Abstract
Significance: There are a number of redox-active anticancer agents currently in development based on the premise that altered redox homeostasis is necessary for cancer cell's survival.
Recent Advances: This review focuses on the relatively few agents that target cellular redox homeostasis to have entered clinical trial as anticancer drugs.
Critical Issues: The success rate of redox anticancer drugs has been disappointing compared to other classes of anticancer agents. This is due, in part, to our incomplete understanding of the functions of the redox targets in normal and cancer tissues, leading to off-target toxicities and low therapeutic indexes of the drugs. The field also lags behind in the use biomarkers and other means to select patients who are most likely to respond to redox-targeted therapy.
Future Directions: If we wish to derive clinical benefit from agents that attack redox targets, then the future will require a more sophisticated understanding of the role of redox targets in cancer and the increased application of personalized medicine principles for their use. Antioxid. Redox Signal. 26, 262–273.
Keywords: : redox, signaling, cancer, therapy, clinical
Introduction
A balance between oxidants and antioxidants exists under physiological conditions even though reactive oxygen species (ROS) are continuously generated. The maintenance of redox homeostasis is accomplished by elimination of ROS by scavenging systems. The two main sources of ROS are the mitochondria and NADPH oxidases (76). Other ROS producers are the cytochrome P450 system, xanthine oxidase, and nitric oxide (NO) synthase. ROS are eliminated by intracellular enzymes, including superoxide dismutase (SOD), glutathione (GSH) peroxidase, catalase, thioredoxin (Trx) reductase, and glutathione S-transferase (GST). Cells maintain a redox balance, and under normal conditions, low levels of locally produced ROS act as mitogens driving cell survival and proliferation. Oxidative stress and aberrant redox signaling result from a loss of this redox balance and can lead to cancer development because of resulting DNA mutations, genomic instability, and protumorigenic signaling (1). Cancer cells can also become dependent on high levels of ROS formation required for an altered redox environment that maintains the procancerous state, thus presenting an “Achilles Heel” for developing treatment options based on the loss of normal redox homeostatsis (76). The clinical outcome of approaches that attempt to exploit this cancer redox vulnerability will be discussed in the review in the context of mechanisms of redox regulation described below.
Mechanisms of Redox Regulation
Mitochondria drive a number of cellular functions from metabolism to apoptosis, while providing the cell's energy needs (52). Redox signaling molecules emanating from ROS generated by the mitochondria are H2O2, NO, and superoxide, or other less significant naturally occurring redox species such as peroxynitrite, prostaglandin-like molecules, and 4-hydroxynonenal. To generate redox signals, they must bring about reversible change in the activity of a protein. Generally, this involves modification of cysteine residues (17).
S-oxidation (cysteine oxidation)
S-glutathionylation and S-nitrosylation are processes that regulate key mitochondrial functions, including nutrient oxidation, oxidative phosphorylation, ROS production, the mitochondrial permeability transition, apoptosis, and mitochondrial fission and fusion (61). All modifications act as redox switches, altering function and enabling proteins to respond to the reduction potential of a particular redox couple.
The Trx system
The cytoplasmic Trx system comprising thioredoxin 1 (Trx1), thioredoxin reductase 1 (TrxR-1), and NADPH, one of the major antioxidant systems, maintains intracellular redox homeostasis through reversible cysteine modification (81). Trx1 regulates redox-sensitive transcription factors by acting as a cofactor to reduce specific cysteine residues (58, 53) and acts as a growth factor that stimulates cancer cell proliferation (8, 77). The Trx system maintains cellular redox homeostasis by scavenging ROS and regulating redox enzymes. There is also a mitochondrial Trx system comprising Trx-2 and TrxR-2 that attenuates some of the effects of Trx-1 and increases in mitochondrial ROS (116). The absence of Trx-1 in homozygous null mice leads to embryonic lethality and of Trx-2 to embryonic lethality at the time of maturation of the mitochondria, both associated with a massive increase in apoptosis (68). Expression of Trx1 and TrxR and activity of TrxR is upregulated in many human cancers resulting in proliferation, survival, chemoresistance, and inhibition of apoptosis (7).
The glutathione system
Glutathione/Glutaredoxin (GRX) maintain cellular redox through S-glutathionylation and reduction, although this is dependent on Trx redox regulation (63). GRX, a small dithiol protein, regulates certain enzyme activities via a disulfide exchange reaction and when overexpressed protects cells from H2O2-induced apoptosis. Elevated GSH levels are observed in various types of tumors, making them more resistant to chemotherapy (14, 107). Removing this molecular shield has been an approach to reinstate death signals driving cancer cells to apoptosis.
NO metabolism
NO metabolism modifies protein thiol groups to S-nitrosothiols, which may then be further modified to disulfide, sulfenic acid, or S-GSH-modified protein all referred to as redox switches (67). These posttranslational modifications are reversible and able to react sensitively to levels of ROS (17).
Redox transcription factors
There are a number of reported redox-sensitive transcription factors (Table 1) (13, 23).
Table 1.
Transcription Factor | Redox Regulator | Consequence |
---|---|---|
AP-1 | Trx-1, nucleoredoxin | Enhanced DNA binding |
β-catenin | Trx-1,nucleoredoxin, Ref-1/APE | Enhanced DNA binding |
Egr-1 | Ref-1/APE | Enhanced DNA binding |
FOXO | Cys oxidation PTP1b | Block nuclear export |
GR | Trx-1 | Enhanced DNA binding |
HIF-1 | Trx-1, APE/Ref-1 | HIF-1 stability, HIF-1 activation |
HSF1 | H2O2 | Altered DNA binding |
TP53 | Trx-1, APE/Ref-1 | Enhanced DNA binding |
NF-κB | Trx-1, APE/Ref-1 | Enhanced DNA binding |
NRF2 | Trx-1 | Prevent degradation |
Sp1 | Trx-1 | Enhanced DNA binding |
TTF | APE/Ref-1 | Enhanced DNA binding |
Drugs Targeting Redox Systems and Redox Regulators
The drugs that we describe in this review have been explored clinically for targeting redox processes and are summarized in Table 2.
Table 2.
Drug | Target | Clinical activity | Statusa |
---|---|---|---|
Aurofin | Thioredoxin reductase | Ongoing trials |
NCT02089048 NCT01419691 I NCT01737502 I |
Motexafin gadolinium | Thioredoxin reductase, ribonucleotide reductase | Radio/chemosensitizer | Not approved by FDA |
Motexafin lutetium | Apoptosis | Photosensitization | No longer being developed |
PX-12 | Thioredoxin, HIF-1 | Stable disease | Iv use no longer pursued |
Arsenic trioxide | Mitochondria, Trx-1/-2,GSH | FDA approved for APL |
NCT00275067 NCT00661544 NCT00258245 |
Buthionine sulfoximine | γ-glutamylcysteine ligase (GSH) | Chemosensitizer no clinical advantage |
No longer being developed |
Telcyta (TLK-286) | Glutathione S-transferase -ρi | Chemosensitizer no clinical advantage |
Development status unknown |
Telintra (TLK199) | Glutathione S-transferase -pi | Chemosensitizer | Development status unknown |
Disulfiram | Acetaldehyde dehydrogenase, GSH/GSSG | Chemosensitizer no clinical advantage |
No longer being developed |
NOV-002 | γ-glutamyl-transpeptidase, GSH/GSSG | Chemosensitizer no clinical advantage |
No longer being developed |
N-acetylcysteine | GSH prodrug | Decrease side effects, no clinical advantage |
NCT02241876, |
PX-478 | HIF-1 | Phase I trial only | Not currently being developed |
EZN-2968 | HIF-1 | Lowers HIF-1 | NCT02564614 |
BAY 87-2243 | HIF-1/2, mitochondrial complex-1 | Toxic at doses used | development status unknown |
Topotecan | topoisomerase I (HIF-1) | Trend to lower HIF-1, a partial tumor response | FDA-approved drug, development status as HIF-1 inhibitor unknown |
EZN-2208 | Topoisomerase I (HIF-1) | HIF-1 activity unknown, no clinical survival advantage | Company not developing clinical products |
Bortezomib | Proteasome (NF-κB) | Significant clinical activity, not known if NF-κB related |
FDA-approved drug |
Fenretinide | Retinoic acid receptor agonist, increases ROS | Induces apoptosis, not known if activity related to ROS |
NCT01535157 NCT00646230 |
α-Tocopherol | Free radical scavenger | Large-scale clinical cancer prevention trials show no clinical benefit | NCT02397486 |
Vitamin C | Free radical scavenger, H2O2 prodrug | No convincing evidence of clinical benefit | Development of high-dose iv vitamin C continues |
Curcumin | Free radical scavenger | No convincing evidence of clinical advantage | Development of more bioavailable forms continues |
Refers to an ongoing clinical trial from the NIH Clinical Trials database at ClinicalTrials.gov.
