Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 3/2016

Open Access 01.03.2016 | Review Article

Targeting the hypoxic fraction of tumours using hypoxia-activated prodrugs

verfasst von: Roger M. Phillips

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 3/2016

Abstract

The presence of a microenvironment within most tumours containing regions of low oxygen tension or hypoxia has profound biological and therapeutic implications. Tumour hypoxia is known to promote the development of an aggressive phenotype, resistance to both chemotherapy and radiotherapy and is strongly associated with poor clinical outcome. Paradoxically, it is recognised as a high-priority target and one of the therapeutic strategies designed to eradicate hypoxic cells in tumours is a group of compounds known collectively as hypoxia-activated prodrugs (HAPs) or bioreductive drugs. These drugs are inactive prodrugs that require enzymatic activation (typically by 1 or 2 electron oxidoreductases) to generate cytotoxic species with selectivity for hypoxic cells being determined by (1) the ability of oxygen to either reverse or inhibit the activation process and (2) the presence of elevated expression of oxidoreductases in tumours. The concepts underpinning HAP development were established over 40 years ago and have been refined over the years to produce a new generation of HAPs that are under preclinical and clinical development. The purpose of this article is to describe current progress in the development of HAPs focusing on the mechanisms of action, preclinical properties and clinical progress of leading examples.

Introduction

One of the characteristic features of solid tumour biology is the presence of a poor and inadequate blood supply [1]. This leads to the establishment of microenvironments that are characterised by gradients of oxygen tension, nutrients, extracellular pH, catabolites and reduced cell proliferation, all of which vary as a function of distance from a supporting blood vessel (Fig. 1). These microenvironments can be chronic in nature caused by poor blood supply (diffusion limited) or acute caused by the temporal opening and closing of blood vessels (perfusion limited). Hypoxia in tumours has been the focus of intense research for over 60 years, and both diffusion-limited hypoxia and perfusion-limited hypoxia are established features of solid tumours [2]. A third mechanism to explain the induction of hypoxia in tumours has been described, namely longitudinal arteriole gradients whereby oxygen-rich inflowing blood vessels branch and coalesce to form poorly oxygenated outflowing blood [3]. In this model, hypoxia would be formed along the axis of the vessel over a multimillimetre range, which contrasts with the submillimetre distances typically associated with perfusion- and diffusion-limited hypoxia. The origins of tumour hypoxia are therefore linked to the abnormal vascular supply that develops within tumours, and there is a substantial body of evidence demonstrating that hypoxia is a common feature of most if not all-solid tumours.
The presence of hypoxia in tumours has significant biological and therapeutic implications. Biologically, hypoxia is implicated in promoting resistance to apoptosis [4], suppression of DNA repair pathways and promotion of genomic instability [5] increased invasion and metastasis [6], promotion of angiogenesis [7], modulation of tyrosine kinase-mediated cell signalling pathways [8], evasion from immune surveillance [9], induction of autophagy [10], hypoxia-driven changes in central metabolic pathways [11], global changes in the metabolome [12], production of L-2-hydroxyglutarate leading to altered histone methylation [13], metabolic adaptation to hypoxia-induced reductive stress [14] and the provision of a niche where cancer stem cells reside [15]. The plethora of effects on tumour biology is mediated largely by hypoxia-inducible factors (HIF) [16] although HIF1-independent hypoxia responses have also been described [17]. In terms of therapeutic implications, the seminal work conducted by Gray in the 1950s [18] provided the first evidence that hypoxia is an underlying cause of resistance to radiotherapy. Since then, hypoxia has been strongly implicated in resistance to several cytotoxic chemotherapy drugs and targeted therapeutics [19, 20]. Multiple mechanisms contribute to hypoxia-induced drug resistance, but as pointed out by Wilson and Hay [21], the generalisation that hypoxia causes resistance to all cytotoxic drugs must be viewed with caution as some drugs are effective under hypoxic conditions. This note of caution should also be extended to include targeted therapeutics following the demonstration that some (such as dasatinib) are preferentially active against cell lines in vitro under hypoxic conditions [22].
Whilst the extent and severity of hypoxia in tumours varies between tumour types and within individual tumours, the combined biological and therapeutic implications of hypoxia have a significant bearing on prognosis. There is now an extensive body of evidence demonstrating that hypoxia can adversely affect clinical outcome [23, 24] and this makes hypoxia a high-priority therapeutic target. The importance of hypoxia as a target has been recognised for many years, but the translation of preclinical strategies designed to target hypoxic cells into mainstream clinical use has remained stubbornly difficult to achieve. Two main approaches are being used to eradicate hypoxic cells (1) the use of ‘bioreductive drugs’ or ‘hypoxia-activated prodrugs (HAPs)’ and (2) molecularly targeted drugs aimed at exploiting biochemical responses to hypoxia, particularly HIF pathways. The current status of HIF-targeted strategies is beyond the scope of this article which focuses on HAPs, their mechanism of action and recent progress in the preclinical and clinical evaluation of leading compounds in this class of drugs. This article also describes novel approaches where HAP-based approaches are being used to improve the selectivity of targeted therapeutics.

Hypoxia-activated prodrugs (HAPs): general principles

The concept of hypoxia-activated prodrugs arose largely from the seminal work on quinone-based derivatives of Mitomycin C by Alan Sartorelli in the early 1970s [25]. Initially, these early studies focused primarily on enzyme-activated prodrugs in a process called bioreductive activation under aerobic conditions, but this concept was extended to include hypoxia following the demonstration that Mitomycin C preferentially killed hypoxic cells in vitro [26]. Over recent years, the term HAP has become established, but HAPs and bioreductive drugs are terms that are often used interchangeably. The principles underpinning the development of HAPs have been refined over the years, and the ‘ideal HAP’ should possess the following properties (1) ability to penetrate from a blood vessel to hypoxic cells within its pharmacokinetic lifespan; (2) preferential activation oxygen conditions that are low enough to prevent activation in normal tissues; (3) the reduced product should have the ability to kill non-proliferating cells typically found within the hypoxic fraction of tumours; and (4) the reduced product should have the ability to diffuse back into the proliferating aerobic fraction and exert a ‘bystander effect’ (Fig. 1).
Five different chemical entities have been shown to be capable of selectively targeting hypoxic cells, and these include nitro (hetero)-cyclic compounds, aromatic N-oxides, aliphatic N-oxides, quinone and transition metal complexes [27]. Whilst these are chemically distinct classes of compounds, a modular concept for the design of HAPs has been described with the three main components being (1) a trigger, (2) a linker and (3) an effector [28]. The effector is the cytotoxic component that is capable of killing cells within the hypoxic microenvironment, and these have typically been potent DNA-interactive agents. The purpose of the linker is to deactivate the effector, whilst the trigger group is the critical group that determines prodrug activation and hypoxia selectivity. Numerous trigger groups have been characterised, and these have to be enzymatically reduced (primarily by oxidoreductases) in order to release or activate the effector [28]. Both one- and two-electron oxidoreductases can catalyse the reduction of the prodrug, and selectivity is determined by the ability of oxygen to reverse the activation process and/or the overexpression of oxidoreductases in tumour tissue (Fig. 1). In general terms, one-electron reduction generates a prodrug radical species that can be back-oxidised in the presence of oxygen to generate the parental prodrug and reactive oxygen species. Host defence mechanisms can detoxify these radical species, thereby reducing toxic effects in oxygenated tissue, but in the absence of oxygen, the prodrug radical species undergoes further reduction/disproportionation or fragmentation reactions to generate products that are cytotoxic [21] Two-electron reduction in contrast bypasses the oxygen-sensitive prodrug radical intermediate, and activation of the prodrug is typically oxygen insensitive. In this case, selectivity is largely determined by the presence of elevated levels of enzyme in tumour tissue [29]. The mechanisms governing hypoxia-activated prodrug activation are summarised in Fig. 1, and important exceptions to these generalised mechanisms are identified here and in the main body of text below. It is also important to state that hypoxia-directed therapeutic agents are unlikely to demonstrate single-agent activity and should be used in combination with radiotherapy and/or chemotherapy that targets aerobic cells. Furthermore, in order to become effective components of combination therapies, HAPs need to be sufficiently safe with minimal side effects.
Whilst the concepts underpinning bioreductive drug and HAP activation are elegant and simple, success in this field has been very difficult to achieve and the stark reality is that despite over 40 years of pre-clinical and clinical research, no HAP has so far been approved for use in humans. Despite this lack of success, important lessons have been learnt from preclinical and clinical studies that have helped shape the design and evaluation of new HAPs. There is currently considerable optimism in the field, and in the following sections, recent progress in the pre-clinical and clinical evaluation of a series of selected HAPs (Fig. 2) is presented together with some emerging strategies for the next generation of HAPs.

TH-302 (Evofosfamide)

TH-302 is a 2-nitroimidazole HAP of bromo-isophosphoramide, the synthesis and initial preclinical evaluation of which was published in 2008 [30]. It was initially developed by Threshold Pharmaceuticals, and TH-302 is currently being developed in partnership with Merck. Its preclinical pharmacology has been described in detail elsewhere [31]. Its mechanism of action is presented in Fig. 3, and key features of its pharmacology include (1) reduction by one-electron oxidoreductases or radiolytic reduction leading to fragmentation and release of the potent DNA alkylating agent bromo-isophosphoramide mustard; (2) hypoxia-dependent induction of γH2AX, DNA cross-linking and cell cycle arrest; (3) broad activity against a range of cell lines with hypoxic cytotoxicity ratios (HCR) ranging from 11 to 600; (4) cells deficient in homology-dependent DNA repair, BRCA1, BRCA2 and FANCA exhibited marked sensitivity to TH-302 under hypoxia; (5) enhanced potency against H460 multicell spheroids compared to monolayer cultures; (6) exhibited a bystander effect in in vitro multicell layer models and in vivo; (7) broad anti-tumour activity across a panel of human tumour xenografts with clear evidence of selective eradication of hypoxic cells and neighbouring cells via the bystander effect; and (8) good pharmacokinetic and safety profiles in mice, rats, dogs and monkeys. More recent studies have confirmed that TH-302 targets hypoxic cells and potentiates the activity of doxorubicin and docetaxel in human tumour xenografts [32] and its activity in vivo can be enhanced by the induction of transient hypoxia [33]. As part of a combination regimen with cytotoxic drugs, targeted therapeutics or radiotherapy, TH302 has been shown to enhance anti-tumour activity and reduce the ability of hypoxic cells to repopulate tumours following re-oxygenation [3440]. Activity is not confined to solid tumours but extends to acute myeloid leukaemia where TH-302 was effective against hypoxic cells that reside within the bone marrow microenvironment [41].
TH-302 is undergoing clinical trial, and phase I studies were published in 2011 [42, 43]. As a single agent administered intravenously, TH-302 was well tolerated. At maximum tolerated dose, adverse events included nausea, skin rash, fatigue and vomiting, and depending on the regimen used, dose-limiting toxicities were grade 3 skin and mucosal toxicities or grade 3 fatigue and vaginitis/proctitis [43]. Two partial responses in patients with metastatic small cell lung cancer and melanoma were observed with stable disease seen in 27 out of 57 patients [43]. A phase I study of TH-302 in combination with doxorubicin demonstrated that despite the haematologic toxicity of doxorubicin increasing when combined with TH-302, toxicities were manageable and partial responses were observed in 5 out of 15 patients with advanced soft tissue sarcoma [42]. Furthermore, one patient with advanced melanoma participating in the phase I studies had complete resolution of Cullen’s sign together with extracranial response in lung, liver and lymph node metastasis [44]. Promising clinical activity has also been reported in phase II trials involving TH-302 in combination with doxorubicin in advanced soft tissue sarcoma [45] and in combination with gemcitabine in patients with advanced pancreatic cancer [46]. TH-302 is undergoing further phase II trials against non-small cell lung cancer (with pemetrexed, NCT02093962) and advanced melanoma (NCT01864538), and phase III clinical trials against soft tissue sarcoma (NCT01440088) and pancreatic cancer (NCT01746979, MAESTRO study) are ongoing. The results of phase III studies are eagerly anticipated.

