PB, Anderson RF, Wilson WR (1987) Hypoxia-selec- hypoxic irradiation. Jpn J Cancer Res 91:433–438 tive radiosensitization of mammalian cells by nitracrine, Shibamoto Y, Zhou L, Hatta H, Mori M, Nishimoto S (2001) In an electron-affinic DNA intercalator. Int J Radiat Biol Relat vivo evaluation of a novel antitumor prodrug, 1-(2’-oxo- Stud Phys Chem Med 51:641–654 propyl)-5-fluorouracil (OFU001), which releases 5-fluo- Roberts KB, Urdaneta N, Vera R, Vera A, Gutierrez E, Aguilar Y, rouracil upon hypoxic irradiation. Int J Radiat Biol Phys Ott S, Medina I, Sempere P, Rockwell S, Sartorelli AC, Fischer 49:407–413 90 G. Ahn and J. M. Brown

Shibamoto Y, Tachi Y, Tanabe K, Hatta H, Nishimoto S (2004) In Tan C, Noronha RG de, Roecker AJ, Pyrzynska B, Khwaja F, vitro and in vivo evaluation of novel antitumor prodrugs of Zhang Z, Zhang H, Teng Q, Nicholson AC, Giannakakou 5-fluoro-2’-deoxyuridine activated by hypoxic irradiation. P, Zhou W, Olson JJ, Pereira MM, Nicolaou KC, Van Meir Int J Radiat Biol Phys 58:397–402 EG (2005) Identification of a novel small-molecule inhibi- Shibata T, Giaccia AJ, Brown JM (2002) Hypoxia-inducible reg- tor of the hypoxia-inducible factor 1 pathway. Cancer Res ulation of a prodrug-activating enzyme for tumor-specific 65:605–612 gene therapy. Neoplasia 4:40–48 Teicher BA, Rose CM (1984) Oxygen-carrying perfluorochemi- Shweiki D, Itin A, Soffer D, Keshet E (1992) Vascular endo- cal emulsion as an adjuvant to radiation therapy in mice. thelial growth factor induced by hypoxia may mediate Cancer Res 44:4285–4288 hypoxia-initiated angiogenesis. Nature 359:843–845 Teicher BA, Herman TS, Holden SA, Rudolph MB (1991) Siim BG, Van Zijl P, Brown JM (1996) Tirapazamine-induced Effect of oxygenation, pH and hyperthermia on RSU-1069 DNA damage measured using the comet assay correlates in vitro and in vivo with radiation in the FSaIIC murine with cytotoxicity towards hypoxic tumour cells in vitro. Br fibrosarcoma. Cancer Lett 59:109–117 J Cancer 73:952–960 Tercel M, Lee AE, Hogg A, Anderson RF, Lee HH, Siim BG, Siim BG, Denny WA, Wilson WR (1997) Nitro reduction as an Denny WA, Wilson WR (2001) Hypoxia-selective antitumor electronic switch for bioreductive drug activation. Oncol agents. 16. Nitroarylmethyl quaternary salts as bioreduc- Res 9:357–369 tive prodrugs of the alkylating agent mechlorethamine. J Simpson JR, Bauer M, Perez CA, Wasserman TH, Emami B, Med Chem 44:3511–3522 Doggett RL, Byhardt RW, Phillips TL, Mowry PA (1989) Theodoropoulos VE, Lazaris AC, Sofras F, Gerzelis I, Tsoukala Radiation therapy alone or combined with misonidazole V, Ghikonti I, Manikas K, Kastriotis I (2004) Hypoxia- in the treatment of locally advanced non-small cell lung inducible factor 1 alpha expression correlates with angio- cancer: report of an RTOG prospective randomized trial. genesis and unfavorable prognosis in bladder cancer. Eur Int J Radiat Oncol Biol Phys 16:1483–1491 Urol 46:200–208 Smith PJ, Blunt NJ, Desnoyers R, Giles Y, Patterson LH (1997a) Thomlinson RH, Gray LH (1955) The histological structure of DNA topoisomerase II-dependent