APL, acute promyelocytic leukemia; FDA, Food and Drug Administration; GSH, glutathione; GSSG, oxidized glutathione; ROS, reactive oxygen species.
Trx-Trx reductase inhibitors
Auranofin
Since the selenoenzyme thioredoxin reductase (TrxR) is a putative target for cytotoxic gold complexes, a group of structurally diverse gold(III) compounds, including the clinically used antiarthritic gold(I) drugs auranofin, approved for the treatment of rheumatoid arthritis in 1985, and aurothiomalate, were investigated for the mechanism of TrxR inhibition (33). All gold(III) or gold(I) compounds were found to produce potent enzyme inhibition but only after prereduction of TrxR with NADPH. TrxR inhibition was attributed to structural modification occurring, upon cofactor binding, with auranofin's gold(I) coordination to the active site selenocysteine (29, 31, 62, 75). An alternate mechanism has been described for gold(III) compounds through oxidative protein damage and inhibition of the ubiquitin–proteasome system by targeting deubiquitinases involved in cell cycle regulation, protein degradation, gene expression, and DNA repair (15, 57). Both these mechanisms of action result in apoptosis. Although the use of auranofin for the treatment of rheumatoid arthritis is declining because of adverse effects associated with its long-term use (90), recent clinical trials have been initiated exploring its use as an anticancer agent. A pharmacokinetic Phase I, open-label study in healthy adult subjects was initiated in 2014 to determine terminal phase pharmacokinetic parameters in blood and gold measurements in stool samples (NCT02089048).
Auranofin has also been evaluated in a number of Phase II studies as a single agent or in combination with chemotherapy. A Phase I and II study to evaluate the safety and effectiveness of auranofin to treat patients with chronic lymphocytic leukemia, small lymphocytic lymphoma, or prolymphocytic lymphoma is awaiting study results (NCT01419691). An ongoing Phase I/II clinical trial initiated in 2012 is examining the combination of sirolimus (rapamycin) and auranofin in patients with advanced or recurrent nonsmall-cell lung cancer or small-cell lung cancer (NCT01737502).
Texaphyrins
Motexafin gadolinium (MGd, Xcytrin) an inhibitor of TrxR and ribonucleotide reductase, was developed by Pharmacyclics as a radiosensitizing agent initially for use in brain metastases (60). Although the mechanism of sensitization remains unclear, it has been proposed that MGd is endocytosed by the clathrin-dependent pathway producing high concentrations in malignant cells, which along with the synergistic effect of irradiation causes apoptosis (5, 10). MGd was evaluated in a number of Phase II trials for lymphomas, multiple myeloma, renal cell cancer, glioblastoma multiforme with radiation, and in NSCLC with chemotherapy (65). Pharmacyclics completed a multicenter US Phase III clinical trial of MGd in NSCLC patients with metastatic tumors of the brain who require whole brain radiotherapy (NCT00054795), but it did not receive Food and Drug Administration (FDA) approval.
Antrin® (motexafin lutetium), another texaphyrin by Pharmacyclics (94), was developed for photodynamic therapy (photoangioplasty) to promote a redox-sensitive apoptotic cell death pathway in vascular cells for the reduction of atherosclerosis involving peripheral arteries. It has been reported to preferentially accumulate in tumor cells and absorb light, forming an extended high-energy conformational state producing high quantum yields of singlet oxygen and a local cytotoxic effect. A number of clinical trials were initiated and terminated (NCT00005067, NCT00005808, NCT00087191) and this agent is no longer being developed.
PX-12
The thioredoxin inhibitor, PX-12 (1-methylpropyl 2-imidazolyl disulfide), has shown both an in vitro and promising in vivo antitumor activity (54). Irreversible thio-alkylation of the Cys73 thioredoxin residue, by PX-12, was shown to result in inhibition of Trx redox activity and subsequent inhibition of the hypoxia-induced increase in HIF-1α protein and vascular endothelial growth factor (VEGF) secretion (110, 111). This inhibition is accompanied by the rapid metabolism of PX-12 producing two inactive metabolites, volatile 2-butanethiol and 2-mercaptoimidazole. PX-12, the first Trx-1 inhibitor to enter clinical trials, was administered as a 1-h intravenous (IV) infusion (79). The infusion time was subsequently increased to 3, 24, and 72 h (9, 78, 80) to overcome lung irritation that was observed at the higher dose levels due to expulsion of the volatile metabolite (9). Although there were no objective responses based on response evaluation criteria in solid tumor criteria in the first Phase I trial with 1- and 3-h infusions, seven (18%) patients achieved durable stable disease, receiving at least six cycles of therapy (range, 6–14 cycles). These included two patients with sarcoma and one patient each with colorectal, appendiceal, and renal cancer. A patient with appendiceal adenocarcinoma had a minor response of 322 days (79).
PX-12 was also investigated in a Phase II trial as a 3-h infusion at two dose levels in patients with previously treated advanced pancreatic cancer (78). Plasma Trx-1 levels were found elevated in only 3 of 28 (11%) patients screened for study. Although the therapy was well tolerated, the best response was stable disease in two patients with unexpectedly low baseline Trx-1. The development of P-12 as an IV agent has been terminated.
Arsenic trioxide (ATO)
The traditional Chinese medicine Pi Shuang or arsenic trioxide [As2O3 (ATO)] was and is still used to treat cancer and other conditions (11, 36). Work in the 1970s to investigate the potential of ATO to treat acute promyelocytic leukemia (82, 114) led to the approval of ATO (TRISENOX) by the FDA for the treatment of leukemia in 2000 (82). Several mechanisms have been proposed to explain its anticancer activity, which occurs mainly through mitochondrial dysfunction, as it was determined that ATO could lower both reduced and oxidized forms of thioredoxin 1 (Trx1) with no effect on Trx1 redox potential (115). The result is altered cellular redox homeostasis through hydrogen peroxide generation, GSH depletion, and Trx-1 downregulation (59). Lu et al. determined that ATO irreversibly inhibited mammalian TrxR, with both the N-terminal redox-active dithiol and the C-terminal selenothiol-active participating in the reaction (43). Numerous clinical studies have investigated ATO use as a single agent in the treatment of solid tumors, however, therapeutic efficacy has been limited (100). Because of this, the combination of ATO with other clinically used chemotherapeutic drugs has been evaluated to improve its therapeutic efficacy in treating human solid tumors. There are currently a number of ongoing clinical trials of ATO in combination with chemotherapy in solid tumors, including in malignant glioma (NCT00275067) and in multiple myeloma (NCT00661544, NCT00258245).
GSH inhibitors
Buthionine sulfoximine
Elevated GSH has been associated with cancer cell resistance to various cytotoxic agents (14, 15). Thus, lowering GSH has been sought by specific inhibition of γ-glutamylcysteine ligase, a key enzyme of GSH biosynthesis. Such an inhibitor, L-S,R buthionine sulfoximine (BSO) (38), was shown to enhance the cytotoxicity to cancer cells of chemotherapeutic agents experimentally both in vitro and in vivo (42, 95). Early clinical trials of BSO evaluated its ability to enhance the cytotoxicity of chemotherapeutic agents (6). A Phase I dose escalation trial of IV BSO administered as a short infusion every 12 h, with and without the alkylating drug L-PAM, showed that BSO, alone and in combination with L-PAM, could be safely given to patients, but that a schedule of short infusions every 12 h did not result in GSH depletion <30% of baseline values (7). In subsequent clinical studies, BSO was evaluated in combination with melphalan in children with neuroblastoma (NCT00005835) (2, 3) and patients with persistent or recurrent malignant melanoma (NCT00661336), which was not completed due to lack of patients. Ultimately, BSOs short half-life (96) led to focused attempts to find alternatives for modification of GSH levels.
Telcyta
Telcyta (TLK-286), a GSH analogue that inhibits glutathione S-transferase pi isoform (GST P1-1), has been tested clinically in combination with platinum, taxanes, and anthracyclines in tumors with very high GST-PI-1 levels (102). It is a prodrug that is activated by GST P1-1 to produce an anticancer alkylating agent and releases a glutathione derivative (25). Telcyta has been tested in Phase II and III clinical trials for the treatment of ovarian, nonsmall cell lung and breast cancer, as a single agent and in combination with other chemotherapeutic agents. NCT00350948, a Phase III randomized study of Telcyta and doxorubicin in platinum refractory ovarian cancer (ASSIST-5), was terminated after data provided showed that women treated with Telcyta had poorer outcomes than those treated with standard of care.
Telintra (TLK199), an inhibitor of GST-P1-1, was granted orphan status and approved to be evaluated in a Phase III study for the treatment of myelodisplastic syndrome (MDS). Other exploratory studies have been discontinued. Telintra, whose inhibition of GST-PI-1 was shown to lead to activation of Jun kinase (91), was shown to cause clinically significant and sustained reduction in red blood cell transfusion responses in MDS patients (87, 88). Telintra has been evaluated in a number of Phase 1 and 2 trials as both a liposomal preparation for injection (NCT00035867) and an oral tablet (NCT00701870), although a number of trials have been terminated due to lack of enrollment. Hematologic improvement was observed in patients who had failed or progressed following a range of prior therapies and supportive care regimens. In 2014, Telik, the company developing Telintra, merged into MabVax and the current development status of Telintra is unknown.