EO9 (Apaziquone)

EO9 is an indolequinone derivative of Mitomycin C that was originally synthesised in 1987 at the University of Amsterdam [47]. It has a chequered history, details of which have been reviewed recently [48]. Its mechanism of action (Fig. 4) involves reduction by one- and/or two-electron oxidoreductases to generate DNA-damaging species in both aerobic and hypoxic conditions. The two-electron reductase NAD(P)H:Quinone oxidoreductase 1 (NQO1 or DT-diaphorase) plays a central role in the bioreductive activation process and largely determines its ability to target aerobic or hypoxic cells [29]. EO9 has the ability to function as a classical HAP but only in cell lines that have low or no NQO1 [49, 50]. Under these conditions, one-electron reduction by enzymes such as cytochrome P450 reductase generates the semi-quinone radical which can redox cycle back to the parent compound in the presence of oxygen. HCR values in excess of 100 have been reported in a number of cell lines but particularly marked hypoxia selectivity occurs in cell lines that harbour the C609T single-nucleotide polymorphism that is devoid of NQO1 activity [49, 51]. In cell lines with high NQO1, however, the oxygen-insensitive two-electron reduction pathway dominates and HCR values close to 1 are typically reported [50]. In these cell lines, a good correlation between NQO1 activity and chemosensitivity under aerobic conditions exists [50]. Preclinical studies therefore demonstrated that EO9 has the ability to target both the aerobic fraction of NQO1-rich tumours (where selectivity is determined by elevated levels of NQO1 in tumours) and the hypoxic fraction of NQO1-deficient tumours [29].
Based on these and other favourable preclinical properties [52], EO9 was selected for clinical evaluation under the auspices of the New Drug Development Office in Amsterdam with phase I studies reporting partial responses in two patients with carcinomas of unknown origin and one in bile duct cancer [53, 54]. The results of phase II studies were, however, disappointing with no partial or complete responses observed. These studies concluded that EO9 had no clinical activity against NSCLC, pancreatic, breast, colorectal and gastric cancers [55, 56]. Several possible explanations were put forward to explain the poor results of these studies including the fact that EO9 was not evaluated as a classical HAP as it was only tested as a single agent. Furthermore, tumour enzymology and the presence of hypoxia in patient’s tumours were not incorporated into the design of the trials [57]. Whilst these issues represent important deficiencies in clinical trial design, it was argued that at least some patients would have had the right ‘biochemical machinery’ to activate EO9 and other explanations for why EO9 failed must exist.
Research focused on the issue of impaired drug delivery to tumours as a possible explanation. Whilst the factors that determine drug delivery to tumours are complex, systemic pharmacokinetic profiles and the ability to extravasate and penetrate through several layers of tumour cells to reach the hypoxic fraction are two key parameters [58]. Phase I studies had already established that EO9 was rapidly cleared from the systemic circulation following intravenous administration with half-lives of less than 20 min [54]. This combined with experimental evidence demonstrating that EO9 does not rapidly penetrate multicell layers in vitro suggested that EO9 will not penetrate more than a few cell layers from a blood vessel within its pharmacokinetic lifespan [59]. One method pursued to tackle this problem was to develop analogues of EO9 with better drug delivery properties, but an alternative approach designed to use EO9’s bad properties to our advantage was adopted. In the case of superficial transitional cell carcinoma of the bladder, chemotherapy is administered directly into the bladder by a catheter (intravesical administration). Intravesical administration of EO9 into the bladder would (1) circumvent the drug delivery problem observed following intravenous administration; (2) retention within the bladder for up to 1 h would extend the time EO9 was in contact with the tumour and enhance penetration; and (3) any drug that reached the systemic circulation would be rapidly cleared, thereby reducing the risk of systemic toxicity.
Following the demonstration that superficial transitional cell carcinoma of the bladder possessed the correct biochemical machinery required to activate EO9 [60], a new clinical trial was developed. Spectrum Pharmaceuticals sponsored the phase I study, and EO9 was administered directly into the bladder (intravesical administration) once a week for 6 weeks followed by assessment of anti-tumour efficacy 2 weeks after the final instillation. Prior to the administration of EO9, patients with multiple tumour lesions had all but one tumour surgically removed with the remaining tumour left to serve as a ‘marker lesion’ for assessing response. Complete response was defined as total ablation of the marker lesion, and eight complete responses were obtained out of a total of 12 patients entered into the study [61]. Using an identical trial design, similar complete response rates (30 out of 45 patients) were reported in phase II studies [62], and recurrence-free rates at 2 years were good in comparison with other marker lesion studies [63, 64]. Following the demonstration that a single intravesical administration of EO9 within 24 h of transurethral resection was well tolerated with a good safety profile [65], two phase III trials commenced using this new administration schedule (NCT00598806 and NCT00461591). In April 2012, Spectrum Pharmaceuticals announced that the results of these two trials did not meet their primary endpoint of a statistically significant difference in the rate of tumour recurrence at 2 years, but analysis of the pooled data from both studies showed a statistically significant effect in favour of EO9. A further phase III study using a multi-instillation schedule (once a week for 6 weeks) has been planned (NCT01410565).

AQ4N (Banoxantrone)

AQ4N is an aliphatic N-oxide that is metabolised by cytochrome P450 (CYP) isozymes and inducible nitric oxide synthase (iNOS) to AQ4, a potent inhibitor of topoisomerase II [66, 67]. Its mechanism of action is summarised in Fig. 5 and involves an initial two-electron reduction step to the mono-N-oxide (AQ4M) followed by a further two-electron reduction to generate AQ4. Selectivity for hypoxic cells occurs because oxygen outcompetes AQ4N for the haem-centred active site of CYPs and oxygen therefore effectively inhibits the reduction in AQ4N [68]. Activation not only occurs in tumour cells but also in hypoxic tumour-associated macrophages where the induction of iNOS under hypoxic conditions led to reduction in AQ4N and killing of tumour cells via a bystander effect [69]. One of the key features of its mechanism of action is that AQ4 is a stable reduction product that strongly binds non-covalently to the DNA of hypoxic cells. Following the eradication of aerobic cells by radiotherapy and/or chemotherapy, hypoxic cells undergo a period of re-oxygenation and can repopulate the tumour. AQ4 inhibits the topoisomerase activity of hypoxic cells as they attempt to re-enter the cell cycle during these periods of re-oxygenation, and in preclinical models in vivo, AQ4N in combination with radiotherapy substantially enhances anti-tumour efficacy [70, 71]. Similar effects were seen with combinations of AQ4N and thiotepa, cyclophosphamide, cisplatin and the structurally related drug mitoxantrone [7173]. More recent studies have demonstrated that the use of AQ4N following androgen deprivation therapy using bicalutamide (which induces hypoxia in tumours) significantly improves treatment outcome in prostate cancer xenografts [74]. Similarly, recent studies have also demonstrated that AQ4N has anti-metastatic properties by targeting hypoxic lesions in the lymph nodes and lymphatics in xenograft models [75]. With regard to drug delivery to the hypoxic fraction, studies in tumour models in vivo demonstrate that AQ4N has the ability to penetrate into hypoxic regions of tumour [73, 76]. Taken together, the preclinical profile of AQ4N clearly illustrates its potential as a HAP that is capable of tackling the key issue of tumour repopulation by hypoxic cells following the eradication of the aerobic fraction by conventional radio/chemotherapy. In addition to its HAP properties, AQ4N has also been shown to have anti-angiogenic properties under aerobic conditions, selectively targeting endothelial cells at low doses, inhibiting migration, tube formation, aortic ring vessel sprouting and invasion of proliferating endothelial cells [77]. Whilst the mechanism of action responsible for its anti-angiogenic properties is not fully understood (significant disruption of microtubule networks was observed), this mechanism could potentially contribute to the anti-tumour activity of AQ4N.
AQ4N has undergone clinical evaluation, and three phase I studies have been reported, two of which have evaluated AQ4N as a single agent [78, 79] and one in combination with radiotherapy [80]. As a single agent, AQ4N was well tolerated up to a maximum tolerated dose of 768 mg/m2 (administered intravenously as a 30-min infusion on days 1, 8 and 15 of a 28-day cycle) with the most common adverse events being fatigue, diarrhoea, nausea, vomiting, anorexia and blue discolouration of skin and body fluids [79]. The pharmacokinetic profile of AQ4N was dose dependent with low levels of AQ4M, and no AQ4 detected in the systemic circulation. Three patients had stable disease, two with bronchoalveolar lung cancer and ovarian cancer and the third with collecting duct renal cancer had prolonged stable disease for 25 months [79]. In a phase I proof-of-principle pharmacodynamics study, AQ4N at a dose of 200 mg/m2 (single dose administered intravenously using a 30-min infusion) was administered 12–36 h before multiple samples of tumour and normal tissue were surgically removed from each patient. AQ4N and its metabolites were analysed by LC/MS, the distribution of AQ4 relative to blood vessels determined by confocal microscopy and the relationship between AQ4 levels and the expression of the endogenous hypoxia marker Glut-1 determined by immunohistochemistry [78]. This study demonstrated that AQ4N was activated selectively in hypoxic regions of solid tumours and the levels of AQ4 detected exceeded those required for activity in animal models. In addition, high levels of AQ4 were detected in glioblastoma multiforme, indicating that AQ4N effectively crossed the blood–brain barrier [78]. In combination with radiotherapy, AQ4N was well tolerated up to 447 mg/m2 administered intravenously with no dose-limiting toxicity and tumour AQ4 concentrations also exceeded levels required for activity in preclinical models. Of the eighteen patients that were assessable for response, one had a partial response, two had >50 % tumour volume reduction and nine patients had stable disease [80]. Whether these responses were due to AQ4N or radiotherapy alone was not possible to determine but the results of this study illustrates the potential value of combination studies of AQ4N with radiotherapy. Regrettably, the clinical development of AQ4N has stalled, but new analogues of AQ4N are under development by OncoTherics.

PR-104

PR-104 is a nitroaromatic compound that is a water-soluble phosphate ester ‘pre-prodrug’ of PR-104A, originally developed by the University of Auckland [81]. Its mechanism of action has been described in detail elsewhere [27] and is summarised in Fig. 6. Briefly, PR-104 undergoes rapid hydrolysis by systemic phosphatases to generate PR-104A that is metabolised by one- and/or two-electron oxidoreductases to the para-hydroxylamine PR-104H and para-amine PR-104M metabolites resulting in interstrand DNA cross-linking. Various oxidoreductases have been shown to catalyse the reduction in PR-104A including cytochrome P450 reductase [82], FAD-dependent oxidoreductase domain containing 2 (FOXRED2 [83]) and aldo–ketoreductase 1C3 (AKR1C3) [84]. PR-104 can target both hypoxic and aerobic cells with one-electron reduction pathways catalysed by cytochrome P450 reductase accounting for the majority of activity under hypoxia [85]. Reduction by AKR1C3 (oxygen-insensitive two-electron reduction process) accounts for activity under aerobic conditions [84]. Hypoxia selectivity in vitro is governed by the fact that the product of one-electron reduction undergoes redox cycling back to the parent compound and hypoxia together with reductase activity and repair of DNA interstrand cross-links are key variables that determine response to PR-104 [86]. More recent studies have also demonstrated that PR-104 is particularly active against hypoxic regions of triple-negative breast cancers that have dysfunctional homologous recombination repair pathways [87].
PR-104 has activity against a range of in vivo preclinical models, and its properties mean it can target hypoxic tumours and/or the aerobic fraction of tumours expressing AKR1C3. Against T cell acute lymphoblastic leukaemia xenografts, single-agent PR-104 treatment proved more efficacious compared to a combination of vincristine, dexamethasone and l-asparaginase and activity correlated with AKR1C3 expression [88]. This study also concludes that AKR1C3 expression could be used as a biomarker to select patients most likely to benefit from PR-104 treatment in future clinical trials. Similar studies reported complete responses in acute lymphoblastic leukaemia models and objective responses in other solid tumours, but in contrast, tumour response did not correlate with AKR1C3 levels [89]. Other studies have demonstrated that PR-104 could be used to eradicate acute lymphoblastic leukaemia cells residing in hypoxic niches in the bone marrow [90]. Against solid tumours, experimental and in silico modelling demonstrate that PR104/PR104A is able to penetrate into severely hypoxic regions of tumours where it is preferentially metabolised to cytotoxic metabolites [91]. A combination of experimental and modelling techniques have also demonstrated that PR-104A can exert a bystander effect that is predicted to contribute significantly to the anti-tumour efficacy of PR-104 [92]. PR-104 has demonstrated anti-tumour activity as a single agent against a range of solid tumour xenografts and greater than additive effects have been reported when PR-104 is used in combination with chemotherapy agents such as gemcitabine, docetaxel and sorafenib [81, 93] and/or radiotherapy [87]. The use of pharmacological approaches to induce tumour hypoxia has also been shown to potentiate the activity of PR-104 [94].
PR-104 is undergoing clinical trials, and several phase I clinical trials have been completed. As a single agent, a maximum tolerated dose of 1100 mg/m2 was reported following a one dose every 21-day schedule [95] and 675 mg/m2 when given on a one dose per week for 3-week schedule [96]. In both studies, PR-104 was administered by a 1-h intravenous infusion. Dose-limiting toxicities included fatigue, febrile neutropenia and infection following a once a week, every 21-day schedule and thrombocytopenia and to a lesser extent neutropenia using the more intensive schedule [95, 96]. No objective responses were reported in these studies despite the fact that PR-104A plasma AUC values exceeded the levels required for activity in preclinical models [95]. In combination with either gemcitabine or docetaxel, severe dose-limiting myelotoxicity occurred, the impact of which was reduced by prophylactic G-CSF in the case of docetaxel [97]. A combination of PR-104 and sorafenib in advanced hepatocellular carcinoma was also poorly tolerated with significant thrombocytopenia and neutropenia reported [98]. PR-104A undergoes glucuronidation [99], and it was suggested that reduced clearance due to compromised glucuronidation in patients with advanced hepatocellular carcinoma of PR-104A was partly responsible for the toxicity observed [98]. Recent studies in mice that do not significantly glucuronidate PR-104A confirm that the development of analogues of PR104 that are not readily glucuronidated may be able to exploit elevated AKR1C3 and/or hypoxia in hepatocellular carcinoma in humans [93]. Based on strong preclinical data, a phase I/II study in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL) has demonstrated clinical activity in 10 out of 31 patients with AML and 2 out of 10 patients with ALL [100]. PR-104 treatment also decreased the number of hypoxic cells in the bone marrow. These positive results indicate that PR-104 is able to exploit the hypoxic niche in acute leukaemias and further clinical evaluation in this setting is warranted.

Tirapazamine (TPZ)

TPZ is an aromatic N-oxide that was originally developed in the mid-1980s, and its pharmacological properties have been extensively reviewed elsewhere [21]. Its mechanism of action is summarised in Fig. 7, and briefly, TPZ is reduced by one-electron reductases (such as cytochrome P450 reductase) to generate a radical species which in the absence of oxygen undergoes further spontaneous reactions, leading to the formation of DNA-damaging oxidising hydroxyl or benzotriazinyl radicals [101]. Selectivity for hypoxic cells is determined by the ability of oxygen to reverse the one-electron reduction step resulting in back oxidation to the parent compound and the formation of superoxide. The two-electron reductase NQO1 can also reduce TPZ, but this is considered a bioprotective mechanism as it bypasses the TPZ radical and forms the relatively non-toxic mono-N-oxide [102]. High hypoxia selectivity has been reported in a number of cell lines, and in vivo studies demonstrated that TPZ in combination with radiotherapy and cisplatin was highly effective against a range of human tumour xenografts [103] and TPZ entered clinical trial in early 1990s.
TPZ has been extensively evaluated in the clinic. Both phase I and II studies generated positive results, particularly phase II studies where TPZ was used in combination with cisplatin, etoposide and/or radiotherapy [104107]. Unfortunately, several phase III clinical trials have failed to demonstrate any survival advantage by adding TPZ to chemotherapy or radiotherapy in non-small cell lung cancer [108], head and neck cancer [109] and cervical cancer [110]. Reasons for the failure of TPZ include failure of radiotherapy protocol compliance and lack of stratification of patients based on tumour hypoxia levels [111, 112]. Subsequent subgroup analysis of these trials using a range of endogenous markers of hypoxia proved of limited benefit in both head and neck cancers and NSCLC trials [113115]. Whilst better methodologies for measuring hypoxia could have been employed [116, 117], the lack of a correlation between clinical response and hypoxia markers supports the clinical findings that inclusion of TPZ into combination protocols has limited if any clinical benefit. An alternative explanation for the failure of TPZ is relatively poor drug penetration into hypoxic regions of tumours. Because TPZ can be activated under comparatively mild hypoxia, it has been shown that TPZ is metabolised too rapidly to penetrate deeply into severely hypoxic tissue [118]. Using a combination of in silico models and experimental approaches, analogues of TPZ that have better penetration and metabolism properties have been developed with SN30000 (now known as CEN-209 following licensing to Centella) emerging as a candidate for clinical development [119]. Details of its mechanism of action have been described elsewhere [120], and SN30000 is likely to proceed to phase I clinical trials shortly. It is hoped that the valuable experience gained from the TPZ clinical trials is incorporated into the design of these trials [111].