cytotoxicity of alkyklam- some human lung cancers and the possible implications for inoanthraquinones and their N-oxides. Cancer Chemother radiotherapy. Br J Cancer 9:539–549 Pharmacol 39:455–461 Tomasz M, Lipman R, Chowdary D, Pawlak J, Verdine GL, Smith PJ, Desnoyers R, Blunt N, Giles Y, Patterson LH, Watson Nakanishi K (1987) Isolation and structure of a covalent JV (1997b) Flow cytometric analysis and confocal imaging cross-link adduct between mitomycin C and DNA. Science of the anticancer alkylaminoanthraquinones and their N- 235:1204–1208 oxides in intact human cells using 647-nm krypton laser Torre MH, Gambino D, Araujo J, Cerecetto H, Gonzalez M, excitation. Cytometry 27:43–53 Lavaggi ML, Azqueta A, Lopez De Cerain A, Vega AM, Smithen CE, Clarke ED, Dale JA, Jacobs RS, Wardman Abram U, Costa-Filho AJ (2005) Novel Cu(II) quinoxaline P, Watts ME, Woodcock M (1980) Novel (nitro-1- N1, N4-dioxide complexes as selective hypoxic cytotoxins. imidazolyl)alkanolamines as potential radiosensitisers Eur J Med Chem 40:473–480 with improved therapeutic properties. In: Brady LW (ed) Treat J, Johnson E, Langer C, Belani C, Haynes B, Greenberg R, Radiation sensitizers: their use in the clinical management Rodriguez R, Drobins P, Miller W Jr, Meehan L, McKeon A, of cancer. Masson, New York, pp 22–32 Devin J, Roemeling R von, Viallen R (1998) Tirapazamine Song CW, Park H, Griffin R (2001) Improvement of tumor oxy- with cisplatin in patients with advanced non-small-cell genation by mild hyperthermia. Radiat Res 155:515–528 lung cancer: a phase II study. J Clin Oncol 16:3524–3527 Squillace DP, Ruben SL, Reid JM, Ames MM (2000) HPLC Trotter MJ, Chaplin DJ, Olive PL (1989) Use of a carbocya- analysis and murine pharmacokinetics of the cytotoxic nine dye as a marker of functional vasculature in murine bioreductive agent NLCQ-1 (NSC 709257). Proc Am Assoc tumours. Br J Cancer 59:706–709 Cancer Res 41:706 Unruh A, Ressel A, Mohamed HG, Johnson RS, Nadrowitz R, Stoeltzing O, McCarty MF, Wey JS, Fan F, Liu W, Belcheva Richter E, Katschinski DM, Wenger RH (2003) The hypoxia- A, Bucana CD, Semenza GL, Ellis LM (2004) Role of inducible factor-1D is a negative factor for tumor therapy. hypoxia-inducible factor 1D in gastric cancer cell growth, Oncogene 22:3213–3220 angiogenesis, and vessel maturation. J Natl Cancer Inst Urtasun RC, Band P, Chapman JD, Feldstein ML, Mielke B, 96:946–956 Fryer C (1976) Radiation and high dose metronidazole in Stratford IJ (1982) Mechanisms of hypoxic cell radiosensitiz- supratentorial glioblastomas. N Engl J Med 294:1364–1367 ers and the development of new sensitizers. Int J Radiat Urtasun RC, Palmer M, Kinney B, Belch A, Hewit J, Hanson J Oncol Biol Phys 8:391–398 (1998) Intervention with the hypoxic tumor cell sensitizer Stratford IJ, Walling JM, Silver ARJ (1986) The differential etanidazole in the combined modality treatment of limited toxicity of RSU 1069: cell survival studies indicating inter- stage small-cell lung cancer. A one-institution study. Int J action with DNA as possible mode of action. Br J Cancer Radiat Oncol Biol