Disulfiram
Disulfiram (1,1′,1′′,1′′′-[disulfanediylbis(carbonothioylnitrilo)]tetraethane, Antabuse), discovered almost 100 years ago as an agent that inhibits acetaldehyde dehydrogenase resulting in an immediate hangover when alcohol is ingested and used as a support for the treatment of chronic alcoholism, also shifts the ratio of GSH:oxidized glutathione (GSSG) to the oxidized state and induces apoptosis (12). It has been evaluated in a clinical trial for metastatic melanoma (NCT00256230) in a study completed in 2011. Another study with disulfiram in combination with ATO (NCT00571116) was halted due to slow accrual and lack of study findings.
NOV-002
A unique antitumor agent NOV-002, a stabilized formulation of GSSG and cisplatin at a ratio of 1000:1, (developed by Novelos and now Cellectar Biosciences, Inc.), has been reported to alter the GSH:GSSG ratio by acting as a competitive substrate for gamma-glutamyl-transpeptidase (GGT) (105). GGT catalyzes the transfer of the gamma-glutamyl moiety of GSH to an acceptor protein to give S-glutathionylation proteins. NOV-002 has been reported to inhibit tumor cell proliferation, survival, and invasion, and in some settings can ameliorate cytotoxic chemotherapy-induced hematopoietic and immune suppression through its ability to modulate cellular redox by upregulation of the expression of SOD-3 and glutathione peroxidase-2 (21). NOV-002 has been studied in combination with standard chemotherapy for Stage IIIb/IV NSCLC in Phase III (NCT00347412) and in a neoadjuvant breast cancer setting in Phase II (NCT00499122). In a neoadjuvant trail of women with newly diagnosed Stage IIB or IIIC breast cancer, NOV-002 in combination with doxorubicin and cyclophosphamide followed by docetaxel met the primary endpoint of a pathologic complete response of 38%. Unfortunately, in a study of 903 NSCLC patients, NOV-002 and carboplatin failed to improve overall survival compared to the paclitaxel and carboplatin alone. In another study of NOV-002 in combination with carboplatin in chemotherapy-resistant ovarian cancer (NCT00345540) failed to reach its endpoint of ≥2 responses and its development has been discontinued.
N-acetylcysteine
N-acetylcysteine (NAC; Mucomyst) is a prodrug of l-cysteine, a precursor of GSH, and hence, particularly during times of oxidative stress (20), has been shown to support the body's antioxidant and NO systems (64). Because de novo synthesis is the primary mechanism by which glutathione is replenished (22), NAC has been tested in a number of clinical trials with the objective of improving cancer outcomes (64). NAC have been evaluated for ability to suppress colon polyps (28), as an adjunct to standard therapy in the eradication of Helicobacter pylori (NCT01109381; the study was terminated due to poor efficacy) (41), and in Phase II and III studies to explore if NAC could decrease the risk of ototoxicity in patients on hemodialysis who are receiving gentamicin (NCT01271088, NCT01131468) (30). In addition, it is currently being explored for its ability to attenuate cisplatin-induced toxicities (NCT02241876) and its ability to reduce mucositis in head and neck patients receiving radiation therapy (NCT02123511). However, a 2-year supplementation of NAC, with or without retinyl palmitate in more than 2500 patients, resulted in no benefit in terms of survival, event-free survival, or second primary tumors for patients with head and neck cancer or with lung cancer, most of whom were previous or current smokers (108).
HIF inhibitors
PX-478
A number of agents have been studied clinically that are reported to inhibit HIF-1 activity directly or indirectly (51). PX-478, a small molecule that downregulates hypoxia-induced increase as well as constitutive expression of HIF-1α and HIF-1 transcription factor activity (101, 112), was evaluated in an open-label, oral dose escalation Phase I trial in patients with advanced cancers to examine safety, tolerability, pharmacokinetics, pharmacodynamic, and antitumor activity (NCT00522652) (104). Pharmocodynamic studies revealed dose-proportional inhibition of HIF-1α levels. Adverse events included gastrointestinal (GI) nausea, diarrhea, and vomiting, as well as fatigue. One patient experienced prolonged Grade 3 thrombocytopenia at the highest dose level. Pharmacokinetic analyses showed evidence of conversion of PX-478 to melphalan consistent with its anticipated mechanism of HIF-1α inhibition.
EZN-2968, a locked nucleic acid antisense oligonucleotide inhibitor of HIF-1α that causes downregulation of HIF-1α mRNA and protein, was studied as a 2-h iv infusion in a pilot trial in patients with refractory solid tumors (NCT00466583) (49). The purpose of the study was to evaluate modulation of HIF-1 mRNA and protein levels as well as antitumor response. Of 10 patients enrolled, one patient with duodenal neuroendocrine tumor had prolonged stabilization of disease (24 weeks). Four of six patients with paired tumor biopsies showed reduction in HIF-1 mRNA levels compared to baseline, while reduction in HIF-1 protein and mRNA levels of some target genes was observed in two patients. The trial provided preliminary proof-of-concept for modulation of HIF-1α mRNA and protein expression. EZN-2968 was acquired by Santaris and subsequently by ROCHE (RO7070179) and is now being evaluated in a Phase Ib proof-of-mechanism study in adults with hepatocellular carcinoma (HCC) (NCT02564614). The primary outcome of this study that began enrolling patients in November 2015 is a change from baseline of HIF-1α mRNA levels in tumor tissue.
BAY 87-2243, a small molecule that inhibits HIF-1α and HIF-2α protein accumulation under hypoxic conditions in vitro, inhibits mitochondrial complex I activity but has no effect on complex III activity. Under conditions of glucose depletion that favor mitochondrial ATP generation in cancer cells as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range (27). BAY 87-2243 interference with mitochondrial function resulted in reduced hypoxia-induced HIF-1 activity in tumors and prompted exploration of its use with radiation as a therapeutic approach to overcome chemo- and radiotherapy resistance of hypoxic tumors (45). It was found that BAY 87-2243 markedly decreased nuclear HIF-1α expression and pimonidazole hypoxic fraction (a measure of tumor hypoxia); however, its application before, after, or during RT did not improve local tumor control demonstrating that its radiosensitizing effect depends on treatment schedule. An open-label, Phase I study of BAY 87-2243 given orally once daily in subjects with advanced malignancies treated five subjects but was prematurely discontinued due to adverse GI events and it was determined that BAY 87-2243 was not tolerated at the doses used.
Topotecan
Topotecan (TPT), an FDA-approved camptothecin analogue that poisons topoisomerase I generating double-strand DNA breaks currently used as second-line therapy for patients with small-cell lung cancer or ovarian cancer, was found in a high-throughput screen using a cell-based assay for HIF-1 transcriptional activity (69, 84). It was found to block HIF-1α translation by a Top1-dependent but DNA damage-independent mechanism (85, 86). In xenograft models, it was shown that administration of daily TPT in combination with the anti-VEGF antibody bevacizumab exerted a synergistic antitumor activity, providing a rationale for clinical development of this combination strategy (83). A pilot study examined daily oral TPT administered to patients with advanced solid tumors expressing HIF-1α in at least 10% of tumor cells (26, 56) (NCT00182676). In 4 of 15 patients, HIF-1α nuclear staining became undetectable after treatment and decreased levels of VEGF and glucose transporter-1 mRNA were seen in three patients. Decreased tumor blood flow and permeability were observed by dynamic contrast magnetic resonance imaging (DCE-MRI) in seven of ten patients after one cycle. One patient had a partial response accompanied by inhibition of HIF-1α in tumor and reduction in tumor blood flow on DCE-MRI. While trends in both HIF-1α modulation and radiologic correlates were seen, these were not statistically significant due to low patient numbers.
EZN-2208, a PEGylated form of the small-molecule topoisomerase 1 inhibitor SN38 (the active metabolite irinotecan; CPT-11), with improved pharmacokinetics showed good antitumor activity in preclinical models of solid tumors and lymphomas, including CPT-11-resistant tumors (73). Its antitumor activity is possibly explained by the ability of this agent to inhibit HIF-1α accumulation (93). EZN-2208 was evaluated in a Phase I clinical trial and found to have an acceptable safety profile as a 1-h infusion (74) with stable disease as the best response (NCT00520390). A Phase II trial in metastatic breast cancer demonstrated activity in patients with triple negative breast cancer and in platinum-resistant patients (70) (NCT00520390). EZN-2208 was also evaluated in combination with bevacizumab in solid tumors (NCT01251926) (50) as well as with cetuximab in metastatic colorectal cancer (NCT00931840) where it was found to be well tolerated but provided no survival advantage (34). Enzon is no longer developing any clinical products.