TH-4000

The vast majority of HAPs developed to date result in the generation of metabolites that damage DNA either directly or indirectly. TH-4000 represents a significant departure from this mechanism and is one of the first hypoxia-activated molecularly targeted therapeutic to be developed. Discovered at the University of Auckland and initially referred to as PR-610 or Hypoxin™, TH-4000 is a hypoxia-activated EGFR tyrosine kinase inhibitor (TKI) that is now being developed by Threshold Pharmaceuticals. It is designed to release an irreversible EGFR TKI within hypoxic regions of tumours (Fig. 8), thereby improving selectivity and circumventing some of the toxicities observed with existing EGFR TKIs. Preclinical studies have been published in abstract form [121, 122] where TH-4000 was shown to be more active against NSCLC xenografts with wild-type and mutant EGFR than Erlotinib. TH-4000 is currently undergoing phase II clinical evaluation against EGFR-mutant, T790M-negative patients with advanced NSCLC (NCT02454842) and metastatic squamous cell carcinoma of the head and neck or skin (NCT02449681).

Novel HAPs in preclinical development

Whilst HAPs designed to release DNA-damaging species are still being developed, other HAPs have been designed to release inhibitors of DNA damage response pathways [123, 124]. Release of the Chk1/Aurora A inhibitor CH-01 (Fig. 9) following reduction of a 4-nitrobenzyl hypoxia trigger leads to fragmentation and release of active Chk1/Aurora A inhibitor with potent activity against severely hypoxic cells in vitro [123]. Similarly, attachment of a DNA-PK inhibitor to a nitroimidazole hypoxia trigger group led to inactivation of the complex (BCCA621C, Fig. 9) and no cytotoxic activity under aerobic conditions was reported [124]. Under severe hypoxia, however, the inhibitor was released and was able to radiosensitise hypoxic H460 cells in vitro with a sensitiser enhancement ratio of 1.85. HAPs targeting the O6-alkylguanine DNA alkyltransferase (AGT, Fig. 9) pathway have also been developed and have shown hypoxia selective activity in a range of cell lines in vitro [125]. This approach could lead to the selective depletion of AGT in tumour tissue without corresponding depletion in normal tissue leading to sensitisation of tumours to O6 guanine targeting cytotoxic drugs such as temozolomide. Using a similar concept to TH-4000, a HAP strategy has been developed to release EGFR inhibitors using Cobalt (III) as the hypoxia-sensitive trigger group (Fig. 9) [126]. Hypoxia selective inhibition of EGFR was demonstrated in vitro, and anticancer activity in vivo against A431 and Calu3 human tumour xenografts was demonstrated. HAP strategies are not only being used to deliver targeted small molecule but have recently been extended to include siRNA approaches. Azobenzene hypoxia trigger groups have been linked to siRNA for hypoxia-targeted delivery of siRNA along with PEGylated nanopreparations in a proof-of-concept study. Using multicell spheroids, the construct was shown to penetrate into the spheroid mass, and in HeLa cells engineered to stably express GFP, a 30–40 % down-regulation of GFP was detected only under hypoxic (0.5 % oxygen) conditions [127].