Phys 40:337–342 53:339–344 Van den Bogaert W, Van der Schueren E, Horiot J-C, De Vil- Sun X, Liu M, Wei Y, Liu F, Zhi X, Xu R, Krissansen GW (2005) hena M, Schraub S, Svoboda V, Arcangeli G, De Pauw M, Overexpression of von Hippel-Lindau tumor suppres- Van Glabbeke M (1995) The EORTC randomized trial on sor protein and antisense HIF-1alpha eradicates glioma. three fractions per day and misonidazole (trial no. 22811) Cancer Gene Ther in advanced head and neck cancer: long-term results and Sutherland RM (1974) Selective chemotherapy of noncycling side effects. Radiother Oncol 35:91–99 cells in an in vitro tumor model. Cancer Res 34:3501– Varghese AJ, Gulyas S, Mohindra JK (1976) Hypoxia-depend- 3503 ent reduction of 1-(2-nitro-1-imidazolyl)3-methoxy-2-pro- Combinations of Hypoxia-Targeting Compounds and Radiation-Activated Prodrugs with Ionizing Radiation 91

panol by Chinese hamster ovary cells and KHT tumor cells Whitmore GF, Gulyas S (1986) Studies on the toxicity of RSU- in vitro and in vivo. Cancer Res 36:3761–3765 1069. Int J Radiat Oncol Biol Phys 12:1219–1222 Volpato M, Seargent J, Loadman PM, Phillips RM (2005) For- Widder J, Dobrowsky W, Schmid R, Pokrajac B, Selzer E, Potter mation of DNA interstrand cross-links as a marker of mito- R (2004) Hyperfractionated accelerated radiochemother- mycin C bioreductive activation and chemosensitivity. Eur apy (HFA-RCT) with mitomycin C for advanced head and J Cancer 41:1331–1338 neck cancer. Radiother Oncol 73:173–177 Walton MI, Workman P (1987) Nitroimidazole bioreductive Williams KJ, Telfer BA, Xenaki D, Sheridan MR, Desbaillets I, metabolism. Quantitation and characterisation of mouse Peters HJW, Honess D, Harris AL, Dachs GU, Van der Kogel tissue benznidazole nitroreductases in vivo and in vitro. A, Stratford IJ (2005) Enhanced response to radiotherapy Biochem Pharmacol 36 887–896 in tumours deficient in the function of hypoxia-inducible Walton MI, Workman P (1993) Pharmacokinetics and biore- factor-1. Radiother Oncol 75:89–98 ductive metabolism of the novel benzotriazine di-N- Wilson WR, Denny WA, Stewart GM, Fenn A, Probert JC (1986) oxide hypoxic cell cytotoxin tirapazamine (WIN 59075; Reductive metabolism and hypoxia-selective toxicity of SR 4233; NSC 130181) in mice. J Pharmacol Exp Ther nitracrine. Int J Radiat Oncol Biol Phys 12:1235–1238 265:938–947 Wilson WR, Van Zijl P, Denny WA (1992) Bis-bioreductive Walton MI, Wolf CR, Workman P (1992) The role of cyto- agents as hypoxia-selective cytotoxins: nitracrine N-oxide. chrome P450 and cytochrome P450 reductase in the reduc- Int J Radiat Oncol Biol Phys 22:693–696 tive bioactivation of the novel benzotriazine di-N-oxide Wilson WR, Moselen JW, Cliffe S, Denny WA, Ware DC (1994) hypoxic cytotoxin 3-amino-1,2,4-benzotriazine-1,4-diox- Exploiting tumor hypoxia through bioreductive release of ide (SR 4233, WIN 59075) by mouse liver. Biochem Phar- diffusible cytotoxins: the cobalt(III)- com- macol 44:251–259 plex SN24771. Int J Radiat Oncol Biol Phys 29:323–327 Wang J, Biedermann KA, Brown JM (1992) Repair of DNA Wilson WR, Denny WA, Pullen