Nuclear factor kappa B inhibitors
Signaling by the nuclear factor kappa B (NF-kB) pathway controls the expression of many genes involved in critical physiological responses, including oxidative stress responses and apoptosis (72). NF-κB signaling can be targeted at multiple points, for example, IKK activation, IκB degradation, and NF-κB DNA binding. Most development efforts have focused on inhibitors of IKKβ, to block the phosphorylation of IκBα, preventing its degradation and maintaining NF-κB in an inactive state in the cytoplasm. In 2006, the list of NF-κB inhibitors of this pathway totaled 785 although as yet no specific NF-kB inhibitors have reached clinical studies (37). Indirect inhibitors include the proteasome inhibitor, bortezomib, and the promiscuous curcumin (below). Bortezomib (VELCADE, previously known as PS-341), a proteasome inhibitor, was one of the first compounds used to inhibit the function of NF-κB (90). Bortezomib has shown significant efficacy in hematologic and solid tumors, including multiple myeloma as well as lung, breast, prostate, pancreatic, and head-neck carcinomas, yet it is unclear if its effect are in fact mediated through the inhibition of NF-kB (47). Bortezomib in combination with temozolomide in a Phase I/II study in advance refractory solid tumors or melanoma was terminated due to lack of efficacy (NCT00512798). Other histone deacetylase inhibitors, vorinostat and romidepsin, which block the posttranslational acetylation of NF-kB to regulate its activity, are approved for treating T-cell lymphoma.
ROS generators
Fenretinide
Fenretinide (N-4-hydroxyphenyl-retinamide or 4-HPR), a semisynthetic retinoid, was initially developed as a low-dose chemopreventative agent (18). Apoptosis by fenretinide has been linked to generation of ROS. It has been shown to bind to and activate retinoic acid receptors resulting in the induction of cell differentiation and apoptosis (16). The mechanism of its induced apoptosis has been hypothesized to operate through coenzyme Q of the mitochondrial respiratory chain (101, 113). Fenretinide has also been reported to cause the de novo synthesis of ceramides associated with increase of ROS resulting in cell death through apoptosis and/or necrosis (39). A study of 14 women with breast cancer, where fenretinide was administered before scheduled biopsy, lumpectomy, or mastectomy, found that fenretinide and its major metabolite accumulated preferentially in fatty tissue of the breast versus plasma (4, 101a). The data supported its study in premenopausal/ER-negative breast cancer prevention (NCT00001378) (32). A Phase III clinical trial of fenretinide suggested that it reduces breast cancer relapse in premenopausal women and provided a significant risk reduction of second breast cancer (NCT00002646) (109). Fenretinide has also been evaluated in a number of other cancer indications and is currently being evaluated in a trial in combination with ketoconazole in recurrent ovarian cancer (NCT01535157) and in Phase I safety trial children with recurrent/resistant neuroblastoma (NCT00646230) or ALL, AML, and NHL (NCT01187810).
Dietary antioxidants
Tocopherol
Nutrition supplements and nutraceuticals with vague claims of health benefit are widely used by the general public, probably more so than antioxidants proposed to reduce levels of harmful oxidation product. Only a few have undergone rigorous clinical evaluation. Because of its ability to neutralize free radicals, the fat soluble antioxidant vitamin E has been suggested to possess anticancer activity by protecting cells against oxidative damage; yet, clinical support has not been forthcoming (99). One component of vitamin E, α-tocopherol, has been evaluated in a number of clinical settings to determine its ability to reduce oxidative stress relating to cancer (35). Through the process of neutralizing a free radical, α-tocopherol is oxidized, its antioxidant capacity is lost, and the presence of other antioxidants, such as vitamin C, is required to regenerate the antioxidant capacity of α-tocopherol (106). The consumption of 400 mg per day α-tocopherol and 400 mg ascorbate has been shown to dramatically reduce the formation of lipid-soluble fecal mutagens (24). A double-blind placebo-controlled clinical cancer prevention trial of high-dose α-tocopherol and beta-carotene for lung cancer prevention in smokers found no benefit (43, 103), but a secondary finding was a 32% reduction in the incidence of prostate cancer in men given daily supplements of synthetic α-tocopherol (46). This finding was further evaluated in a subsequent Phase III randomized controlled clinical trial funded primarily by NCI to determine if selenium and vitamin E (dl-alpha tocopheryl acetate) taken as dietary supplements alone or together could help prevent prostate cancer (NCT00006392) (44). Initially planned for a minimum of 7 years, the trial was terminated early because at a 3-year time point, more cases of prostate cancer in men taking only vitamin E were appearing. By the planned end of the study, the incidence of prostate cancer in this group was found to be significantly higher at 17% compared to placebo (55).
Vitamin E has also been tested for its ability to lessen the harmful effects of medical treatments such as dialysis and radiation and to reduce unwanted side effects of drugs such as hair loss with doxorubicin and the lung damage with amiodarone (99). A number of randomized placebo-controlled studies in adults and children found that topical vitamin E might reduce oral mucositis induced by chemotherapy or radiotherapy (NCT00311116). In addition, the incidence and severity of peripheral neurotoxicity induced by taxanes or platinum-based chemotherapy by alpha-tocopherol were found reduced in five randomized trials, although this finding was not confirmed in two subsequent randomized placebo-controlled studies (35). Hence, despite several large prospective cohort studies, each have failed to find significant associations between vitamin E intake and the risk of cancer (55) or clinical benefit; yet, studies to evaluate its ability to reduce toxicities are still continuing (NCT02397486).
Vitamin C
High-dose vitamin C given by oral and IV routs has been studied as a treatment for patients with cancer since the 1970s. However, carefully controlled clinical trials in the late 1970s failed to show that oral vitamin C had any therapeutic benefit against cancer (19). More recently, interest has revived in the use of IV vitamin C that can reach peak plasma concentrations greater that 25-fold those of oral doses, with the suggestion that vitamin C may act as a prodrug for hydrogen peroxide delivery to tumors, without the presence of hydrogen peroxide in the blood (71). A recent systematic review of the literature found no credible evidence for an antitumor effect of vitamin C in combination with chemotherapy in however form of administration (48).
Curcumin
Curcumin [1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione], the spice turmeric derived from the rhizome of Curcuma longa L, has both antioxidant and anti-inflammatory properties (66) by being able to scavenge reactive oxygen and nitrogen species in vitro (97, 98). It also has numerous other reported activities, including the ability to modulate signaling molecules such as proinflammatory cytokines, apoptotic proteins, NF-κB, cyclooxygenase-2, 5-LOX, STAT3, C-reactive protein, and prostaglandin E(2), among others. Curcumin has undergone clinical trials in patients with colorectal, pancreatic, breast, prostate, and lung cancer, and multiple myeloma [see review by Gupta et al. (40)]. Most studies involved relatively small numbers of patients and limited tumor shrinkages were seen although some biomarkers changed and some patient symptoms improved. Given the pleiotropic effects of curcumin, it is difficult to ascribe any of its effects in cancer to its redox activity (92).
Conclusions
This review covers 21 redox modifying agents that have been tested in clinical trials for anticancer activity. What are the lessons that can be learned from these agents? Three of these were natural products (α-tocopherol, vitamin C, and curcumin), widely expected to be clinically active based on epidemiological and anecdotal evidence, yet none showed convincing antitumor activity. Three other agents are FDA approved, but only one (ATO) for a mechanism that involves potential redox activity. Many agents are no longer being developed, or their development status is unknown. It is noteworthy that very few of the agents were tested specifically in patients known to express the redox drug target, who might be expected to respond.
The approval number for redox agents is about what would be expected for chemotherapy drugs that enter cancer clinical trials (around 1:20), but much less than for molecularly targeted drugs (1:5). Most of the redox agents were developed against specific targets but were not tested clinically in this way. There are four agents still in clinical trials so that the number approved could be higher, but not significantly so. The reason for the disappointing success rate for redox anticancer drugs probably also relates to the complexity and our incomplete knowledge of the role of redox mechanisms in cancer, and the difficulty of identifying good molecular targets that are critical for tumor growth and progression. There are also the practical difficulties of making redox-active agents that can survive the journey to the tumor through the homeostatically redox-regulated body environment. However, recent advance in chemistry for designing irreversibly acting drugs that often have fine-tuned redox reactive groups may help improve the success of developing these agents in the future.
It is also increasingly clear that redox signaling is a critical process for normal cells and only when redox signaling is deranged becomes important for cancer cells. Drugs often fail because of unanticipated toxic side effects on normal tissues and this appears to be the case for many of the redox drugs tested so far. With increasing knowledge of the role of normal redox signaling, and not just pathological processes where redox defenses are overwhelmed, which has been the major focus until now, it can be hoped that untoward effects on normal tissues will become less of an issue for new generations of redox drugs. This is also the need for more rigorous validation of future redox target so that we can be sure that they are molecularly and clinically relevant to cancer. With this in mind, our improving knowledge of physiological redox mechanisms and the application of advances being made in biomarker development for patient selection for personalized cancer medicine, it can be hoped that the next generation of redox cancer drugs will show more target selectivity, less off target effects, and overall, increased success.