Concluding remarks and future directions

One of the most pressing unmet clinical needs is the development of therapeutic agents that can eradicate the hypoxic fraction of tumour cells. Despite extensive efforts to target and kill hypoxic cells over several decades, the need for such therapeutic strategies remains a significant objective. In the field of HAPs, several compounds have made it through pre-clinical development into clinical trial, but success has so far proved elusive. These failures, however, have helped shape the development and testing of new HAPs, and there is now genuine optimism that success is imminent. Of the compounds undergoing clinical development, TH-302 is currently the ‘gold standard’ and the results of phase III trials are eagerly awaited. As described in this article, there are a number of other HAPs in clinical trial and behind these, there is a pipeline of other agents undergoing preclinical evaluation or awaiting clinical trial. Of particular interest is the development of HAP strategies designed to release targeted therapeutics (pioneered by TH-4000) within the hypoxic microenvironment of tumours, and this is an exciting development. It should also be noted that HAPs represent one approach to targeting tumour hypoxia and other areas are being actively pursued [128]. One such avenue is tumour metabolism, and as the biology underpinning the metabolic phenotype of tumour cells and the metabolic interplay between tumour and host cells under hypoxia are unravelled, novel therapeutic targets and strategies will emerge. Despite its lack of immediate success, the field of HAP development has produced a wealth of knowledge, understanding and expertise. It is hoped that the novel approaches to targeting hypoxia under development now will take note of the principles and experience gained from over 40 years of developing HAPs and incorporate them into the design of appropriate preclinical and clinical studies.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Hockel M, Vaupel P (2001) Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93:266–276PubMedCrossRef Hockel M, Vaupel P (2001) Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93:266–276PubMedCrossRef
3.
Zurück zum Zitat Koch CJ, Jenkins WT, Jenkins KW, Yang XY, Shuman AL, Pickup S, Riehl CR, Paudyal R, Poptani H, Evans SM (2013) Mechanisms of blood flow and hypoxia production in rat 9L-epigastric tumors. Tumor Microenviron Ther 1:1–13PubMedCentralPubMedCrossRef Koch CJ, Jenkins WT, Jenkins KW, Yang XY, Shuman AL, Pickup S, Riehl CR, Paudyal R, Poptani H, Evans SM (2013) Mechanisms of blood flow and hypoxia production in rat 9L-epigastric tumors. Tumor Microenviron Ther 1:1–13PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, Giaccia AJ (1996) Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379:88–91PubMedCrossRef Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, Giaccia AJ (1996) Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379:88–91PubMedCrossRef
6.
Zurück zum Zitat Chang Q, Jurisica I, Do T, Hedley DW (2011) Hypoxia predicts aggressive growth and spontaneous metastasis formation from orthotopically grown primary xenografts of human pancreatic cancer. Cancer Res 71:3110–3120PubMedCrossRef Chang Q, Jurisica I, Do T, Hedley DW (2011) Hypoxia predicts aggressive growth and spontaneous metastasis formation from orthotopically grown primary xenografts of human pancreatic cancer. Cancer Res 71:3110–3120PubMedCrossRef
7.
Zurück zum Zitat Harris AL (2002) Hypoxia–a key regulatory factor in tumour growth. Nat Rev Cancer 2:38–47PubMedCrossRef Harris AL (2002) Hypoxia–a key regulatory factor in tumour growth. Nat Rev Cancer 2:38–47PubMedCrossRef
8.
Zurück zum Zitat Gluck AA, Aebersold DM, Zimmer Y, Medova M (2015) Interplay between receptor tyrosine kinases and hypoxia signaling in cancer. Int J Biochem Cell Biol 62:101–114PubMedCrossRef Gluck AA, Aebersold DM, Zimmer Y, Medova M (2015) Interplay between receptor tyrosine kinases and hypoxia signaling in cancer. Int J Biochem Cell Biol 62:101–114PubMedCrossRef
9.
Zurück zum Zitat Barsoum IB, Smallwood CA, Siemens DR, Graham CH (2014) A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 74:665–674PubMedCrossRef Barsoum IB, Smallwood CA, Siemens DR, Graham CH (2014) A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 74:665–674PubMedCrossRef
10.
Zurück zum Zitat Viry E, Paggetti J, Baginska J, Mgrditchian T, Berchem G, Moussay E, Janji B (2014) Autophagy: an adaptive metabolic response to stress shaping the antitumor immunity. Biochem Pharmacol 92:31–42PubMedCrossRef Viry E, Paggetti J, Baginska J, Mgrditchian T, Berchem G, Moussay E, Janji B (2014) Autophagy: an adaptive metabolic response to stress shaping the antitumor immunity. Biochem Pharmacol 92:31–42PubMedCrossRef
11.
Zurück zum Zitat Sun RC, Denko NC (2014) Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth. Cell Metab 19:285–292PubMedCentralPubMedCrossRef Sun RC, Denko NC (2014) Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth. Cell Metab 19:285–292PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Kucharzewska P, Christianson HC, Belting M (2015) Global profiling of metabolic adaptation to hypoxic stress in human glioblastoma cells. PLoS ONE 10:e0116740PubMedCentralPubMedCrossRef Kucharzewska P, Christianson HC, Belting M (2015) Global profiling of metabolic adaptation to hypoxic stress in human glioblastoma cells. PLoS ONE 10:e0116740PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Intlekofer AM, Dematteo RG, Venneti S, Finley LW, Lu C, Judkins AR, Rustenburg AS, Grinaway PB, Chodera JD, Cross JR, Thompson CB (2015) Hypoxia induces production of L-2-hydroxyglutarate. Cell Metab 22:304–311PubMedCrossRef Intlekofer AM, Dematteo RG, Venneti S, Finley LW, Lu C, Judkins AR, Rustenburg AS, Grinaway PB, Chodera JD, Cross JR, Thompson CB (2015) Hypoxia induces production of L-2-hydroxyglutarate. Cell Metab 22:304–311PubMedCrossRef
14.
Zurück zum Zitat Oldham WM, Clish CB, Yang Y, Loscalzo J (2015) Hypoxia-mediated increases in l-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab 22:291–303PubMedCrossRef Oldham WM, Clish CB, Yang Y, Loscalzo J (2015) Hypoxia-mediated increases in l-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab 22:291–303PubMedCrossRef
15.
Zurück zum Zitat Ye J, Wu D, Wu P, Chen Z, Huang J (2014) The cancer stem cell niche: cross talk between cancer stem cells and their microenvironment. Tumour Biol 35:3945–3951PubMedCrossRef Ye J, Wu D, Wu P, Chen Z, Huang J (2014) The cancer stem cell niche: cross talk between cancer stem cells and their microenvironment. Tumour Biol 35:3945–3951PubMedCrossRef
16.
Zurück zum Zitat Balamurugan K (2016) HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer 138:1058-1066PubMedCrossRef Balamurugan K (2016) HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer 138:1058-1066PubMedCrossRef
17.
Zurück zum Zitat Pires IM, Blokland NJ, Broos AW, Poujade FA, Senra JM, Eccles SA, Span PN, Harvey AJ, Hammond EM (2014) HIF-1alpha-independent hypoxia-induced rapid PTK6 stabilization is associated with increased motility and invasion. Cancer Biol Ther 15:1350–1357PubMedCentralPubMedCrossRef Pires IM, Blokland NJ, Broos AW, Poujade FA, Senra JM, Eccles SA, Span PN, Harvey AJ, Hammond EM (2014) HIF-1alpha-independent hypoxia-induced rapid PTK6 stabilization is associated with increased motility and invasion. Cancer Biol Ther 15:1350–1357PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Gray LH, Conger AO, Ebert M, Hornsey S, Scott OCA (1953) The concentration of oxygen dissolved in tissue at the time of irradiation as a factor in radiotherapy. Br J Radiol 26:638–648PubMedCrossRef Gray LH, Conger AO, Ebert M, Hornsey S, Scott OCA (1953) The concentration of oxygen dissolved in tissue at the time of irradiation as a factor in radiotherapy. Br J Radiol 26:638–648PubMedCrossRef
20.
Zurück zum Zitat Tredan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 99:1441–1454PubMedCrossRef Tredan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 99:1441–1454PubMedCrossRef
21.
22.
Zurück zum Zitat Ahmadi M, Ahmadihosseini Z, Allison SJ, Begum S, Rockley K, Sadiq M, Chintamaneni S, Lokwani R, Hughes N, Phillips RM (2014) Hypoxia modulates the activity of a series of clinically approved tyrosine kinase inhibitors. Br J Pharmacol 171:224–236PubMedCentralPubMedCrossRef Ahmadi M, Ahmadihosseini Z, Allison SJ, Begum S, Rockley K, Sadiq M, Chintamaneni S, Lokwani R, Hughes N, Phillips RM (2014) Hypoxia modulates the activity of a series of clinically approved tyrosine kinase inhibitors. Br J Pharmacol 171:224–236PubMedCentralPubMedCrossRef
23.
Zurück zum Zitat Hammond EM, Asselin MC, Forster D, O’Connor JP, Senra JM, Williams KJ (2014) The meaning, measurement and modification of hypoxia in the laboratory and the clinic. Clin Oncol (R Coll Radiol) 26:277–288CrossRef Hammond EM, Asselin MC, Forster D, O’Connor JP, Senra JM, Williams KJ (2014) The meaning, measurement and modification of hypoxia in the laboratory and the clinic. Clin Oncol (R Coll Radiol) 26:277–288CrossRef
24.
Zurück zum Zitat Vaupel P, Mayer A (2007) Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 26:225–239PubMedCrossRef Vaupel P, Mayer A (2007) Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 26:225–239PubMedCrossRef
25.
Zurück zum Zitat Lin AJ, Cosby LA, Shansky CW, Sartorelli AC (1972) Potential bioreductive alkylating agents. 1. Benzoquinone derivatives. J Med Chem 15:1247–1252PubMed Lin AJ, Cosby LA, Shansky CW, Sartorelli AC (1972) Potential bioreductive alkylating agents. 1. Benzoquinone derivatives. J Med Chem 15:1247–1252PubMed
26.
Zurück zum Zitat Teicher BA, Sartorelli AC (1980) Nitrobenzyl halides and carbamates as prototype bioreductive alkylating agents. J Med Chem 23:955–960PubMedCrossRef Teicher BA, Sartorelli AC (1980) Nitrobenzyl halides and carbamates as prototype bioreductive alkylating agents. J Med Chem 23:955–960PubMedCrossRef
27.
Zurück zum Zitat Guise CP, Mowday AM, Ashoorzadeh A, Yuan R, Lin WH, Wu DH, Smaill JB, Patterson AV, Ding K (2014) Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia. Chin J Cancer 33:80–86PubMedCentralPubMedCrossRef Guise CP, Mowday AM, Ashoorzadeh A, Yuan R, Lin WH, Wu DH, Smaill JB, Patterson AV, Ding K (2014) Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia. Chin J Cancer 33:80–86PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Denny WA, Wilson WR, Hay MP (1996) Recent developments in the design of bioreductive drugs. Br J Cancer Suppl 27:S32–S38PubMedCentralPubMed Denny WA, Wilson WR, Hay MP (1996) Recent developments in the design of bioreductive drugs. Br J Cancer Suppl 27:S32–S38PubMedCentralPubMed
29.
Zurück zum Zitat Workman P (1994) Enzyme-directed bioreductive drug development revisited: a commentary on recent progress and future prospects with emphasis on quinone anticancer agents and quinone metabolizing enzymes, particularly DT-diaphorase. Oncol Res 6:461–475PubMed Workman P (1994) Enzyme-directed bioreductive drug development revisited: a commentary on recent progress and future prospects with emphasis on quinone anticancer agents and quinone metabolizing enzymes, particularly DT-diaphorase. Oncol Res 6:461–475PubMed
30.
Zurück zum Zitat Duan JX, Jiao H, Kaizerman J, Stanton T, Evans JW, Lan L, Lorente G, Banica M, Jung D, Wang J, Ma H, Li X, Yang Z, Hoffman RM, Ammons WS, Hart CP, Matteucci M (2008) Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs. J Med Chem 51:2412–2420PubMedCrossRef Duan JX, Jiao H, Kaizerman J, Stanton T, Evans JW, Lan L, Lorente G, Banica M, Jung D, Wang J, Ma H, Li X, Yang Z, Hoffman RM, Ammons WS, Hart CP, Matteucci M (2008) Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs. J Med Chem 51:2412–2420PubMedCrossRef
31.
Zurück zum Zitat Meng F, Evans JW, Bhupathi D, Banica M, Lan L, Lorente G, Duan JX, Cai X, Mowday AM, Guise CP, Maroz A, Anderson RF, Patterson AV, Stachelek GC, Glazer PM, Matteucci MD, Hart CP (2012) Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302. Mol Cancer Ther 11:740–751PubMedCrossRef Meng F, Evans JW, Bhupathi D, Banica M, Lan L, Lorente G, Duan JX, Cai X, Mowday AM, Guise CP, Maroz A, Anderson RF, Patterson AV, Stachelek GC, Glazer PM, Matteucci MD, Hart CP (2012) Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302. Mol Cancer Ther 11:740–751PubMedCrossRef
32.
Zurück zum Zitat Saggar JK, Tannock IF (2014) Activity of the hypoxia-activated pro-drug TH-302 in hypoxic and perivascular regions of solid tumors and its potential to enhance therapeutic effects of chemotherapy. Int J Cancer 134:2726–2734PubMedCrossRef Saggar JK, Tannock IF (2014) Activity of the hypoxia-activated pro-drug TH-302 in hypoxic and perivascular regions of solid tumors and its potential to enhance therapeutic effects of chemotherapy. Int J Cancer 134:2726–2734PubMedCrossRef
33.
