Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Biopharm Drug Dispos ; 45(2): 83-92, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38492211

ABSTRACT

AST-001 is a chemically synthesized inactive nitrogen mustard prodrug that is selectively cleaved to a cytotoxic aziridine (AST-2660) via aldo-keto reductase family 1 member C3 (AKR1C3). The purpose of this study was to investigate the pharmacokinetics and tissue distribution of the prodrug, AST-001, and its active metabolite, AST-2660, in mice, rats, and monkeys. After single and once daily intravenous bolus doses of 1.5, 4.5, and 13.5 mg/kg AST-001 to Sprague-Dawley rats and once daily 1 h intravenous infusions of 0.5, 1.5, and 4.5 mg/kg AST-001 to cynomolgus monkeys, AST-001 exhibited dose-dependent pharmacokinetics and reached peak plasma levels at the end of the infusion. No significant accumulation and gender differences were observed after 7 days of repeated dosing. In rats, the half-life of AST-001 was dose independent and ranged from 4.89 to 5.75 h. In cynomolgus monkeys, the half-life of AST-001 was from 1.66 to 5.56 h and increased with dose. In tissue distribution studies conducted in Sprague-Dawley rats and in liver cancer PDX models in female athymic nude mice implanted with LI6643 or LI6280 HepG2-GFP tumor fragments, AST-001 was extensively distributed to selected tissues. Following a single intravenous dose, AST-001 was not excreted primarily as the prodrug, AST-001 or the metabolite AST-2660 in the urine, feces, and bile. A comprehensive analysis of the preclinical data and inter-species allometric scaling were used to estimate the pharmacokinetic parameters of AST-001 in humans and led to the recommendation of a starting dose of 5 mg/m2 in the first-in-human dose escalation study.


Subject(s)
Nitrogen Mustard Compounds , Prodrugs , Animals , Female , Mice , Rats , Aldo-Keto Reductase Family 1 Member C3/drug effects , Macaca fascicularis , Mice, Nude , Rats, Sprague-Dawley , Nitrogen Mustard Compounds/pharmacokinetics , Aziridines/pharmacokinetics , Dose-Response Relationship, Drug
2.
Cancer Chemother Pharmacol ; 83(6): 1183-1189, 2019 06.
Article in English | MEDLINE | ID: mdl-30868237

ABSTRACT

PURPOSE: Despite positive responses in phase II clinical trials, the bioreductive prodrug apaziquone failed to achieve statistically significant activity in non-muscle invasive bladder cancer in phase III trials. Apaziquone was administered shortly after transurethral resection and here we test the hypothesis that haematuria inactivates apaziquone. METHODS: HPLC analysis was used to determine the ability of human whole blood to metabolise apaziquone ex vivo. An in vitro model of haematuria was developed and the response of RT112 and EJ138 cells following a 1-h exposure to apaziquone was determined in the presence of urine plus or minus whole blood or lysed whole blood. RESULTS: HPLC analysis demonstrated that apaziquone is metabolised by human whole blood with a half-life of 78.6 ± 23.0 min. As a model for haematuria, incubation of cells in media containing up to 75% buffered (pH 7.4) urine and 25% whole blood was not toxic to cells for a 1-h exposure period. Whole blood (5% v/v) significantly (p < 0.01) reduced the potency of apaziquone in this experimental model. Lysed whole blood also significantly (p < 0.05) reduced cell growth, although higher concentrations were required to achieve an effect (15% v/v). CONCLUSIONS: The results of this study demonstrate that haematuria can reduce the potency of apaziquone in this experimental model. These findings impact upon the design of further phase III clinical trials and strongly suggest that apaziquone should not be administered immediately after transurethral resection of non-muscle invasive bladder cancer when haematuria is common.