Abbreviations Used
- ATO
arsenic trioxide
- BSO
buthionine sulfoximine
- DCE-MRI
dynamic contrast magnetic resonance imaging
- FDA
Food and Drug Administration
- GGT
gamma-glutamyl-transpeptidase
- GI
gastrointestinal
- GRX
glutaredoxin
- GSH
glutathione
- GSSG
oxidized glutathione
- GST
glutathione S-transferase
- IV
intravenous
- MDS
myelodisplastic syndrome
- MGd
motexafin gadolinium
- NAC
N-acetylcysteine
- NF-kB
nuclear factor kappa B
- NO
nitric oxide
- ROS
reactive oxygen species
- SOD
superoxide dismutase
- TPT
topotecan
- Trx
thioredoxin
- TrxR
thioredoxin reductase
- VEGF
vascular endothelial growth factor
Acknowledgments
Supported by grants CA163541, CA163098, and CA188206 (GP).
Author Disclosure Statement
D.L.K. is an employee and stockholder of PHusis Therapeutics, G.P. is a founder and stockholder of PHusis Therapeutics.
References
- 1.Acharya A, Das I, Chandhok D, and Saha T. Redox regulation in cancer: a double-edge sword with therapeutic potential. Oxid Med Cell Longev 3: 23–34, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Anderson CP, Seeger RC, Matthay KK, Perentesis JP, Neglia JP, Bailey HH, Villablanca JG, Groshen S, Hasenauer B, Maris JM, Seeger RC, and Reynolds CP. The combination of buthionine sulfoximine (BSO) and melphalan (L-PAM) is active against recurrent neuroblastoma. Med Pediatr Oncol 33: 158, 1999 [Google Scholar]
- 3.Anderson CP, Matthay KK, Perentesis JP, Neglia JP, Bailey HH, Villablanca JG, Groshen S, Hasenauer B, Maris JM, Seeger RC, and Reynolds CP. Pilot study of intravenous melphalan combined with continuous infusion L-S,R-buthionine sulfoximine for children with recurrent neuroblastoma. Pediatr Blood Cancer 62: 1739–1746, 2015 [DOI] [PubMed] [Google Scholar]
- 4.Anita S, Modiano M, Lee JJ, Peng YM, Xu MJ, Vallar H, Dalton W, and Lippman S. Breast tissue accumulation of retinamides in a randomized short-term study of fenretinide. Clin Cancer Res 9: 2400–2405, 2003 [PubMed] [Google Scholar]
- 5.Arambula JF, Preihs C, Borthwick D, Magda D, and Sessler JL. Texaphyrins: tumor localizing redox active expanded porphyrins. Anticancer Agents Med Chem 11: 222–232, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bailey HH, Ripple G, Tutsch KD, Arzoomanian RZ, Alberti D, Feierabend C, Mahvi D, Schink J, Pomplun M, Mulcahy RT, and Wilding G. Phase I study of continuous-infusion of L-S,R-buthionine sulfoximine with intravenous melphalan. J Natl Cancer Inst 89: 1789–1796, 1997 [DOI] [PubMed] [Google Scholar]
- 7.Bailey HH, Mulcahy RT, Tutsch KD, Arzoomanian RZ, Alberti D, Tombes MB, Wilding G, Pomplun M, and Spriggs DR. Phase I clinical trial of intravenous L-buthionine sulfoximine and melphalan: an attempt at modulation of glutathione. J Clin Oncol 12: 194–205, 1994 [DOI] [PubMed] [Google Scholar]
- 8.Baker A, Payne CM, Briehl MM, and Powis G. Thioredoxin, a gene found overexpressed in human cancer, inhibits apoptosis in vitro and in vivo. Cancer Res 57: 5162–5167, 1997 [PubMed] [Google Scholar]
- 9.Baker AF, Adab KN, Raghunand N, Chow HHS, Stratton SP, Squire SW, Boice M, Pestano LA, Kirkpatrick DL, and Dragovich T. A phase IB trial of 24-hour intravenous PX-12, a thioredoxin-1 inhibitor, in patients with advanced gastrointestinal cancers. Invest New Drugs 31: 631–641, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Berndt C, Kurz T, Bannenbera S, Jacob R, Holmgren A, and Brunk UT. Ascorbate and endocytosed Motexafin gadolinium induce lysosomal rupture. Cancer Lett 307: 119–123, 2011 [DOI] [PubMed] [Google Scholar]
- 11.Bian Z, Chen S, Cheng C, Wang J, Xiao H, and Qin H. Developing new drugs from annals of Chinese medicine. Acta Pharmaceutica Sinica B 2: 1–7, 2012 [Google Scholar]
- 12.Brar SS, Grigg C, Wilson KS, Holder WD, Jr, Dreau D, Austin C, Foster M, Ghio AJ, Whorton AR, Stowell GW, Whittall LB, Whittle RR, White DP, and Kennedy TP. Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease. Mol Cancer Ther 3: 1049–1060, 2004 [PubMed] [Google Scholar]
- 13.Brigelius-Flohé R. and Flohé L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid Redox Signal 15: 2335–2381, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Calvert P, Yao KS, Hamilton TC, and O'Dwyer PJ. Clinical studies of reversal of drug resistance based on glutathione. Chem Biolo Interact 111–112: 213–224, 1998 [DOI] [PubMed] [Google Scholar]
- 15.Chen X, Shi X, Zhao C, Li X, Lan X, Liu S, Huang H, Liu N, Liao S, Zang D, Song W, Liu Q, Carter BZ, Dou QP, Wang X, and Liu J. Anti-rheumatic agent auranofin induced apoptosis in chronic myeloid leukemia cells resistant to imatinib through both Bcr/Abl-dependent and -independent mechanisms. Oncotarget 5: 9118–9132, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen YR, Zhou G, and Tan TH. c-Jun N-terminal kinase mediates apoptotic signaling induced by N-(4-hydroxyphenyl) retinamide. Mol Pharmacol 56: 1271–1279, 1999 [DOI] [PubMed] [Google Scholar]
- 17.Collins Y, Chouchani ET, Janes AM, Menger KE, Cochemé HM, and Murphy MP. Mitochondrial redox signaling at a glance. J Cell Sci 125: 801–806, 2012 [DOI] [PubMed] [Google Scholar]
- 18.Costa A, De Palo G, Decensi A, Formelli F, Chiesa F, Nava M, Camerini T, Marubini E, and Veronesi U. Retinoids in cancer chemoprevention. Clinical trials with the synthetic analogue fenretinide. Ann N Y Acad Sci 768: 148–162, 1995 [DOI] [PubMed] [Google Scholar]
- 19.Creagan ET, Moertel CG, O'Fallon JR, Schutt AJ, O'Connell MJ, Rubin J, and Frytak S. Failure of high-dose vitamin C (ascorbic acid) therapy to benefit patients with advanced cancer. A controlled trial. N Engl J Med 301: 687–690, 1979 [DOI] [PubMed] [Google Scholar]
- 20.Dekhuijzen PN. Antioxidant properties of N-acetylcysteine: their relevance in relation to chronic obstructive pulmonary disease. Eur Respir J 23: 629–636, 2004 [DOI] [PubMed] [Google Scholar]
- 21.Diaz-Montero CM, Wang Y, Shao L, Feng W, Zidan AA, Pazoles CJ, Montero AJ, and Zhou D. The glutathione disulfide mimetic NOV-002 inhibits cyclophosphamide-induced hematopoietic and immune suppression by reducing oxidative stress. Free Rad Biol Med 52: 1560–1568, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dickinson DA, Moellering DR, Iles KE, Patel RP, Levonen AL, Wigley A, Darley-Usmar VM, and Forman HJ. Cytoprotection against oxidative stress and the regulation of glutathione synthesis. Biol Chem 384: 527–537, 2003 [DOI] [PubMed] [Google Scholar]
- 23.Ding S, Li C, Cheng N, Cui X, Xu X, and Zhou G. Redox regulation in cancer stem cells Oxid Med Cell Longev 2015: 7507982015, 2015 [DOI] [PMC free article] [PubMed]
- 24.Dion PW, Bright-See EB, Smith CC, and Bruce WR. The effect of dietary ascorbic acid and alpha-tocopherol on fecal mutagenicity. Mutat Res 102: 27–37, 1982 [DOI] [PubMed] [Google Scholar]
- 25.Dourado DF, Fernandes PA, Ramos MJ, and Mannervik B. Mechanism of glutathione transferase P1-1-catalyzed activation of the prodrug canfosfamide (TLK286, TELCYTA). Biochemistry, 52: 8069–8078, 2013 [DOI] [PubMed] [Google Scholar]
- 26.Duffy AG, Melillo G, Turkbey B, Allen D, Choyke PL, Chen C, Raffeld M, Doroshow JH, Murgo A, and Kummar S. A pilot trial of oral topotecan (TPT) in patients with refractory advanced solid neoplasms expressing HIF-1alpha. J Clin Oncol 28, 2010. (suppl; abstr e13518) [Google Scholar]
- 27.Ellinghaus P, Heisler I, Unterschemmann K, Haerter M, Beck H, Greschat S, Ehrmann A, Summer H, Flamme I, Oehme F, Thierauch K, Michels M, Hess-Stumpp H, and Ziegelbauer K. BAY 87–2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I. Cancer Med 2: 611–624, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Estensen RD, Levy M, Klopp SJ, Galbraith AR, Mandel JS, Blomquist JA, and Wattenberg LW. N-acetylcysteine suppression of the proliferative index in the colon of patients with previous adenomatous colonic polyps. Cancer Lett 147: 109–114, 1999 [DOI] [PubMed] [Google Scholar]