Zurück zum Zitat Wojtkowiak JW, Cornnell HC, Matsumoto S, Saito K, Takakusagi Y, Dutta P, Kim M, Zhang X, Leos R, Bailey KM, Martinez G, Lloyd MC, Weber C, Mitchell JB, Lynch RM, Baker AF, Gatenby RA, Rejniak KA, Hart C, Krishna MC, Gillies RJ (2015) Pyruvate sensitizes pancreatic tumors to hypoxia-activated prodrug TH-302. Cancer Metab 3:2PubMedCentralPubMedCrossRef Wojtkowiak JW, Cornnell HC, Matsumoto S, Saito K, Takakusagi Y, Dutta P, Kim M, Zhang X, Leos R, Bailey KM, Martinez G, Lloyd MC, Weber C, Mitchell JB, Lynch RM, Baker AF, Gatenby RA, Rejniak KA, Hart C, Krishna MC, Gillies RJ (2015) Pyruvate sensitizes pancreatic tumors to hypoxia-activated prodrug TH-302. Cancer Metab 3:2PubMedCentralPubMedCrossRef
34.
Zurück zum Zitat Hu J, Van Valckenborgh E, Xu D, Menu E, De Raeve H, De Bryune E, Xu S, Van Camp B, Handisides D, Hart CP, Vanderkerken K (2013) Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro. Mol Cancer Ther 12:1763–1773PubMedCrossRef Hu J, Van Valckenborgh E, Xu D, Menu E, De Raeve H, De Bryune E, Xu S, Van Camp B, Handisides D, Hart CP, Vanderkerken K (2013) Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro. Mol Cancer Ther 12:1763–1773PubMedCrossRef
35.
Zurück zum Zitat Liapis V, Labrinidis A, Zinonos I, Hay S, Ponomarev V, Panagopoulos V, DeNichilo M, Ingman W, Atkins GJ, Findlay DM, Zannettino AC, Evdokiou A (2015) Hypoxia-activated pro-drug TH-302 exhibits potent tumor suppressive activity and cooperates with chemotherapy against osteosarcoma. Cancer Lett 357:160–169PubMedCentralPubMedCrossRef Liapis V, Labrinidis A, Zinonos I, Hay S, Ponomarev V, Panagopoulos V, DeNichilo M, Ingman W, Atkins GJ, Findlay DM, Zannettino AC, Evdokiou A (2015) Hypoxia-activated pro-drug TH-302 exhibits potent tumor suppressive activity and cooperates with chemotherapy against osteosarcoma. Cancer Lett 357:160–169PubMedCentralPubMedCrossRef
36.
Zurück zum Zitat Meng F, Bhupathi D, Sun JD, Liu Q, Ahluwalia D, Wang Y, Matteucci MD, Hart CP (2015) Enhancement of hypoxia-activated prodrug TH-302 anti-tumor activity by Chk1 inhibition. BMC Cancer 15:422PubMedCentralPubMedCrossRef Meng F, Bhupathi D, Sun JD, Liu Q, Ahluwalia D, Wang Y, Matteucci MD, Hart CP (2015) Enhancement of hypoxia-activated prodrug TH-302 anti-tumor activity by Chk1 inhibition. BMC Cancer 15:422PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Peeters SG, Zegers CM, Biemans R, Lieuwes NG, van Stiphout RG, Yaromina A, Sun JD, Hart CP, Windhorst AD, van Elmpt W, Dubois LJ, Lambin P (2015) TH-302 in combination with radiotherapy enhances the therapeutic outcome and is associated with pretreatment [18F]HX4 hypoxia PET imaging. Clin Cancer Res 21:2984–2992PubMedCrossRef Peeters SG, Zegers CM, Biemans R, Lieuwes NG, van Stiphout RG, Yaromina A, Sun JD, Hart CP, Windhorst AD, van Elmpt W, Dubois LJ, Lambin P (2015) TH-302 in combination with radiotherapy enhances the therapeutic outcome and is associated with pretreatment [18F]HX4 hypoxia PET imaging. Clin Cancer Res 21:2984–2992PubMedCrossRef
38.
Zurück zum Zitat Saggar JK, Tannock IF (2015) Chemotherapy rescues hypoxic tumor cells and induces their reoxygenation and repopulation-an effect that is inhibited by the hypoxia-activated prodrug TH-302. Clin Cancer Res 21:2107–2114PubMedCrossRef Saggar JK, Tannock IF (2015) Chemotherapy rescues hypoxic tumor cells and induces their reoxygenation and repopulation-an effect that is inhibited by the hypoxia-activated prodrug TH-302. Clin Cancer Res 21:2107–2114PubMedCrossRef
39.
Zurück zum Zitat Sun JD, Liu Q, Ahluwalia D, Li W, Meng F, Wang Y, Bhupathi D, Ruprell AS, Hart CP (2015) Efficacy and safety of the hypoxia-activated prodrug TH-302 in combination with gemcitabine and nab-paclitaxel in human tumor xenograft models of pancreatic cancer. Cancer Biol Ther 16:438–449PubMedCentralPubMedCrossRef Sun JD, Liu Q, Ahluwalia D, Li W, Meng F, Wang Y, Bhupathi D, Ruprell AS, Hart CP (2015) Efficacy and safety of the hypoxia-activated prodrug TH-302 in combination with gemcitabine and nab-paclitaxel in human tumor xenograft models of pancreatic cancer. Cancer Biol Ther 16:438–449PubMedCentralPubMedCrossRef
40.
Zurück zum Zitat Yoon C, Lee HJ, Park DJ, Lee YJ, Tap WD, Eisinger-Mathason TS, Hart CP, Choy E, Simon MC, Yoon SS (2015) Hypoxia-activated chemotherapeutic TH-302 enhances the effects of VEGF-A inhibition and radiation on sarcomas. Br J Cancer 113:46–56PubMedCrossRef Yoon C, Lee HJ, Park DJ, Lee YJ, Tap WD, Eisinger-Mathason TS, Hart CP, Choy E, Simon MC, Yoon SS (2015) Hypoxia-activated chemotherapeutic TH-302 enhances the effects of VEGF-A inhibition and radiation on sarcomas. Br J Cancer 113:46–56PubMedCrossRef
41.
Zurück zum Zitat Portwood S, Lal D, Hsu YC, Vargas R, Johnson MK, Wetzler M, Hart CP, Wang ES (2013) Activity of the hypoxia-activated prodrug, TH-302, in preclinical human acute myeloid leukemia models. Clin Cancer Res 19:6506–6519PubMedCrossRef Portwood S, Lal D, Hsu YC, Vargas R, Johnson MK, Wetzler M, Hart CP, Wang ES (2013) Activity of the hypoxia-activated prodrug, TH-302, in preclinical human acute myeloid leukemia models. Clin Cancer Res 19:6506–6519PubMedCrossRef
42.
Zurück zum Zitat Ganjoo KN, Cranmer LD, Butrynski JE, Rushing D, Adkins D, Okuno SH, Lorente G, Kroll S, Langmuir VK, Chawla SP (2011) A phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology 80:50–56PubMedCrossRef Ganjoo KN, Cranmer LD, Butrynski JE, Rushing D, Adkins D, Okuno SH, Lorente G, Kroll S, Langmuir VK, Chawla SP (2011) A phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology 80:50–56PubMedCrossRef
43.
Zurück zum Zitat Weiss GJ, Infante JR, Chiorean EG, Borad MJ, Bendell JC, Molina JR, Tibes R, Ramanathan RK, Lewandowski K, Jones SF, Lacouture ME, Langmuir VK, Lee H, Kroll S, Burris HA 3rd (2011) Phase 1 study of the safety, tolerability, and pharmacokinetics of TH-302, a hypoxia-activated prodrug, in patients with advanced solid malignancies. Clin Cancer Res 17:2997–3004PubMedCrossRef Weiss GJ, Infante JR, Chiorean EG, Borad MJ, Bendell JC, Molina JR, Tibes R, Ramanathan RK, Lewandowski K, Jones SF, Lacouture ME, Langmuir VK, Lee H, Kroll S, Burris HA 3rd (2011) Phase 1 study of the safety, tolerability, and pharmacokinetics of TH-302, a hypoxia-activated prodrug, in patients with advanced solid malignancies. Clin Cancer Res 17:2997–3004PubMedCrossRef
44.
Zurück zum Zitat Weiss GJ, Lewandowski K, Oneall J, Kroll S (2011) Resolution of Cullen’s sign in patient with metastatic melanoma responding to hypoxia-activated prodrug TH-302. Dermatol Rep 3:e56CrossRef Weiss GJ, Lewandowski K, Oneall J, Kroll S (2011) Resolution of Cullen’s sign in patient with metastatic melanoma responding to hypoxia-activated prodrug TH-302. Dermatol Rep 3:e56CrossRef
45.
Zurück zum Zitat Chawla SP, Cranmer LD, Van Tine BA, Reed DR, Okuno SH, Butrynski JE, Adkins DR, Hendifar AE, Kroll S, Ganjoo KN (2014) Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. J Clin Oncol 32:3299–3306PubMedCentralPubMedCrossRef Chawla SP, Cranmer LD, Van Tine BA, Reed DR, Okuno SH, Butrynski JE, Adkins DR, Hendifar AE, Kroll S, Ganjoo KN (2014) Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. J Clin Oncol 32:3299–3306PubMedCentralPubMedCrossRef
46.
Zurück zum Zitat Borad MJ, Reddy SG, Bahary N, Uronis HE, Sigal D, Cohn AL, Schelman WR, Stephenson J Jr, Chiorean EG, Rosen PJ, Ulrich B, Dragovich T, Del Prete SA, Rarick M, Eng C, Kroll S, Ryan DP (2015) Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol 33:1475–1481PubMedCrossRef Borad MJ, Reddy SG, Bahary N, Uronis HE, Sigal D, Cohn AL, Schelman WR, Stephenson J Jr, Chiorean EG, Rosen PJ, Ulrich B, Dragovich T, Del Prete SA, Rarick M, Eng C, Kroll S, Ryan DP (2015) Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol 33:1475–1481PubMedCrossRef
47.
Zurück zum Zitat Oostveen EA, Speckamp WN (1987) Mitomycin analogs I. Indolequinones as (potential)bis alkylating agents. Tetrahedron 43:255–262CrossRef Oostveen EA, Speckamp WN (1987) Mitomycin analogs I. Indolequinones as (potential)bis alkylating agents. Tetrahedron 43:255–262CrossRef
48.
Zurück zum Zitat Phillips RM, Hendriks HR, Peters GJ, Pharmacology E, Molecular Mechanism G (2013) EO9 (Apaziquone): from the clinic to the laboratory and back again. Br J Pharmacol 168:11–18PubMedCentralPubMedCrossRef Phillips RM, Hendriks HR, Peters GJ, Pharmacology E, Molecular Mechanism G (2013) EO9 (Apaziquone): from the clinic to the laboratory and back again. Br J Pharmacol 168:11–18PubMedCentralPubMedCrossRef
49.
Zurück zum Zitat Plumb JA, Workman P (1994) Unusually marked hypoxic sensitization to indoloquinone EO9 and mitomycin C in a human colon-tumour cell line that lacks DT-diaphorase activity. Int J Cancer 56:134–139PubMedCrossRef Plumb JA, Workman P (1994) Unusually marked hypoxic sensitization to indoloquinone EO9 and mitomycin C in a human colon-tumour cell line that lacks DT-diaphorase activity. Int J Cancer 56:134–139PubMedCrossRef
50.
Zurück zum Zitat Robertson N, Haigh A, Adams GE, Stratford IJ (1994) Factors affecting sensitivity to EO9 in rodent and human tumour cells in vitro: DT-diaphorase activity and hypoxia. Eur J Cancer 30A:1013–1019PubMedCrossRef Robertson N, Haigh A, Adams GE, Stratford IJ (1994) Factors affecting sensitivity to EO9 in rodent and human tumour cells in vitro: DT-diaphorase activity and hypoxia. Eur J Cancer 30A:1013–1019PubMedCrossRef
51.
Zurück zum Zitat Traver RD, Siegel D, Beall HD, Phillips RM, Gibson NW, Franklin WA, Ross D (1997) Characterization of a polymorphism in NAD(P)H: quinone oxidoreductase (DT-diaphorase). Br J Cancer 75:69–75PubMedCentralPubMedCrossRef Traver RD, Siegel D, Beall HD, Phillips RM, Gibson NW, Franklin WA, Ross D (1997) Characterization of a polymorphism in NAD(P)H: quinone oxidoreductase (DT-diaphorase). Br J Cancer 75:69–75PubMedCentralPubMedCrossRef
52.
Zurück zum Zitat Hendriks HR, Pizao PE, Berger DP, Kooistra KL, Bibby MC, Boven E, Dreef-van der Meulen HC, Henrar RE, Fiebig HH, Double JA et al (1993) EO9: a novel bioreductive alkylating indoloquinone with preferential solid tumour activity and lack of bone marrow toxicity in preclinical models. Eur J Cancer 29A:897–906PubMedCrossRef Hendriks HR, Pizao PE, Berger DP, Kooistra KL, Bibby MC, Boven E, Dreef-van der Meulen HC, Henrar RE, Fiebig HH, Double JA et al (1993) EO9: a novel bioreductive alkylating indoloquinone with preferential solid tumour activity and lack of bone marrow toxicity in preclinical models. Eur J Cancer 29A:897–906PubMedCrossRef
53.
Zurück zum Zitat McLeod HL, Graham MA, Aamdal S, Setanoians A, Groot Y, Lund B (1996) Phase I pharmacokinetics and limited sampling strategies for the bioreductive alkylating drug EO9. EORTC Early Clinical Trials Group. Eur J Cancer 32A:1518–1522PubMedCrossRef McLeod HL, Graham MA, Aamdal S, Setanoians A, Groot Y, Lund B (1996) Phase I pharmacokinetics and limited sampling strategies for the bioreductive alkylating drug EO9. EORTC Early Clinical Trials Group. Eur J Cancer 32A:1518–1522PubMedCrossRef
54.
Zurück zum Zitat Schellens JH, Planting AS, van Acker BA, Loos WJ, de Boer-Dennert M, van der Burg ME, Koier I, Krediet RT, Stoter G, Verweij J (1994) Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09. J Natl Cancer Inst 86:906–912PubMedCrossRef Schellens JH, Planting AS, van Acker BA, Loos WJ, de Boer-Dennert M, van der Burg ME, Koier I, Krediet RT, Stoter G, Verweij J (1994) Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09. J Natl Cancer Inst 86:906–912PubMedCrossRef
55.
Zurück zum Zitat Dirix LY, Tonnesen F, Cassidy J, Epelbaum R, ten Bokkel Huinink WW, Pavlidis N, Sorio R, Gamucci T, Wolff I, Te Velde A, Lan J, Verweij J (1996) EO9 phase II study in advanced breast, gastric, pancreatic and colorectal carcinoma by the EORTC Early Clinical Studies Group. Eur J Cancer 32A:2019–2022PubMedCrossRef Dirix LY, Tonnesen F, Cassidy J, Epelbaum R, ten Bokkel Huinink WW, Pavlidis N, Sorio R, Gamucci T, Wolff I, Te Velde A, Lan J, Verweij J (1996) EO9 phase II study in advanced breast, gastric, pancreatic and colorectal carcinoma by the EORTC Early Clinical Studies Group. Eur J Cancer 32A:2019–2022PubMedCrossRef
56.
Zurück zum Zitat Pavlidis N, Hanauske AR, Gamucci T, Smyth J, Lehnert M, te Velde A, Lan J, Verweij J (1996) A randomized phase II study with two schedules of the novel indoloquinone EO9 in non-small-cell lung cancer: a study of the EORTC Early Clinical Studies Group (ECSG). Ann Oncol 7:529–531PubMedCrossRef Pavlidis N, Hanauske AR, Gamucci T, Smyth J, Lehnert M, te Velde A, Lan J, Verweij J (1996) A randomized phase II study with two schedules of the novel indoloquinone EO9 in non-small-cell lung cancer: a study of the EORTC Early Clinical Studies Group (ECSG). Ann Oncol 7:529–531PubMedCrossRef
57.
Zurück zum Zitat Connors TA (1996) Bioreductive agents, hypoxic cells and therapy. Eur J Cancer 32A:1833–1834PubMedCrossRef Connors TA (1996) Bioreductive agents, hypoxic cells and therapy. Eur J Cancer 32A:1833–1834PubMedCrossRef
58.
Zurück zum Zitat Minchinton AI, Tannock IF (2006) Drug penetration in solid tumours. Nat Rev Cancer 6:583–592PubMedCrossRef Minchinton AI, Tannock IF (2006) Drug penetration in solid tumours. Nat Rev Cancer 6:583–592PubMedCrossRef
59.
Zurück zum Zitat Phillips RM, Loadman PM, Cronin BP (1998) Evaluation of a novel in vitro assay for assessing drug penetration into avascular regions of tumours. Br J Cancer 77:2112–2119PubMedCentralPubMedCrossRef Phillips RM, Loadman PM, Cronin BP (1998) Evaluation of a novel in vitro assay for assessing drug penetration into avascular regions of tumours. Br J Cancer 77:2112–2119PubMedCentralPubMedCrossRef