Subject(s)
Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Chromatography, High Pressure Liquid , Hematuria/complications , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aziridines/pharmacokinetics , Aziridines/pharmacology , Cell Line, Tumor , Half-Life , Humans , In Vitro Techniques , Indolequinones/pharmacokinetics , Indolequinones/pharmacology , Research Design
3.
Expert Opin Drug Metab Toxicol ; 13(7): 783-791, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28637373

ABSTRACT

INTRODUCTION: Apaziquone (also known as EO9 and QapzolaTM) is a prodrug that is activated to DNA damaging species by oxidoreductases (particularly NQO1) and has the ability to kill aerobic and/or hypoxic cancer cells. Areas covered: Whilst its poor pharmacokinetic properties contributed to its failure in phase II clinical trials when administered intravenously, these properties were ideal for loco-regional therapies. Apaziquone demonstrated good anti-cancer activity against non-muscle invasive bladder cancer (NMIBC) when administered intravesically to marker lesions and was well tolerated with no systemic side effects. However, phase III clinical trials did not reach statistical significance for the primary endpoint of 2-year recurrence in apaziquone over placebo although improvements were observed. Post-hoc analysis of the combined study data did indicate a significant benefit for patients treated with apaziquone, especially when the instillation of apaziquone was given 30 min or more after surgery. A further phase III study is ongoing to test the hypotheses generated in the unsuccessful phase III studies conducted to date. Expert opinion: Because of its specific pharmacological properties, Apaziquone is excellently suited for local therapy such as NMIBC. Future studies should include proper biomarkers.


Subject(s)
Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aziridines/pharmacokinetics , Aziridines/pharmacology , Humans , Indolequinones/pharmacokinetics , Indolequinones/pharmacology , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms/pathology
4.
Sci Rep ; 5: 14554, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26416623

ABSTRACT

In this study, the potential effects of bacteria on the efficacy of frequently used chemotherapies was examined. Bacteria and cancer cell lines were examined in vitro and in vivo for changes in the efficacy of cancer cell killing mediated by chemotherapeutic agents. Of 30 drugs examined in vitro, the efficacy of 10 was found to be significantly inhibited by certain bacteria, while the same bacteria improved the efficacy of six others. HPLC and mass spectrometry analyses of sample drugs (gemcitabine, fludarabine, cladribine, CB1954) demonstrated modification of drug chemical structure. The chemoresistance or increased cytotoxicity observed in vitro with sample drugs (gemcitabine and CB1954) was replicated in in vivo murine subcutaneous tumour models. These findings suggest that bacterial presence in the body due to systemic or local infection may influence tumour responses or off-target toxicity during chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Cladribine/pharmacokinetics , Colonic Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Vidarabine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Biotransformation , Cladribine/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Escherichia coli/growth & development , Female , Injections, Intralesional , Injections, Subcutaneous , Listeria/growth & development , Mice , Neoplasm Transplantation , Skin/drug effects , Skin/microbiology , Skin/pathology , Treatment Outcome , Vidarabine/pharmacokinetics , Vidarabine/pharmacology , Gemcitabine
6.
Cancer Chemother Pharmacol ; 69(4): 1039-49, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22186884

ABSTRACT

PURPOSE: Preclinical studies evaluated the anti-tumor activity and mechanism of action of AMP423, a naphthyl derivative of 2-cyanoaziridine-1-carboxamide with structural similarity to the pro-oxidant anti-tumor agent imexon. METHODS: The cytotoxic potency was evaluated in vitro against a variety of human cancer cell lines. Mechanism-of-action studies were performed in the human 8226/S myeloma cell line and its imexon-resistant variant, 8226/IM10. In vivo activity was evaluated against human myeloma and lymphoma xenografts in SCID mice. Pharmacokinetics and toxicology were investigated in non-tumor-bearing mice. RESULTS: The 72-h IC(50)s for all cell types ranged from 2 to 36 µM, across a wide variety of human cancer cell lines. AMP423 was active in SCID mice bearing 8226/S myeloma and SU-DHL-6 B-cell lymphoma tumors, with a median tumor growth delay (T-C) of 21 days (P = 0.0002) and 5 days (P = 0.004), respectively, and a median tumor growth inhibition (T/C) of 33.3% (P = 0.03) and 82% (P = 0.01), respectively. In non-tumor-bearing mice, AMP423 was not myelosuppressive. Mechanistic studies show that AMP423's mode of cell death is a mixture of necrosis and apoptosis, with generation of reactive oxygen species, inhibition of protein synthesis, and a decrease in reduced sulfhydryl levels, but no alkylation of nucleophiles. Unlike its structural analog imexon, which causes cell cycle arrest in G(2)/M, AMP423 induces the accumulation of cells in S-phase. CONCLUSIONS: AMP423 has pro-oxidant effects similar to imexon, has greater cytotoxic potency in vitro, and has anti-tumor activity in hematologic tumors in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Lymphoma, B-Cell/drug therapy , Multiple Myeloma/drug therapy , Naphthalenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Aziridines/chemistry , Aziridines/pharmacokinetics , Cell Cycle Checkpoints , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Hexanones/chemistry , Hexanones/pharmacokinetics , Hexanones/pharmacology , Humans , Lymphoma, B-Cell/metabolism , Male , Mice , Mice, SCID , Multiple Myeloma/metabolism , Naphthalenes/chemistry , Naphthalenes/pharmacokinetics , Xenograft Model Antitumor Assays
7.
Free Radic Res ; 45(10): 1184-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21762045