- 29.Fan C, Zheng W, Fu X, Li X, Wong YS, and Chen T. Enhancement of auranofin-induced lung cancer cell apoptosis by selenocystine, a natural inhibitor of TrxR1 in vitro and in vivo. Cell Death Dis 5:e1191, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Feldman L, Efrati S, Eviatar E, Abramsohn R, Yarovoy I, Gersch E, Averbukh Z, and Weissgarten J. Gentamicin-induced ototoxicity in hemodialysis patients is ameliorated by N-acetylcysteine. Kidney Int 72: 359–363, 2007 [DOI] [PubMed] [Google Scholar]
- 31.Fiskus W, Saba N, Shen M, Ghias M, Liu J, Gupta SD, Chauhan L, Rao R, Gunewardena S, Schorno K, Austin CP, Maddocks K, Byrd J, Melnick A, Huang P, Wiestner A, and Bhalla KN. Auranofin induces lethal oxidative and endoplasmic reticulum stress and exerts potent preclinical activity against chronic lymphocytic leukemia. Cancer Res 74: 2520–2532, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Formelli F, Clerici M, Campa T, Di Mauro MG, Magni A, Mascotti G, Moglia D, De Palo G, Costa A, and Veronesi U. Five-year administration of fenretinide: pharmacokinetics and effects on plasma retinol concentrations. J Clin Oncol 11: 2036–2042, 1993 [DOI] [PubMed] [Google Scholar]
- 33.Gabbiani C, Mastrobuoni G, Sorrentino F, Dani B, Rigobello MP, Bindoli A, Cinellu MA, Pieraccini G, Messoria L, and Casini A. Thioredoxin reductase, an emerging target for anticancer metallodrugs. Enzyme inhibition by cytotoxic gold(III) compounds studied with combined mass spectrometry and biochemical assays. Med Chem Commun 2: 50–54, 2011 [Google Scholar]
- 34.Garrett CR, Bekaii-Saab TS, Ryan T, Fisher GA, Clive S, Kavan P, Shacham-Shmueli E, Buchbinder A, and Goldberg RM. Randomized phase 2 study of pegylated SN-38 (EZN-2208) or irinotecan plus cetuximab in patients with advanced colorectal cancer. Cancer 119: 4223–4230, 2013 [DOI] [PubMed] [Google Scholar]
- 35.Geeraert L. CAM-Cancer Consortium. Vitamin E during cancer treatment. Available www.cam-cancer.org/CAM-Summaries/Dietary-approaches/Vitamin-E-during-cancer-treatment September 30, 2015
- 36.Gibaud S. and Jaouen G. Arsenic-based drugs: from Fowler's solution to modern anticancer chemotherapy. Topics Organomet Chem 32: 1–20, 2010 [Google Scholar]
- 37.Gilmore TD. and Herscovitch M. Inhibitors of NF-κB signaling: 785 and counting. Oncogene 25: 6887–6899, 2006 [DOI] [PubMed] [Google Scholar]
- 38.Griffith OW. and Meister AJ. Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine (S-n-butyl homocysteine sulfoximine). J Biol Chem 254: 7558–7560, 1979 [PubMed] [Google Scholar]
- 39.Guilbault C, De Sanctis JB, Wojewodka G, Saeed Z, Lachance C, Skinner TA, Vilela RM, Kubow S, Lands LC, Hajduch M, Matouk E, and Radzioch D. Fenretinide corrects newly found ceramide deficiency in cystic fibrosis. Am J Resp Cell Mol Biol 38: 47–56, 2008 [DOI] [PubMed] [Google Scholar]
- 40.Gupta SC, Patchva S, and Aggarwal BB. Therapeutic roles of curcumin: lessons learned from clinical trials. AAPS J 15: 195–218, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gurbuz AK, Ozel AM, Ozturk R, Yildirim S, Yazgan Y, and Demirturk L. Effect of N-acetyl cysteine on Helicobacter pylori. South Med J 98: 1095–1097, 2005 [DOI] [PubMed] [Google Scholar]
- 42.Hamilton TC, Winker MA, Louie KG, Batist G, Behrens BC, Tsuruo T, Grotzinger KR,. McKoy WM, Young RC, and Ozols RF. Augmentation of adriamycin, melphalan, and cisplatin cytotoxicity in drug-resistant and -sensitive human ovarian carcinoma cell lines by buthionine sulfoximine mediated glutathione depletion. Biochem Pharmacol 34: 2583–2586, 1985 [DOI] [PubMed] [Google Scholar]
- 43.Heinonen OP. The alpha-tocopherol, beta-carotene lung cancer prevention study: design, methods, participants characteristics, and compliance. AEP 4: 1–10, 1994 [DOI] [PubMed] [Google Scholar]
- 44.Heinonen OP, Albanes D, Virtamo J, et al. Prostate cancer and supplementation with alpha-tocopherol and beta-carotene: incidence and mortality in a controlled trial. J Nat Cancer Inst 90: 440–446, 1998 [DOI] [PubMed] [Google Scholar]
- 45.Helbig L, Koi L, Brüchner K, Gurtner K, Hess-Stumpp H, Unterschemmann K, Baumann M, Zips D, and Yaromina A. BAY 87–2243, a novel inhibitor of hypoxia-induced gene activation, improves local tumor control after fractionated irradiation in a schedule-dependent manner in head and neck human xenografts. Rad Oncol 9: 207, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Helzlsouer KJ, Huang HY, Alberg AJ, Hoffman S, Burke A, Norkus EP, Morris JS, and Comstock GW. Association between alpha-tocopherol, gamma-tocopherol, selenium, and subsequent prostate cancer. J Nat Cancer Inst 92: 2018–2023, 2000 [DOI] [PubMed] [Google Scholar]
- 47.Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K, Mitsiades C, Munshi NC, Richardson PG, Carrasco RD, and Anderson KC. Bortezomib induces canonical nuclear factor-kappa B activation in multiple myeloma cells. Blood 114: 1046–1052, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jacobs C, Hutton B, Ng T, Shorr R, and Clemons M. Is there a role for oral or intravenous ascorbate (vitamin C) in treating patients with cancer? A systematic review. Oncologist 20: 210–223, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jeong W, Rapisarda A, Park SR, Kinders RJ, Chen A, Melillo G, Turkbey B, Steinberg SM, Choyke P, Doroshow JH, and Kummar S. Pilot trial of EZN-2968, an antisense oligonucleotide inhibitor of hypoxia-inducible factor-1 alpha (HIF-1α), in patients with refractory solid tumors. Cancer Chemother Pharmacol 73: 343–348, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jeong W, Park SR, Rapisarda A, Fer N, Kinders RJ, Chen A, Melillo G, Turkbey B, Steinberg SM, Choyke P, Doroshow JH, and Kummar S. Weekly EZN-2208 (PEGylated SN-38) in combination with bevacizumab in patients with refractory solid tumors. Invest New Drugs 32: 340–346, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jones DT, Pugh CW, Wigfield S, Stevens MFG, and Harris AL. Novel thioredoxin inhibitors paradoxically increase hypoxia-inducible factor-α expression by decrease functional transcriptional activity, and binding and degradation. Clin Cancer Res 12: 5384–5394, 2006 [DOI] [PubMed] [Google Scholar]
- 52.Kang J. and Peervaiz S. Mitochondria: redox metabolism and dysfunction. Biochem Res Int 2012: Article 896751, 14, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kim HY. and Kim JR. Thioredoxin as a reducing agent for mammalian methionine sulfoxide reductases B lacking resolving cysteine. Biochem Biophys Res Commun 371: 490–494, 2008 [DOI] [PubMed] [Google Scholar]
- 54.Kirkpatrick DL, Kuperus M, Dowdeswell M, Potier N, Donald LJ, Kunkel M, Berggren M, Angulo M, and Powis G. Mechanisms of inhibition of the thioredoxin growth factor system by antitumor 2-imidazolyl disulfides. Biochem Pharmacol 55: 987–994, 1998 [DOI] [PubMed] [Google Scholar]
- 55.Klein EA, Thompson IM, Tangen CM, Crowley JJ, Lucia S, Goodman PJ, Minasian LM, Ford LG, Parnes HL, Gaziano JM, Karp DD, Lieber MM, Walther PJ, Klotz L, Parsons JK, Chin JL, Darke AK, Li0ppman SM, Goodman GE, Meyskens FL, and Baker LH. Vitamin E and the risk of prostate cancer: results of the selenium and vitamin E cancer prevention trial (SELECT). JAMA 306: 1549–1556, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kummar S, Raffeld M, Juwara L, Horneffer Y, Strassberger A, Allen D, Steinberg SM, Rapisarda A, Spencer SD, Figg WD, Chen X, Turkbey IB, Choyke P,. Murgo AJ, Doroshow JH, and Melillo G. Multihistology, target-driven pilot trial of oral topotecan as an inhibitor of hypoxia-inducible factor-1α (HIF-1α) in advanced solid tumors. Clin Cancer Res 17: 5123–5131, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liu N, Li X, Huang H, Zhao C, Liao S, Yang C, et al. Clinically used antirheumatic agent auranofin is a proteasomal deubiquitinase inhibitor and inhibits tumor growth. Oncotarget 5: 5453–5471, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lu J. and Holmgren A. The thioredoxin antioxidant system. Free Radic Biol Med 66: 75–87, 2014 [DOI] [PubMed] [Google Scholar]