60.
Zurück zum Zitat Choudry GA, Stewart PA, Double JA, Krul MR, Naylor B, Flannigan GM, Shah TK, Brown JE, Phillips RM (2001) A novel strategy for NQO1 (NAD(P)H:quinone oxidoreductase, EC 1.6.99.2) mediated therapy of bladder cancer based on the pharmacological properties of EO9. Br J Cancer 85:1137–1146PubMedCentralPubMedCrossRef Choudry GA, Stewart PA, Double JA, Krul MR, Naylor B, Flannigan GM, Shah TK, Brown JE, Phillips RM (2001) A novel strategy for NQO1 (NAD(P)H:quinone oxidoreductase, EC 1.6.99.2) mediated therapy of bladder cancer based on the pharmacological properties of EO9. Br J Cancer 85:1137–1146PubMedCentralPubMedCrossRef
61.
Zurück zum Zitat Puri R, Palit V, Loadman PM, Flannigan M, Shah T, Choudry GA, Basu S, Double JA, Lenaz G, Chawla S, Beer M, Van Kalken C, de Boer R, Beijnen JH, Twelves CJ, Phillips RM (2006) Phase I/II pilot study of intravesical apaziquone (EO9) for superficial bladder cancer. J Urol 176:1344–1348PubMedCrossRef Puri R, Palit V, Loadman PM, Flannigan M, Shah T, Choudry GA, Basu S, Double JA, Lenaz G, Chawla S, Beer M, Van Kalken C, de Boer R, Beijnen JH, Twelves CJ, Phillips RM (2006) Phase I/II pilot study of intravesical apaziquone (EO9) for superficial bladder cancer. J Urol 176:1344–1348PubMedCrossRef
62.
Zurück zum Zitat van der Heijden AG, Moonen PM, Cornel EB, Vergunst H, de Reijke TM, van Boven E, Barten EJ, Puri R, van Kalken CK, Witjes JA (2006) Phase II marker lesion study with intravesical instillation of apaziquone for superficial bladder cancer: toxicity and marker response. J Urol 176:1349–1353 (discussion 1353) PubMedCrossRef van der Heijden AG, Moonen PM, Cornel EB, Vergunst H, de Reijke TM, van Boven E, Barten EJ, Puri R, van Kalken CK, Witjes JA (2006) Phase II marker lesion study with intravesical instillation of apaziquone for superficial bladder cancer: toxicity and marker response. J Urol 176:1349–1353 (discussion 1353) PubMedCrossRef
63.
Zurück zum Zitat Hendricksen K, van der Heijden AG, Cornel EB, Vergunst H, de Reijke TM, van Boven E, Smits GA, Puri R, Gruijs S, Witjes JA (2009) Two-year follow-up of the phase II marker lesion study of intravesical apaziquone for patients with non-muscle invasive bladder cancer. World J Urol 27:337–342PubMedCentralPubMedCrossRef Hendricksen K, van der Heijden AG, Cornel EB, Vergunst H, de Reijke TM, van Boven E, Smits GA, Puri R, Gruijs S, Witjes JA (2009) Two-year follow-up of the phase II marker lesion study of intravesical apaziquone for patients with non-muscle invasive bladder cancer. World J Urol 27:337–342PubMedCentralPubMedCrossRef
64.
Zurück zum Zitat Jain A, Phillips RM, Scally AJ, Lenaz G, Beer M, Puri R (2009) Response of multiple recurrent TaT1 bladder cancer to intravesical apaziquone (EO9): comparative analysis of tumor recurrence rates. Urology 73:1083–1086PubMedCrossRef Jain A, Phillips RM, Scally AJ, Lenaz G, Beer M, Puri R (2009) Response of multiple recurrent TaT1 bladder cancer to intravesical apaziquone (EO9): comparative analysis of tumor recurrence rates. Urology 73:1083–1086PubMedCrossRef
65.
Zurück zum Zitat Hendricksen K, Gleason D, Young JM, Saltzstein D, Gershman A, Lerner S, Witjes JA (2008) Safety and side effects of immediate instillation of apaziquone following transurethral resection in patients with nonmuscle invasive bladder cancer. J Urol 180:116–120PubMedCrossRef Hendricksen K, Gleason D, Young JM, Saltzstein D, Gershman A, Lerner S, Witjes JA (2008) Safety and side effects of immediate instillation of apaziquone following transurethral resection in patients with nonmuscle invasive bladder cancer. J Urol 180:116–120PubMedCrossRef
66.
Zurück zum Zitat Nishida CR, Ortiz de Montellano PR (2008) Reductive heme-dependent activation of the n-oxide prodrug AQ4N by nitric oxide synthase. J Med Chem 51:5118–5120PubMedCentralPubMedCrossRef Nishida CR, Ortiz de Montellano PR (2008) Reductive heme-dependent activation of the n-oxide prodrug AQ4N by nitric oxide synthase. J Med Chem 51:5118–5120PubMedCentralPubMedCrossRef
67.
Zurück zum Zitat Raleigh SM, Wanogho E, Burke MD, McKeown SR, Patterson LH (1998) Involvement of human cytochromes P450 (CYP) in the reductive metabolism of AQ4 N, a hypoxia activated anthraquinone di-N-oxide prodrug. Int J Radiat Oncol Biol Phys 42:763–767PubMedCrossRef Raleigh SM, Wanogho E, Burke MD, McKeown SR, Patterson LH (1998) Involvement of human cytochromes P450 (CYP) in the reductive metabolism of AQ4 N, a hypoxia activated anthraquinone di-N-oxide prodrug. Int J Radiat Oncol Biol Phys 42:763–767PubMedCrossRef
69.
Zurück zum Zitat Mehibel M, Singh S, Chinje EC, Cowen RL, Stratford IJ (2009) Effects of cytokine-induced macrophages on the response of tumor cells to banoxantrone (AQ4 N). Mol Cancer Ther 8:1261–1269PubMedCrossRef Mehibel M, Singh S, Chinje EC, Cowen RL, Stratford IJ (2009) Effects of cytokine-induced macrophages on the response of tumor cells to banoxantrone (AQ4 N). Mol Cancer Ther 8:1261–1269PubMedCrossRef
70.
Zurück zum Zitat McKeown SR, Hejmadi MV, McIntyre IA, McAleer JJ, Patterson LH (1995) AQ4 N: an alkylaminoanthraquinone N-oxide showing bioreductive potential and positive interaction with radiation in vivo. Br J Cancer 72:76–81PubMedCentralPubMedCrossRef McKeown SR, Hejmadi MV, McIntyre IA, McAleer JJ, Patterson LH (1995) AQ4 N: an alkylaminoanthraquinone N-oxide showing bioreductive potential and positive interaction with radiation in vivo. Br J Cancer 72:76–81PubMedCentralPubMedCrossRef
71.
Zurück zum Zitat Patterson LH, McKeown SR, Ruparelia K, Double JA, Bibby MC, Cole S, Stratford IJ (2000) Enhancement of chemotherapy and radiotherapy of murine tumours by AQ4 N, a bioreductively activated anti-tumour agent. Br J Cancer 82:1984–1990PubMedCentralPubMedCrossRef Patterson LH, McKeown SR, Ruparelia K, Double JA, Bibby MC, Cole S, Stratford IJ (2000) Enhancement of chemotherapy and radiotherapy of murine tumours by AQ4 N, a bioreductively activated anti-tumour agent. Br J Cancer 82:1984–1990PubMedCentralPubMedCrossRef
72.
Zurück zum Zitat Friery OP, Gallagher R, Murray MM, Hughes CM, Galligan ES, McIntyre IA, Patterson LH, Hirst DG, McKeown SR (2000) Enhancement of the anti-tumour effect of cyclophosphamide by the bioreductive drugs AQ4N and tirapazamine. Br J Cancer 82:1469–1473PubMedCentralPubMedCrossRef Friery OP, Gallagher R, Murray MM, Hughes CM, Galligan ES, McIntyre IA, Patterson LH, Hirst DG, McKeown SR (2000) Enhancement of the anti-tumour effect of cyclophosphamide by the bioreductive drugs AQ4N and tirapazamine. Br J Cancer 82:1469–1473PubMedCentralPubMedCrossRef
73.
Zurück zum Zitat Tredan O, Garbens AB, Lalani AS, Tannock IF (2009) The hypoxia-activated ProDrug AQ4N penetrates deeply in tumor tissues and complements the limited distribution of mitoxantrone. Cancer Res 69:940–947PubMedCrossRef Tredan O, Garbens AB, Lalani AS, Tannock IF (2009) The hypoxia-activated ProDrug AQ4N penetrates deeply in tumor tissues and complements the limited distribution of mitoxantrone. Cancer Res 69:940–947PubMedCrossRef
74.
Zurück zum Zitat Ming L, Byrne NM, Camac SN, Mitchell CA, Ward C, Waugh DJ, McKeown SR, Worthington J (2013) Androgen deprivation results in time-dependent hypoxia in LNCaP prostate tumours: informed scheduling of the bioreductive drug AQ4N improves treatment response. Int J Cancer 132:1323–1332PubMedCrossRef Ming L, Byrne NM, Camac SN, Mitchell CA, Ward C, Waugh DJ, McKeown SR, Worthington J (2013) Androgen deprivation results in time-dependent hypoxia in LNCaP prostate tumours: informed scheduling of the bioreductive drug AQ4N improves treatment response. Int J Cancer 132:1323–1332PubMedCrossRef
75.
Zurück zum Zitat Gieling RG, Fitzmaurice RJ, Telfer BA, Babur M, Williams KJ (2015) Dissemination via the lymphatic or angiogenic route impacts the pathology, microenvironment and hypoxia-related drug response of lung metastases. Clin Exp Metastasis 32:567–577PubMedCrossRef Gieling RG, Fitzmaurice RJ, Telfer BA, Babur M, Williams KJ (2015) Dissemination via the lymphatic or angiogenic route impacts the pathology, microenvironment and hypoxia-related drug response of lung metastases. Clin Exp Metastasis 32:567–577PubMedCrossRef
76.
Zurück zum Zitat Williams KJ, Albertella MR, Fitzpatrick B, Loadman PM, Shnyder SD, Chinje EC, Telfer BA, Dunk CR, Harris PA, Stratford IJ (2009) In vivo activation of the hypoxia-targeted cytotoxin AQ4N in human tumor xenografts. Mol Cancer Ther 8:3266–3275PubMedCrossRef Williams KJ, Albertella MR, Fitzpatrick B, Loadman PM, Shnyder SD, Chinje EC, Telfer BA, Dunk CR, Harris PA, Stratford IJ (2009) In vivo activation of the hypoxia-targeted cytotoxin AQ4N in human tumor xenografts. Mol Cancer Ther 8:3266–3275PubMedCrossRef
77.
Zurück zum Zitat O’Rourke M, Ward C, Worthington J, McKenna J, Valentine A, Robson T, Hirst DG, McKeown SR (2008) Evaluation of the antiangiogenic potential of AQ4 N. Clin Cancer Res 14:1502–1509PubMedCrossRef O’Rourke M, Ward C, Worthington J, McKenna J, Valentine A, Robson T, Hirst DG, McKeown SR (2008) Evaluation of the antiangiogenic potential of AQ4 N. Clin Cancer Res 14:1502–1509PubMedCrossRef
78.
Zurück zum Zitat Albertella MR, Loadman PM, Jones PH, Phillips RM, Rampling R, Burnet N, Alcock C, Anthoney A, Vjaters E, Dunk CR, Harris PA, Wong A, Lalani AS, Twelves CJ (2008) Hypoxia-selective targeting by the bioreductive prodrug AQ4N in patients with solid tumors: results of a phase I study. Clin Cancer Res 14:1096–1104PubMedCrossRef Albertella MR, Loadman PM, Jones PH, Phillips RM, Rampling R, Burnet N, Alcock C, Anthoney A, Vjaters E, Dunk CR, Harris PA, Wong A, Lalani AS, Twelves CJ (2008) Hypoxia-selective targeting by the bioreductive prodrug AQ4N in patients with solid tumors: results of a phase I study. Clin Cancer Res 14:1096–1104PubMedCrossRef
79.
Zurück zum Zitat Papadopoulos KP, Goel S, Beeram M, Wong A, Desai K, Haigentz M, Milian ML, Mani S, Tolcher A, Lalani AS, Sarantopoulos J (2008) A phase 1 open-label, accelerated dose-escalation study of the hypoxia-activated prodrug AQ4N in patients with advanced malignancies. Clin Cancer Res 14:7110–7115PubMedCrossRef Papadopoulos KP, Goel S, Beeram M, Wong A, Desai K, Haigentz M, Milian ML, Mani S, Tolcher A, Lalani AS, Sarantopoulos J (2008) A phase 1 open-label, accelerated dose-escalation study of the hypoxia-activated prodrug AQ4N in patients with advanced malignancies. Clin Cancer Res 14:7110–7115PubMedCrossRef
80.
Zurück zum Zitat Steward WP, Middleton M, Benghiat A, Loadman PM, Hayward C, Waller S, Ford S, Halbert G, Patterson LH, Talbot D (2007) The use of pharmacokinetic and pharmacodynamic end points to determine the dose of AQ4N, a novel hypoxic cell cytotoxin, given with fractionated radiotherapy in a phase I study. Ann Oncol 18:1098–1103PubMedCrossRef Steward WP, Middleton M, Benghiat A, Loadman PM, Hayward C, Waller S, Ford S, Halbert G, Patterson LH, Talbot D (2007) The use of pharmacokinetic and pharmacodynamic end points to determine the dose of AQ4N, a novel hypoxic cell cytotoxin, given with fractionated radiotherapy in a phase I study. Ann Oncol 18:1098–1103PubMedCrossRef
81.
Zurück zum Zitat Patterson AV, Ferry DM, Edmunds SJ, Gu Y, Singleton RS, Patel K, Pullen SM, Hicks KO, Syddall SP, Atwell GJ, Yang S, Denny WA, Wilson WR (2007) Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA cross-linking agent PR-104. Clin Cancer Res 13:3922–3932PubMedCrossRef Patterson AV, Ferry DM, Edmunds SJ, Gu Y, Singleton RS, Patel K, Pullen SM, Hicks KO, Syddall SP, Atwell GJ, Yang S, Denny WA, Wilson WR (2007) Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA cross-linking agent PR-104. Clin Cancer Res 13:3922–3932PubMedCrossRef
82.
Zurück zum Zitat Guise CP, Wang AT, Theil A, Bridewell DJ, Wilson WR, Patterson AV (2007) Identification of human reductases that activate the dinitrobenzamide mustard prodrug PR-104A: a role for NADPH:cytochrome P450 oxidoreductase under hypoxia. Biochem Pharmacol 74:810–820PubMedCrossRef Guise CP, Wang AT, Theil A, Bridewell DJ, Wilson WR, Patterson AV (2007) Identification of human reductases that activate the dinitrobenzamide mustard prodrug PR-104A: a role for NADPH:cytochrome P450 oxidoreductase under hypoxia. Biochem Pharmacol 74:810–820PubMedCrossRef
83.
Zurück zum Zitat Hunter FW, Jaiswal JK, Hurley DG, Liyanage HD, McManaway SP, Gu Y, Richter S, Wang J, Tercel M, Print CG, Wilson WR, Pruijn FB (2014) The flavoprotein FOXRED2 reductively activates nitro-chloromethylbenzindolines and other hypoxia-targeting prodrugs. Biochem Pharmacol 89:224–235PubMedCrossRef Hunter FW, Jaiswal JK, Hurley DG, Liyanage HD, McManaway SP, Gu Y, Richter S, Wang J, Tercel M, Print CG, Wilson WR, Pruijn FB (2014) The flavoprotein FOXRED2 reductively activates nitro-chloromethylbenzindolines and other hypoxia-targeting prodrugs. Biochem Pharmacol 89:224–235PubMedCrossRef
84.