SM, Thompson KM, Li AE, and chromosome breaks in cells exposed to SR 4233 under Patterson LH, Lee HH (1996) Tertiary amine N-oxides as hypoxia or to ionizing radiation. Cancer Res 52:4473– bioreductive drugs: DACA N-oxide, nitracrine N-oxide and 4477 AQ4N. Br J Cancer 27 (Suppl):S43–S47 Wang J, Biedermann KA, Wolf CR, Brown JM (1993) Metabo- Wilson WR, Tercel M, Anderson RF, Denny WA (1998) Radia- lism of the bioreductive cytotoxin SR 4233 by tumour cells: tion-activated prodrugs as hypoxia-selective cytotoxins: enzymatic studies. Br J Cancer 67:321–325 model studies with nitroarylmetyl quaternary salts. Anti- Wardman P (2001) Electron transfer and oxidative stress cancer Drug Des 13 663–685 as key factors in the design of drugs selectively active in Wilson WR, Pullen SM, Hogg A, Helsby NA, Hicks KO, Denny hypoxia. Curr Med Chem 8:739–761 WA (2002) Quantitation of bystander effects in nitrore- Wardman P, Clarke ED (1976) Oxygen inhibition of nitrore- ductase suicide gene therapy using three-dimensional cell ductase: electron transfer from nitro radical-anions to cultures. Cancer Res 62:1425–1432 oxygen. Biochem Biophys Res Commun 69:942–949 Wilson WR, Edmunds SJ, Valentines S (2005) Mechanism of Wardman P, Dennis MF, Everett SA, Patel KB, Stratford MR, action and antitumour activity of PR-104, a dinitroben- Tracy M (1995) Radicals from one-electron reduction of zamide mustard pre-prodrug that is activated selectively nitro compounds, aromatic N-oxides and quinones: the under hypoxia. National Cancer Research Institute Confer- kinetic basis of hypoxia-selective, bioreductive drugs. Bio- ence, Birmingham, UK chem Soc Symp 61:171–194 Workman P, Stratford IJ (1993) The experimental development Ware DC, Palmer BD, Wilson WR, Denny WA (1993) Hypoxia- of bioreductive drugs and their role in cancer therapy. selective antitumor agents. 7. Metal complexes of aliphatic Cancer Met Rev 12:73–82 mustards as a new class of hypoxia-selective cytotoxins. Wouters BG, Brown JM (1997) Cells at intermediate oxygen Synthesis and evaluation of cobalt(III) complexes of biden- levels can be more important than the “hypoxic fraction” in tate mustards. J Med Chem 36:1839–1846 determining tumor response to fractionated radiotherapy. Watson ER, Halnan KE, Dische S, Saunders MI, Cade IS, Mcewen Radiat Res 147:541–550 JB, Wiernik G, Perrins DJD, Sutherland I (1978) Hyperbaric Zeman EM, Brown JM, Lemmon MJ, Hirst VK, Lee WW (1986) oxygen and radiotherapy: a Medical Research Council trial SR-4233: a new bioreductive agent with high selective tox- in carcinoma of the cervix. Br J Radiol 51:879–887 icity for hypoxic mammalian cells. Int J Radiat Oncol Biol Weissberg JB, Son YH, Papac RJ, Sasaki CT, Fischer DB, Law- Phys 12:1239–1242 rence R, Rockwell S, Sartorelli AC, Fischer JJ (1989) Ran- Zhong XS, Zheng JZ, Reed E, Jiang B-H (2004) SU5416 inhib- domized of mitomycin C as an adjunct to ited VEGF and HIF-1alpha expression through the PI3K/ radiotherapy in head and neck cancer. Int J Radiat Oncol AKT/p70S6K1 signaling pathway. Biochem Biophys Res Biol Phys 17:3–9 Commun 324:471–480