- 59.Lu J, Chew EH, and Holmgren A. Targeting thioredoxin reductase is a basis for cancer therapy by arsenic trioxide. Proc Natl Acad Sci U S A 104: 12288–12293, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Magda D, Lepp C, Gerasimchuk N, Lee I, Sessler JL, Lin A, Biaglow JE, and Miller RA. Redox cycling by motexafin gadolinium enhances cellular response to ionizing radiation by forming reactive oxygen species. Int J Radiat Oncol Biol Phys 51: 1025–1036, 2001 [DOI] [PubMed] [Google Scholar]
- 61.Mailloux RJ, Jin X, and Willmore WG. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol 2: 123–139, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Marzano C, Gandin V, Folda A, Scutari G, Bindoli A, and Rigobello MP. Inhibition of thioredoxin reductase by auranofin induces apoptosis in cisplatin-resistant human ovarian cancer cells. Free Radic Biol Med 42: 872–881, 2007 [DOI] [PubMed] [Google Scholar]
- 63.Michelet L, Zaffagnini M, Massor V, Keryer E, Manacker H, Miginiac-Maslow M, Issakidis- Bourgue E, and Lemaire SD. Thioredoxins, glutaredoxins and glutathionylation: new crosstalks to explore. Photosynth Res 89: 225–245, 2006 [DOI] [PubMed] [Google Scholar]
- 64.Millea PJ. N-Acetylcysteine: multiple clinical applications. Am Fam Physician 80: 265–269, 2009 [PubMed] [Google Scholar]
- 65.Miller RA, Woodburn KW, Fan Q, Lee I, Miles D, Duran G, Sikic B, and Magda D. Motexafin gadolinium: a redox active drug that enhances the efficacy of bleomycin and doxorubicin. Clin Cancer Res 7: 3215–3221, 2001 [PubMed] [Google Scholar]
- 66.Mullaicharam AR. and Maheswaran A. Pharmacological effects of curcumin. Int J Nutr Pharmacol Neurol Dis 2: 92–99, 2012 [Google Scholar]
- 67.Nikitovic D. and Holmgren A. S-nitrosoglutathione is cleaved by the thioredoxin system with liberation of glutathione and redox regulating nitric oxide. J Biol Chem 271: 19280–19185, 1996 [DOI] [PubMed] [Google Scholar]
- 68.Nonn L, Williams RR, Erickson RP, and Powis G. The absence of mitochondrial thioredoxin 2 causes massive apoptosis, exencephaly, and early embryonic lethality in homozygous mice. Mol Cell Biol 23: 916–922, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Onnis B, Rapisarda A, and Melillo G. Development of HIF-1 inhibitors for cancer therapy. J Cell Mol Med 13: 2780–2786, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Osborne CRC, O'Shaughnessy JA, Steinberg MS, Holmes FA, Kim HS, Kocs DM, Richards PD, Vukelja SJ, Berkowitz N, and Buchbinder A. Final analysis of Phase 2 study of EZN-2208 (PEG-SN38) in metastatic breast cancer (MBC) demonstrates activity in patients with triple negative breast cancer (TNBC) and in platinum pretreated patients. Am Soc Clin Oncol Poster #1017, 2012 [Google Scholar]
- 71.Padayatty SJ, Riordan HD, Hewitt SM, Katz A, Hoffer LJ, and Levine M. Intravenously administered vitamin C as cancer therapy: three cases. CMAJ 174: 937–942, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 18: 6853–6866, 1999 [DOI] [PubMed] [Google Scholar]
- 73.Pastorino F, Loi M, Sapra P, Becherini P, Cilli M, Emionite L, Ribatti D, Greenberger LM, Horak ID, and Ponzoni M. Tumor regression and curability of preclinical neuroblastoma models by PEGylated SN38 (EZN-2208), a novel topoisomerase I inhibitor. Clin Cancer Res 16: 4809–4821, 2010 [DOI] [PubMed] [Google Scholar]
- 74.Patnaik A, Papadopoulos KP, Tolcher AW, Beeram M, Urien S, Schaaf LJ, Sanaa Tahiri S, Tanios Bekaii-Saab T, Lokiec FM, Rezaï K, and Buchbinder A. Phase I dose-escalation study of EZN-2208 (PEG-SN38), a novel conjugate of poly(ethylene) glycol and SN38, administered weekly in patients with advanced cancer. Cancer Chemother Pharmacol 71: 1499–1506, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Pessetto ZY, Weir SJ, Sethi G, Broward MA, and Godwin AK. Drug repurposing for gastrointestinal stromal tumor. Mol Cancer Ther 12: 1299–1309, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Polimeni M. and Gazzano E. Is redox signaling a feasible target for overcoming multidrug resistance in cancer chemotherapy? Front Pharmacol 5: 286, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Raffel J, Bhattacharyya AK, Gallegos A, Cui H, Einspahr JG, Alberts DS, and Powis G. Increased expression of thioredoxin-1 in human colorectal cancer is associated with decreased patient survival. J Lab Clin Med 142: 46–51, 2003 [DOI] [PubMed] [Google Scholar]
- 78.Ramanathan RK, Abbruzzese J, Dragovich T, Kirkpatrick L, Guillen JM, Baker AF, Pestano LA, Green S, and Von Hoff DD. A randomized phase II study of PX-12, an inhibitor of thioredoxin in patients with advanced cancer of the pancreas following progression after a gemcitabine-containing combination. Cancer Chemother Pharmacol 67: 503–509, 2011 [DOI] [PubMed] [Google Scholar]
- 79.Ramanathan RK, Kirkpatrick DL, Belani CB, Friedland D, Green SB, Chow H-HS, Cordova CA, Stratton SP, Sharlow ER, Baker A, and Dragovich T. A Phase I pharmacokinetic and pharmacodynamic study of PX-12, a novel inhibitor of thioredoxin-1, in patients with advanced solid tumors. Clin Cancer Res 13: 2109–2114, 2007 [DOI] [PubMed] [Google Scholar]
- 80.Ramanathan RK, Stephenson JJ, Weiss GJ, Pestano LA, Lowe A, Hiscox A, Leos RA, Martin JC, Kirkpatrick L, and Richards DA. A phase I trial of PX-12, a small-molecule inhibitor of thioredoxin-1, administered as a 72-hour infusion every 21 days in patients with advanced cancers refractory to standard therapy. Invest New Drugs 30: 1591–1596, 2012 [DOI] [PubMed] [Google Scholar]
- 81.Raninga PV, Trapani GD, Vuckovic S, Bhatia M, and Tonissen KR. Inhibition of thioredoxin 1 leads to apoptosis in drug-resistant multiple myeloma. Oncotarget 6: 15410–15424, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Rao Y, Li R, and Zhang D. A drug from poison: how the therapeutic effect of arsenic trioxide on acute promyelocytic leukemia was discovered. Sci China Life Sci 56: 495–502, 2013 [DOI] [PubMed] [Google Scholar]
- 83.Rapisarda A, Hollingshead M, Uranchimeg B, Bonomi CA, Borgel SD, Carter JP, Gehrs B, Raffeld M, Kinders RJ, Parchment R, Anver MR, Shoemaker RH, and Melillo G. Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition. Mol Cancer Ther 8: 1867–1877, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Rapisarda A, Uranchimeg B, Scudiero DA, Selby M, Sausville EA, Shoemaker RH, and Melillo G. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway. Cancer Res 62: 4316–4324, 2002 [PubMed] [Google Scholar]
- 85.Rapisarda A, Uranchimeg B, Sordet O, Sordet O, Pommier Y, Shoemaker RH, and Melillo G.Topoisomerase I-mediated inhibition of hypoxia-inducible factor 1: mechanism and therapeutic implications. Cancer Res 64: 1475–1482, 2004 [DOI] [PubMed] [Google Scholar]
- 86.Rapisarda A, Zalek J, Hollingshead M, Braunschweig T, Uranchimeg B, Bonomi CA, Borgel SD, Carter JP, Hewitt SM, Shoemaker RH, and Melillo G. Schedule-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation, angiogenesis, and tumor growth by topotecan in U251-HRE glioblastoma xenografts. Cancer Res 64: 6845–6848, 2004 [DOI] [PubMed] [Google Scholar]
- 87.Raza A, Galili N, Callander N, Ochoa L, Piro L, Emanuel P, Williams S, Burris III H, Faderl S, Estrov Z, Curtin P, Larson RA, Keck JG, Jones M, Meng L, and Brown GL. Phase 1-2a multicenter dose-escalation study of ezatiostat hydrochloride liposomes for injection (Telintra®, TLK199), a novel glutathione analog prodrug in patients with myelodysplastic syndrome. J Hematol Oncol 2: 20–32, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Raza A, Galili N, Smith S, Godwin J, Lancet J, Melchert M, Jones M, Keck JG, Meng L, Brown GL, and List A. Phase 1 multicenter dose-escalation study of ezatiostat hydrochloride (TLK199 tablets), a novel glutathione analog prodrug, in patients with myelodysplastic syndrome. Blood 113: 6523–6540, 2009 [DOI] [PubMed] [Google Scholar]
- 89.This reference has been deleted.