Zurück zum Zitat Guise CP, Abbattista MR, Singleton RS, Holford SD, Connolly J, Dachs GU, Fox SB, Pollock R, Harvey J, Guilford P, Donate F, Wilson WR, Patterson AV (2010) The bioreductive prodrug PR-104A is activated under aerobic conditions by human aldo-keto reductase 1C3. Cancer Res 70:1573–1584PubMedCrossRef Guise CP, Abbattista MR, Singleton RS, Holford SD, Connolly J, Dachs GU, Fox SB, Pollock R, Harvey J, Guilford P, Donate F, Wilson WR, Patterson AV (2010) The bioreductive prodrug PR-104A is activated under aerobic conditions by human aldo-keto reductase 1C3. Cancer Res 70:1573–1584PubMedCrossRef
85.
Zurück zum Zitat Guise CP, Abbattista MR, Tipparaju SR, Lambie NK, Su J, Li D, Wilson WR, Dachs GU, Patterson AV (2012) Diflavin oxidoreductases activate the bioreductive prodrug PR-104A under hypoxia. Mol Pharmacol 81:31–40PubMedCrossRef Guise CP, Abbattista MR, Tipparaju SR, Lambie NK, Su J, Li D, Wilson WR, Dachs GU, Patterson AV (2012) Diflavin oxidoreductases activate the bioreductive prodrug PR-104A under hypoxia. Mol Pharmacol 81:31–40PubMedCrossRef
86.
Zurück zum Zitat Gu Y, Patterson AV, Atwell GJ, Chernikova SB, Brown JM, Thompson LH, Wilson WR (2009) Roles of DNA repair and reductase activity in the cytotoxicity of the hypoxia-activated dinitrobenzamide mustard PR-104A. Mol Cancer Ther 8:1714–1723PubMedCrossRef Gu Y, Patterson AV, Atwell GJ, Chernikova SB, Brown JM, Thompson LH, Wilson WR (2009) Roles of DNA repair and reductase activity in the cytotoxicity of the hypoxia-activated dinitrobenzamide mustard PR-104A. Mol Cancer Ther 8:1714–1723PubMedCrossRef
87.
Zurück zum Zitat Hunter FW, Hsu HL, Su J, Pullen SM, Wilson WR, Wang J (2014) Dual targeting of hypoxia and homologous recombination repair dysfunction in triple-negative breast cancer. Mol Cancer Ther 13:2501–2514PubMedCrossRef Hunter FW, Hsu HL, Su J, Pullen SM, Wilson WR, Wang J (2014) Dual targeting of hypoxia and homologous recombination repair dysfunction in triple-negative breast cancer. Mol Cancer Ther 13:2501–2514PubMedCrossRef
88.
Zurück zum Zitat Moradi Manesh D, El-Hoss J, Evans K, Richmond J, Toscan CE, Bracken LS, Hedrick A, Sutton R, Marshall GM, Wilson WR, Kurmasheva RT, Billups C, Houghton PJ, Smith MA, Carol H, Lock RB (2015) AKR1C3 is a biomarker of sensitivity to PR-104 in preclinical models of T-cell acute lymphoblastic leukemia. Blood 126:1193–1202PubMedCrossRef Moradi Manesh D, El-Hoss J, Evans K, Richmond J, Toscan CE, Bracken LS, Hedrick A, Sutton R, Marshall GM, Wilson WR, Kurmasheva RT, Billups C, Houghton PJ, Smith MA, Carol H, Lock RB (2015) AKR1C3 is a biomarker of sensitivity to PR-104 in preclinical models of T-cell acute lymphoblastic leukemia. Blood 126:1193–1202PubMedCrossRef
89.
Zurück zum Zitat Houghton PJ, Lock R, Carol H, Morton CL, Phelps D, Gorlick R, Kolb EA, Keir ST, Reynolds CP, Kang MH, Maris JM, Wozniak AW, Gu Y, Wilson WR, Smith MA (2011) Initial testing of the hypoxia-activated prodrug PR-104 by the pediatric preclinical testing program. Pediatr Blood Cancer 57:443–453PubMedCentralPubMedCrossRef Houghton PJ, Lock R, Carol H, Morton CL, Phelps D, Gorlick R, Kolb EA, Keir ST, Reynolds CP, Kang MH, Maris JM, Wozniak AW, Gu Y, Wilson WR, Smith MA (2011) Initial testing of the hypoxia-activated prodrug PR-104 by the pediatric preclinical testing program. Pediatr Blood Cancer 57:443–453PubMedCentralPubMedCrossRef
90.
Zurück zum Zitat Benito J, Shi Y, Szymanska B, Carol H, Boehm I, Lu H, Konoplev S, Fang W, Zweidler-McKay PA, Campana D, Borthakur G, Bueso-Ramos C, Shpall E, Thomas DA, Jordan CT, Kantarjian H, Wilson WR, Lock R, Andreeff M, Konopleva M (2011) Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS ONE 6:e23108PubMedCentralPubMedCrossRef Benito J, Shi Y, Szymanska B, Carol H, Boehm I, Lu H, Konoplev S, Fang W, Zweidler-McKay PA, Campana D, Borthakur G, Bueso-Ramos C, Shpall E, Thomas DA, Jordan CT, Kantarjian H, Wilson WR, Lock R, Andreeff M, Konopleva M (2011) Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS ONE 6:e23108PubMedCentralPubMedCrossRef
91.
Zurück zum Zitat Foehrenbacher A, Secomb TW, Wilson WR, Hicks KO (2013) Design of optimized hypoxia-activated prodrugs using pharmacokinetic/pharmacodynamic modeling. Front Oncol 3:314PubMedCentralPubMed Foehrenbacher A, Secomb TW, Wilson WR, Hicks KO (2013) Design of optimized hypoxia-activated prodrugs using pharmacokinetic/pharmacodynamic modeling. Front Oncol 3:314PubMedCentralPubMed
92.
Zurück zum Zitat Foehrenbacher A, Patel K, Abbattista MR, Guise CP, Secomb TW, Wilson WR, Hicks KO (2013) The role of bystander effects in the antitumor activity of the hypoxia-activated prodrug PR-104. Front Oncol 3:263PubMedCentralPubMed Foehrenbacher A, Patel K, Abbattista MR, Guise CP, Secomb TW, Wilson WR, Hicks KO (2013) The role of bystander effects in the antitumor activity of the hypoxia-activated prodrug PR-104. Front Oncol 3:263PubMedCentralPubMed
93.
Zurück zum Zitat Abbattista MR, Jamieson SM, Gu Y, Nickel JE, Pullen SM, Patterson AV, Wilson WR, Guise CP (2015) Pre-clinical activity of PR-104 as monotherapy and in combination with sorafenib in hepatocellular carcinoma. Cancer Biol Ther 16:610–622PubMedCrossRef Abbattista MR, Jamieson SM, Gu Y, Nickel JE, Pullen SM, Patterson AV, Wilson WR, Guise CP (2015) Pre-clinical activity of PR-104 as monotherapy and in combination with sorafenib in hepatocellular carcinoma. Cancer Biol Ther 16:610–622PubMedCrossRef
94.
Zurück zum Zitat Cairns RA, Bennewith KL, Graves EE, Giaccia AJ, Chang DT, Denko NC (2009) Pharmacologically increased tumor hypoxia can be measured by 18F-Fluoroazomycin arabinoside positron emission tomography and enhances tumor response to hypoxic cytotoxin PR-104. Clin Cancer Res 15:7170–7174PubMedCentralPubMedCrossRef Cairns RA, Bennewith KL, Graves EE, Giaccia AJ, Chang DT, Denko NC (2009) Pharmacologically increased tumor hypoxia can be measured by 18F-Fluoroazomycin arabinoside positron emission tomography and enhances tumor response to hypoxic cytotoxin PR-104. Clin Cancer Res 15:7170–7174PubMedCentralPubMedCrossRef
95.
Zurück zum Zitat Jameson MB, Rischin D, Pegram M, Gutheil J, Patterson AV, Denny WA, Wilson WR (2010) A phase I trial of PR-104, a nitrogen mustard prodrug activated by both hypoxia and aldo-keto reductase 1C3, in patients with solid tumors. Cancer Chemother Pharmacol 65:791–801PubMedCrossRef Jameson MB, Rischin D, Pegram M, Gutheil J, Patterson AV, Denny WA, Wilson WR (2010) A phase I trial of PR-104, a nitrogen mustard prodrug activated by both hypoxia and aldo-keto reductase 1C3, in patients with solid tumors. Cancer Chemother Pharmacol 65:791–801PubMedCrossRef
96.
Zurück zum Zitat McKeage MJ, Gu Y, Wilson WR, Hill A, Amies K, Melink TJ, Jameson MB (2011) A phase I trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid tumour patients. BMC Cancer 11:432PubMedCentralPubMedCrossRef McKeage MJ, Gu Y, Wilson WR, Hill A, Amies K, Melink TJ, Jameson MB (2011) A phase I trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid tumour patients. BMC Cancer 11:432PubMedCentralPubMedCrossRef
97.
Zurück zum Zitat McKeage MJ, Jameson MB, Ramanathan RK, Rajendran J, Gu Y, Wilson WR, Melink TJ, Tchekmedyian NS (2012) PR-104 a bioreductive pre-prodrug combined with gemcitabine or docetaxel in a phase Ib study of patients with advanced solid tumours. BMC Cancer 12:496PubMedCentralPubMedCrossRef McKeage MJ, Jameson MB, Ramanathan RK, Rajendran J, Gu Y, Wilson WR, Melink TJ, Tchekmedyian NS (2012) PR-104 a bioreductive pre-prodrug combined with gemcitabine or docetaxel in a phase Ib study of patients with advanced solid tumours. BMC Cancer 12:496PubMedCentralPubMedCrossRef
98.
Zurück zum Zitat Abou-Alfa GK, Chan SL, Lin CC, Chiorean EG, Holcombe RF, Mulcahy MF, Carter WD, Patel K, Wilson WR, Melink TJ, Gutheil JC, Tsao CJ (2011) PR-104 plus sorafenib in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol 68:539–545PubMedCrossRef Abou-Alfa GK, Chan SL, Lin CC, Chiorean EG, Holcombe RF, Mulcahy MF, Carter WD, Patel K, Wilson WR, Melink TJ, Gutheil JC, Tsao CJ (2011) PR-104 plus sorafenib in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol 68:539–545PubMedCrossRef
99.
Zurück zum Zitat Gu Y, Tingle MD, Wilson WR (2011) Glucuronidation of anticancer prodrug PR-104A: species differences, identification of human UDP-glucuronosyltransferases, and implications for therapy. J Pharmacol Exp Ther 337:692–702PubMedCrossRef Gu Y, Tingle MD, Wilson WR (2011) Glucuronidation of anticancer prodrug PR-104A: species differences, identification of human UDP-glucuronosyltransferases, and implications for therapy. J Pharmacol Exp Ther 337:692–702PubMedCrossRef
100.
Zurück zum Zitat Konopleva M, Thall PF, Yi CA, Borthakur G, Coveler A, Bueso-Ramos C, Benito J, Konoplev S, Gu Y, Ravandi F, Jabbour E, Faderl S, Thomas D, Cortes J, Kadia T, Kornblau S, Daver N, Pemmaraju N, Nguyen HQ, Feliu J, Lu H, Wei C, Wilson WR, Melink TJ, Gutheil JC, Andreeff M, Estey EH, Kantarjian H (2015) Phase I/II study of the hypoxia-activated prodrug PR104 in refractory/relapsed acute myeloid leukemia and acute lymphoblastic leukemia. Haematologica 100:927–934PubMedCentralPubMedCrossRef Konopleva M, Thall PF, Yi CA, Borthakur G, Coveler A, Bueso-Ramos C, Benito J, Konoplev S, Gu Y, Ravandi F, Jabbour E, Faderl S, Thomas D, Cortes J, Kadia T, Kornblau S, Daver N, Pemmaraju N, Nguyen HQ, Feliu J, Lu H, Wei C, Wilson WR, Melink TJ, Gutheil JC, Andreeff M, Estey EH, Kantarjian H (2015) Phase I/II study of the hypoxia-activated prodrug PR104 in refractory/relapsed acute myeloid leukemia and acute lymphoblastic leukemia. Haematologica 100:927–934PubMedCentralPubMedCrossRef
101.
Zurück zum Zitat Shinde SS, Hay MP, Patterson AV, Denny WA, Anderson RF (2009) Spin trapping of radicals other than the *OH radical upon reduction of the anticancer agent tirapazamine by cytochrome P450 reductase. J Am Chem Soc 131:14220–14221PubMedCrossRef Shinde SS, Hay MP, Patterson AV, Denny WA, Anderson RF (2009) Spin trapping of radicals other than the *OH radical upon reduction of the anticancer agent tirapazamine by cytochrome P450 reductase. J Am Chem Soc 131:14220–14221PubMedCrossRef
102.
Zurück zum Zitat Patterson AV, Robertson N, Houlbrook S, Stephens MA, Adams GE, Harris AL, Stratford IJ, Carmichael J (1994) The role of DT-diaphorase in determining the sensitivity of human tumor cells to tirapazamine (SR 4233). Int J Radiat Oncol Biol Phys 29:369–372PubMedCrossRef Patterson AV, Robertson N, Houlbrook S, Stephens MA, Adams GE, Harris AL, Stratford IJ, Carmichael J (1994) The role of DT-diaphorase in determining the sensitivity of human tumor cells to tirapazamine (SR 4233). Int J Radiat Oncol Biol Phys 29:369–372PubMedCrossRef
103.
Zurück zum Zitat Marcu L, Olver I (2006) Tirapazamine: from bench to clinical trials. Curr Clin Pharmacol 1:71–79PubMedCrossRef Marcu L, Olver I (2006) Tirapazamine: from bench to clinical trials. Curr Clin Pharmacol 1:71–79PubMedCrossRef
104.
Zurück zum Zitat Le QT, Moon J, Redman M, Williamson SK, Lara PN Jr, Goldberg Z, Gaspar LE, Crowley JJ, Moore DF Jr, Gandara DR (2009) Phase II study of tirapazamine, cisplatin, and etoposide and concurrent thoracic radiotherapy for limited-stage small-cell lung cancer: SWOG 0222. J Clin Oncol 27:3014–3019PubMedCentralPubMedCrossRef Le QT, Moon J, Redman M, Williamson SK, Lara PN Jr, Goldberg Z, Gaspar LE, Crowley JJ, Moore DF Jr, Gandara DR (2009) Phase II study of tirapazamine, cisplatin, and etoposide and concurrent thoracic radiotherapy for limited-stage small-cell lung cancer: SWOG 0222. J Clin Oncol 27:3014–3019PubMedCentralPubMedCrossRef
105.
Zurück zum Zitat Miller VA, Ng KK, Grant SC, Kindler H, Pizzo B, Heelan RT, von Roemeling R, Kris MG (1997) Phase II study of the combination of the novel bioreductive agent, tirapazamine, with cisplatin in patients with advanced non-small-cell lung cancer. Ann Oncol 8:1269–1271PubMedCrossRef Miller VA, Ng KK, Grant SC, Kindler H, Pizzo B, Heelan RT, von Roemeling R, Kris MG (1997) Phase II study of the combination of the novel bioreductive agent, tirapazamine, with cisplatin in patients with advanced non-small-cell lung cancer. Ann Oncol 8:1269–1271PubMedCrossRef
106.
Zurück zum Zitat Rischin D, Peters L, Fisher R, Macann A, Denham J, Poulsen M, Jackson M, Kenny L, Penniment M, Corry J, Lamb D, McClure B (2005) Tirapazamine, cisplatin, and radiation versus fluorouracil, cisplatin, and radiation in patients with locally advanced head and neck cancer: a randomized phase II trial of the Trans-Tasman Radiation Oncology Group (TROG 98.02). J Clin Oncol 23:79–87PubMedCrossRef Rischin D, Peters L, Fisher R, Macann A, Denham J, Poulsen M, Jackson M, Kenny L, Penniment M, Corry J, Lamb D, McClure B (2005) Tirapazamine, cisplatin, and radiation versus fluorouracil, cisplatin, and radiation in patients with locally advanced head and neck cancer: a randomized phase II trial of the Trans-Tasman Radiation Oncology Group (TROG 98.02). J Clin Oncol 23:79–87PubMedCrossRef
107.
Zurück zum Zitat Treat J, Johnson E, Langer C, Belani C, Haynes B, Greenberg R, Rodriquez R, Drobins P, Miller W Jr, Meehan L, McKeon A, Devin J, von Roemeling R, Viallet J (1998) Tirapazamine with cisplatin in patients with advanced non-small-cell lung cancer: a phase II study. J Clin Oncol 16:3524–3527PubMed Treat J, Johnson E, Langer C, Belani C, Haynes B, Greenberg R, Rodriquez R, Drobins P, Miller W Jr, Meehan L, McKeon A, Devin J, von Roemeling R, Viallet J (1998) Tirapazamine with cisplatin in patients with advanced non-small-cell lung cancer: a phase II study. J Clin Oncol 16:3524–3527PubMed