- 90.Rode C. and Thomson MJ. Auranofin: repurposing an old drug for a golden new age. Drugs R D 15: 13–20, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ruscoe JE, Rosario LA, Wang T, Gaté L, Arifoglu P, Wolf CR, Henderson CJ, Ronai Z, and Tew KD. Pharmacologic or genetic manipulation of glutathione S-transferase P1-1 (GSTpi) influences cell proliferation pathways. J Pharmacol Exp Ther 298: 339–345, 2001 [PubMed] [Google Scholar]
- 92.Sa G. and Das T. Anti cancer effects of curcumin: cycle of life and death. Cell Div 3: 14, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sapra P, Kraft P, Pastorino F, Ribatti D, Dumble M, Mehlig M, Wang M, Ponzoni M, Greenberger LM, and Horak ID. Potent and sustained inhibition of HIF-1α and downstream genes by a polyethyleneglycol-SN38 conjugate EZN-2208, results in antiangiogenic effects. Angiogenesis 14: 245–253, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sessler JL. and Miller RA. Texaphyrins: new drugs with diverse clinical applications in radiation and photodynamic therapy. Biochem Pharmacol 59: 733–739, 2000 [DOI] [PubMed] [Google Scholar]
- 95.Siemann DW. and Beyers KL. In vivo therapeutic potential of combination thiol depletion and alkylating chemotherapy. Br J Cancer 68: 1071–1079, 1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Smith AC, Liao JT, Page JG, Wientjes MG, and Grieshaber CK. Pharmacokinetics of buthionine sulfoximine (NSC 326231) and its effect on melphalan-induced toxicity in mice. Cancer Res 49: 5385–5391, 1989 [PubMed] [Google Scholar]
- 97.Sreejayan N. and Rao MN. Nitric oxide scavenging by curcuminoids. J Pharm Pharmacol 49: 105–107, 1997 [DOI] [PubMed] [Google Scholar]
- 98.Sreejayan N. and d Rao MN. Free radical scavenging activity of curcuminoids. Arzneimittelforschung 46: 169–171,1996 [PubMed] [Google Scholar]
- 99.Stone WL. and Papas AM. Tocopherols and the etiology of colon cancer. J Natl Cancer Inst 89: 1006–1014, 1997 [DOI] [PubMed] [Google Scholar]
- 100.Subbarayan PR. and Ardalan B. In the war against solid tumors arsenic trioxide needs partners. J Gastrointest Cancer 45: 363–371, 2014 [DOI] [PubMed] [Google Scholar]
- 101.Suzuki S, Higuchi M, Proske RJ, Oridate N, Hong WK, and Lotan R. Implication of mitochondria-derived reactive oxygen species, cytochrome C and caspase-3 in N-(4-hydroxyphenyl) retinamide-induced apoptosis in cervical carcinoma cells. Oncogene 18: 6380–6387, 1999 [DOI] [PubMed] [Google Scholar]
- 101a.Swerdlow RD, Zwiebel JA, Gravell AE, and Cheson BD. Current clinical trials of fenretinide. Oncology 15: 1595–1596, 2001 [PubMed] [Google Scholar]
- 102.Tew KD. TLK-286: a novel glutathione S-transferase-activated prodrug. Expert Opin Investig Drugs 14: 1047–1054, 2005 [DOI] [PubMed] [Google Scholar]
- 103.The alpha-tocopherol beta carotene cancer prevention study group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med 330: 1029–1035, 1994 [DOI] [PubMed] [Google Scholar]
- 104.Tibes R, Falchook GS, Von Hoff DD, Weiss GJ, Iyengar T, Kurzrock R, Pestano L, Lowe AM, and Herbst RS. Results from a phase I, dose-escalation study of PX-478, an orally available inhibitor of HIF-1α. J Clin Oncol 28: suppl 3076, 2010 [Google Scholar]
- 105.Townsend DM, He L, Hutchens S, Garrett TE, Pazoles CJ, and Tew KD. NOV-002, a glutathione disulfide mimetic, as a modulator of cellular redox balance. Cancer Res 68: 2870–2877, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Traber MG. Vitamin E. In: Present Knowledge in Nutrition, 10th ed, edited by Erdman JWJ, Macdonald IA, Zeisel SH. Washington D.C: Wiley-Blackwell; 2012, pp. 214–229
- 107.Traverso N, Ricciarelli R, Nitti M, Marengo B, furfaro AL, Ronzato MA, Marinari UM, and Domenicotti C. Role of Glutathione in cancer progression and chemoresistance. Oxid Med Cell Longev 2013: Article ID 972913, 10, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.van Zandwijk N, Dalesio O, Pastorino U, de Vries N, and van Tinteren H. EUROSCON, a randomized trial of vitamin A and N-acetylcysteine in patients with head and neck cancer or lung cancer. J Natl Cancer Inst 92: 977–986, 2000 [DOI] [PubMed] [Google Scholar]
- 109.Veronesi U, Mariani L, Decensi A, Formelli F, Camerini T, Miceli R, Di Mauro MG, Costa A, Marubini E, Sporn MB, and De Palo G. Fifteen-year results of a randomized phase III trial of fenretinide to prevent second breast cancer. Ann Oncol 17: 1065–1071, 2006 [DOI] [PubMed] [Google Scholar]
- 110.Welsh SJ, Bellamy WT, Briehl MM, and Powis G. The redox protein thioredoxin-1 (Trx-1) increases hypoxia-inducible factor 1alpha protein expression: Trx-1 overexpression results in increased vascular endothelial growth factor production and enhanced tumor angiogenesis. Cancer Res 62: 5089–5095, 2002 [PubMed] [Google Scholar]
- 111.Welsh SJ, Williams RR, Birmingham A, Newman DJ, Kirkpatriack DL, and Powis G. The thioredoxin redox inhibitors 1-methylpropyl 2-imidazolyl disulfide and pleurotin inhibit hypoxia-induced factor 1α and vascular endothelial growth factor formation. Mol Cancer Ther 2: 235–243, 2003 [PubMed] [Google Scholar]
- 112.Welsh S, Williams R, Kirkpatrick L, Paine-Murrieta G, and Powis G. Antitumor activity and pharmacodynamics properties of PX-478, an inhibitor of hypoxia-inducible factor-1α. Mol Cancer Ther 3: 233–244, 2004 [PubMed] [Google Scholar]
- 113.Wu J, DiPietrantonio A, and Hsieh T. Mechanism of fenretinide (4-HPR)-induced cell death. Apoptosis 6: 377–388, 2001 [DOI] [PubMed] [Google Scholar]
- 114.Zhang T-D, Chen G-Q, Wang Z-G, Wang Z-Y, Chen S-J, and Chen Z. Arsenic trioxide, a therapeutic agent for APL. Oncogene 20: 7146–7153, 2001 [DOI] [PubMed] [Google Scholar]
- 115.Zheng CY, Lam SK, Li YY, and Ho JC. Arsenic trioxide-induced cytotoxicity in small cell lung cancer via altered redox homeostasis and mitochondrial integrity. Int J Oncol 46: 1067–1078, 2015 [DOI] [PubMed] [Google Scholar]
- 116.Zhou J, Damdimopoulos AE, Spyrou G, and Brüne B. Thioredoxin 1 and thioredoxin 2 have opposed regulatory functions on hypoxia-inducible factor-1alpha. J Biol Chem 282: 7482–7490, 2007 [DOI] [PubMed] [Google Scholar]