108.
Zurück zum Zitat Williamson SK, Crowley JJ, Lara PN, Jr., McCoy J, Lau DH, Tucker RW, Mills GM, Gandara DR, Southwest Oncology Group Trial S (2005) Phase III trial of paclitaxel plus carboplatin with or without tirapazamine in advanced non-small-cell lung cancer: Southwest Oncology Group Trial S0003. J Clin Oncol 23:9097–9104CrossRef Williamson SK, Crowley JJ, Lara PN, Jr., McCoy J, Lau DH, Tucker RW, Mills GM, Gandara DR, Southwest Oncology Group Trial S (2005) Phase III trial of paclitaxel plus carboplatin with or without tirapazamine in advanced non-small-cell lung cancer: Southwest Oncology Group Trial S0003. J Clin Oncol 23:9097–9104CrossRef
109.
Zurück zum Zitat Rischin D, Peters LJ, O’Sullivan B, Giralt J, Fisher R, Yuen K, Trotti A, Bernier J, Bourhis J, Ringash J, Henke M, Kenny L (2010) Tirapazamine, cisplatin, and radiation versus cisplatin and radiation for advanced squamous cell carcinoma of the head and neck (TROG 02.02, HeadSTART): a phase III trial of the Trans-Tasman Radiation Oncology Group. J Clin Oncol 28:2989–2995PubMedCrossRef Rischin D, Peters LJ, O’Sullivan B, Giralt J, Fisher R, Yuen K, Trotti A, Bernier J, Bourhis J, Ringash J, Henke M, Kenny L (2010) Tirapazamine, cisplatin, and radiation versus cisplatin and radiation for advanced squamous cell carcinoma of the head and neck (TROG 02.02, HeadSTART): a phase III trial of the Trans-Tasman Radiation Oncology Group. J Clin Oncol 28:2989–2995PubMedCrossRef
110.
Zurück zum Zitat DiSilvestro PA, Ali S, Craighead PS, Lucci JA, Lee YC, Cohn DE, Spirtos NM, Tewari KS, Muller C, Gajewski WH, Steinhoff MM, Monk BJ (2014) Phase III randomized trial of weekly cisplatin and irradiation versus cisplatin and tirapazamine and irradiation in stages IB2, IIA, IIB, IIIB, and IVA cervical carcinoma limited to the pelvis: a Gynecologic Oncology Group study. J Clin Oncol 32:458–464PubMedCentralPubMedCrossRef DiSilvestro PA, Ali S, Craighead PS, Lucci JA, Lee YC, Cohn DE, Spirtos NM, Tewari KS, Muller C, Gajewski WH, Steinhoff MM, Monk BJ (2014) Phase III randomized trial of weekly cisplatin and irradiation versus cisplatin and tirapazamine and irradiation in stages IB2, IIA, IIB, IIIB, and IVA cervical carcinoma limited to the pelvis: a Gynecologic Oncology Group study. J Clin Oncol 32:458–464PubMedCentralPubMedCrossRef
111.
Zurück zum Zitat Brown JM (2012) Imaging tumor sensitivity to a bioreductive prodrug: two for the price of one! Clin Cancer Res 18:1487–1489PubMedCrossRef Brown JM (2012) Imaging tumor sensitivity to a bioreductive prodrug: two for the price of one! Clin Cancer Res 18:1487–1489PubMedCrossRef
112.
Zurück zum Zitat Peters LJ, O’Sullivan B, Giralt J, Fitzgerald TJ, Trotti A, Bernier J, Bourhis J, Yuen K, Fisher R, Rischin D (2010) Critical impact of radiotherapy protocol compliance and quality in the treatment of advanced head and neck cancer: results from TROG 02.02. J Clin Oncol 28:2996–3001PubMedCrossRef Peters LJ, O’Sullivan B, Giralt J, Fitzgerald TJ, Trotti A, Bernier J, Bourhis J, Yuen K, Fisher R, Rischin D (2010) Critical impact of radiotherapy protocol compliance and quality in the treatment of advanced head and neck cancer: results from TROG 02.02. J Clin Oncol 28:2996–3001PubMedCrossRef
113.
Zurück zum Zitat Le QT, Fisher R, Oliner KS, Young RJ, Cao H, Kong C, Graves E, Hicks RJ, McArthur GA, Peters L, O’Sullivan B, Giaccia A, Rischin D (2012) Prognostic and predictive significance of plasma HGF and IL-8 in a phase III trial of chemoradiation with or without tirapazamine in locoregionally advanced head and neck cancer. Clin Cancer Res 18:1798–1807PubMedCentralPubMedCrossRef Le QT, Fisher R, Oliner KS, Young RJ, Cao H, Kong C, Graves E, Hicks RJ, McArthur GA, Peters L, O’Sullivan B, Giaccia A, Rischin D (2012) Prognostic and predictive significance of plasma HGF and IL-8 in a phase III trial of chemoradiation with or without tirapazamine in locoregionally advanced head and neck cancer. Clin Cancer Res 18:1798–1807PubMedCentralPubMedCrossRef
114.
Zurück zum Zitat Lim AM, Rischin D, Fisher R, Cao H, Kwok K, Truong D, McArthur GA, Young RJ, Giaccia A, Peters L, Le QT (2012) Prognostic significance of plasma osteopontin in patients with locoregionally advanced head and neck squamous cell carcinoma treated on TROG 02.02 phase III trial. Clin Cancer Res 18:301–307PubMedCentralPubMedCrossRef Lim AM, Rischin D, Fisher R, Cao H, Kwok K, Truong D, McArthur GA, Young RJ, Giaccia A, Peters L, Le QT (2012) Prognostic significance of plasma osteopontin in patients with locoregionally advanced head and neck squamous cell carcinoma treated on TROG 02.02 phase III trial. Clin Cancer Res 18:301–307PubMedCentralPubMedCrossRef
115.
Zurück zum Zitat Mack PC, Redman MW, Chansky K, Williamson SK, Farneth NC, Lara PN Jr, Franklin WA, Le QT, Crowley JJ, Gandara DR, Swog (2008) Lower osteopontin plasma levels are associated with superior outcomes in advanced non-small-cell lung cancer patients receiving platinum-based chemotherapy: SWOG Study S0003. J Clin Oncol 26:4771–4776PubMedCentralPubMedCrossRef Mack PC, Redman MW, Chansky K, Williamson SK, Farneth NC, Lara PN Jr, Franklin WA, Le QT, Crowley JJ, Gandara DR, Swog (2008) Lower osteopontin plasma levels are associated with superior outcomes in advanced non-small-cell lung cancer patients receiving platinum-based chemotherapy: SWOG Study S0003. J Clin Oncol 26:4771–4776PubMedCentralPubMedCrossRef
116.
Zurück zum Zitat Rischin D, Hicks RJ, Fisher R, Binns D, Corry J, Porceddu S, Peters LJ, Trans-Tasman Radiation Oncology Group S (2006) Prognostic significance of [18F]-misonidazole positron emission tomography-detected tumor hypoxia in patients with advanced head and neck cancer randomly assigned to chemoradiation with or without tirapazamine: a substudy of Trans-Tasman Radiation Oncology Group Study 98.02. J Clin Oncol 24:2098–2104CrossRef Rischin D, Hicks RJ, Fisher R, Binns D, Corry J, Porceddu S, Peters LJ, Trans-Tasman Radiation Oncology Group S (2006) Prognostic significance of [18F]-misonidazole positron emission tomography-detected tumor hypoxia in patients with advanced head and neck cancer randomly assigned to chemoradiation with or without tirapazamine: a substudy of Trans-Tasman Radiation Oncology Group Study 98.02. J Clin Oncol 24:2098–2104CrossRef
117.
Zurück zum Zitat Trinkaus ME, Hicks RJ, Young RJ, Peters LJ, Solomon B, Bressel M, Corry J, Fisher R, Binns D, McArthur GA, Rischin D (2014) Correlation of p16 status, hypoxic imaging using [18F]-misonidazole positron emission tomography and outcome in patients with loco-regionally advanced head and neck cancer. J Med Imaging Radiat Oncol 58:89–97PubMedCrossRef Trinkaus ME, Hicks RJ, Young RJ, Peters LJ, Solomon B, Bressel M, Corry J, Fisher R, Binns D, McArthur GA, Rischin D (2014) Correlation of p16 status, hypoxic imaging using [18F]-misonidazole positron emission tomography and outcome in patients with loco-regionally advanced head and neck cancer. J Med Imaging Radiat Oncol 58:89–97PubMedCrossRef
118.
Zurück zum Zitat Hicks KO, Pruijn FB, Sturman JR, Denny WA, Wilson WR (2003) Multicellular resistance to tirapazamine is due to restricted extravascular transport: a pharmacokinetic/pharmacodynamic study in HT29 multicellular layer cultures. Cancer Res 63:5970–5977PubMed Hicks KO, Pruijn FB, Sturman JR, Denny WA, Wilson WR (2003) Multicellular resistance to tirapazamine is due to restricted extravascular transport: a pharmacokinetic/pharmacodynamic study in HT29 multicellular layer cultures. Cancer Res 63:5970–5977PubMed
119.
Zurück zum Zitat Chitneni SK, Bida GT, Yuan H, Palmer GM, Hay MP, Melcher T, Wilson WR, Zalutsky MR, Dewhirst MW (2013) 18F-EF5 PET imaging as an early response biomarker for the hypoxia-activated prodrug SN30000 combined with radiation treatment in a non-small cell lung cancer xenograft model. J Nucl Med 54:1339–1346PubMedCentralPubMedCrossRef Chitneni SK, Bida GT, Yuan H, Palmer GM, Hay MP, Melcher T, Wilson WR, Zalutsky MR, Dewhirst MW (2013) 18F-EF5 PET imaging as an early response biomarker for the hypoxia-activated prodrug SN30000 combined with radiation treatment in a non-small cell lung cancer xenograft model. J Nucl Med 54:1339–1346PubMedCentralPubMedCrossRef
120.
Zurück zum Zitat Anderson RF, Yadav P, Patel D, Reynisson J, Tipparaju SR, Guise CP, Patterson AV, Denny WA, Maroz A, Shinde SS, Hay MP (2014) Characterisation of radicals formed by the triazine 1,4-dioxide hypoxia-activated prodrug, SN30000. Org Biomol Chem 12:3386–3392PubMedCrossRef Anderson RF, Yadav P, Patel D, Reynisson J, Tipparaju SR, Guise CP, Patterson AV, Denny WA, Maroz A, Shinde SS, Hay MP (2014) Characterisation of radicals formed by the triazine 1,4-dioxide hypoxia-activated prodrug, SN30000. Org Biomol Chem 12:3386–3392PubMedCrossRef
121.
Zurück zum Zitat Patterson AV, Silva S, Guise C, Abbattista M, Bull M, Hsu A, Sun J, Jung D, Grey A, Ashoorzadeh A, Anderson R, Smaill JB (2015) The hypoxia activated EGFR-TKI TH-4000 overcomes erlotinib-resistance in preclinical NSCLC models at plasma levels achieved in phase 1 clinical trial. Cancer Res 75: Abstract number 5358 Patterson AV, Silva S, Guise C, Abbattista M, Bull M, Hsu A, Sun J, Jung D, Grey A, Ashoorzadeh A, Anderson R, Smaill JB (2015) The hypoxia activated EGFR-TKI TH-4000 overcomes erlotinib-resistance in preclinical NSCLC models at plasma levels achieved in phase 1 clinical trial. Cancer Res 75: Abstract number 5358
122.
Zurück zum Zitat Patterson AV, Silva S, Guise C, Bull M, Abbattista M, Hsu A, Sun JD, Hart CP, Pearce TE, Smaill JB (2015) TH-4000, a hypoxia-activated EGFR/Her2 inhibitor to treat EGFR-TKI resistant T790M-negative NSCLC. J Clin Oncol 33: abstract e13548 Patterson AV, Silva S, Guise C, Bull M, Abbattista M, Hsu A, Sun JD, Hart CP, Pearce TE, Smaill JB (2015) TH-4000, a hypoxia-activated EGFR/Her2 inhibitor to treat EGFR-TKI resistant T790M-negative NSCLC. J Clin Oncol 33: abstract e13548
123.
Zurück zum Zitat Cazares-Korner C, Pires IM, Swallow ID, Grayer SC, O’Connor LJ, Olcina MM, Christlieb M, Conway SJ, Hammond EM (2013) CH-01 is a hypoxia-activated prodrug that sensitizes cells to hypoxia/reoxygenation through inhibition of Chk1 and Aurora A. ACS Chem Biol 8:1451–1459PubMedCentralPubMedCrossRef Cazares-Korner C, Pires IM, Swallow ID, Grayer SC, O’Connor LJ, Olcina MM, Christlieb M, Conway SJ, Hammond EM (2013) CH-01 is a hypoxia-activated prodrug that sensitizes cells to hypoxia/reoxygenation through inhibition of Chk1 and Aurora A. ACS Chem Biol 8:1451–1459PubMedCentralPubMedCrossRef
124.
Zurück zum Zitat Lindquist KE, Cran JD, Kordic K, Chua PC, Winters GC, Tan JS, Lozada J, Kyle AH, Evans JW, Minchinton AI (2013) Selective radiosensitization of hypoxic cells using BCCA621C: a novel hypoxia activated prodrug targeting DNA-dependent protein kinase. Tumour Microenvironment and Therapy 1:46–55 Lindquist KE, Cran JD, Kordic K, Chua PC, Winters GC, Tan JS, Lozada J, Kyle AH, Evans JW, Minchinton AI (2013) Selective radiosensitization of hypoxic cells using BCCA621C: a novel hypoxia activated prodrug targeting DNA-dependent protein kinase. Tumour Microenvironment and Therapy 1:46–55
125.
Zurück zum Zitat Zhu R, Baumann RP, Penketh PG, Shyam K, Sartorelli AC (2013) Hypoxia-selective O6-alkylguanine-DNA alkyltransferase inhibitors: design, synthesis, and evaluation of 6-(benzyloxy)-2-(aryldiazenyl)-9H-purines as prodrugs of O6-benzylguanine. J Med Chem 56:1355–1359PubMedCentralPubMedCrossRef Zhu R, Baumann RP, Penketh PG, Shyam K, Sartorelli AC (2013) Hypoxia-selective O6-alkylguanine-DNA alkyltransferase inhibitors: design, synthesis, and evaluation of 6-(benzyloxy)-2-(aryldiazenyl)-9H-purines as prodrugs of O6-benzylguanine. J Med Chem 56:1355–1359PubMedCentralPubMedCrossRef
126.
Zurück zum Zitat Karnthaler-Benbakka C, Groza D, Kryeziu K, Pichler V, Roller A, Berger W, Heffeter P, Kowol CR (2014) Tumor-targeting of EGFR inhibitors by hypoxia-mediated activation. Angew Chem Int Ed Engl 53:12930–12935PubMedCentralPubMedCrossRef Karnthaler-Benbakka C, Groza D, Kryeziu K, Pichler V, Roller A, Berger W, Heffeter P, Kowol CR (2014) Tumor-targeting of EGFR inhibitors by hypoxia-mediated activation. Angew Chem Int Ed Engl 53:12930–12935PubMedCentralPubMedCrossRef
128.
Zurück zum Zitat Karakashev SV, Reginato MJ (2015) Progress toward overcoming hypoxia-induced resistance to solid tumor therapy. Cancer Manag Res 7:253–264PubMedCentralPubMed Karakashev SV, Reginato MJ (2015) Progress toward overcoming hypoxia-induced resistance to solid tumor therapy. Cancer Manag Res 7:253–264PubMedCentralPubMed
Metadaten
Titel
Targeting the hypoxic fraction of tumours using hypoxia-activated prodrugs
verfasst von
Roger M. Phillips
Publikationsdatum
01.03.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 3/2016
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-015-2920-7

Weitere Artikel der Ausgabe 3/2016

Cancer Chemotherapy and Pharmacology 3/2016 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.