ABSTRACT

NRH:quinone oxidoreductase 2 (QR2) is a cytosolic enzyme that catalyzes the reduction of quinones, such as menadione and co-enzymes Q. With the aim of understanding better the mechanisms of action of QR2, we approached this enzyme catalysis via electron paramagnetic resonance (EPR) measurements of the by-products of the QR2 redox cycle. The variation in the production of oxidative species such as H(2)O(2), and subsequent hydroxyl radical generation, was measured during the course of QR2 activity under aerobic conditions and using pure human enzyme. The effects on the activity of the following were compared: (i) synthetic (N-benzyldihydronicotinamide, BNAH) or natural (nicotinamide riboside, NRH) co-substrates; (ii) synthetic (menadione) or natural (co-enzyme Q0, Q2) substrates; (iii) QR2 modulators and inhibitors (melatonin, resveratrol and S29434); (iv) a pro-drug activated via a redox cycle [CB1954, 5-(aziridin-1-yl)-2,4-dinitrobenzamide]. The results were also compared with those obtained with human QR1. The production of hydroxyl radicals is: (i) observed whatever the substrate/co-substrate used; ii) quenched by adding catalase; (iii) not observed with the specific QR2 inhibitor S29434; (iv) observed with the pro-drug CB1954. While QR2 produced free radicals with this pro-drug, QR1 gave no EPR signal showing the strong reducing capacity of QR2. In conclusion, EPR analysis of QR2 enzyme activity through free radical production enables modulators and effective inhibitors to be distinguished.


Subject(s)
Quinone Reductases/chemistry , Quinone Reductases/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Binding Sites , Electron Spin Resonance Spectroscopy , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Free Radicals/chemistry , Free Radicals/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Oxidation-Reduction , Prodrugs/pharmacokinetics , Quinone Reductases/antagonists & inhibitors , Substrate Specificity
8.
Ann Oncol ; 22(7): 1653-1660, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21378203

ABSTRACT

BACKGROUND: This trial describes a first-in-man evaluation of RH1, a novel bioreductive drug activated by DT-diaphorase (DTD), an enzyme overexpressed in many tumours. PATIENTS AND METHODS: A dose-escalation phase I trial of RH1 was carried out. The primary objective was to establish the maximum tolerated dose (MTD) of RH1. Secondary objectives were assessment of toxicity, pharmacokinetic determination of RH1 and pharmacodynamic assessment of drug effect through measurement of DNA cross linking in peripheral blood mononuclear cells (PBMCs) and tumour, DTD activity in tumour and NAD(P)H:quinone oxidoreductase 1 (NQO1) polymorphism status. RESULTS: Eighteen patients of World Health Organization performance status of zero to one with advanced refractory solid malignancies were enrolled. MTD was 1430 µg/m(2)/day with reversible bone marrow suppression being dose limiting. Plasma pharmacokinetic analysis showed RH1 is rapidly cleared from blood (t(1/2) = 12.3 min), with AUC increasing proportionately with dose. The comet-X assay demonstrated dose-related increases in DNA cross linking in PBMCs. DNA cross linking was demonstrated in tumours, even with low levels of DTD. Only one patient was homozygous for NQO1 polymorphism precluding any conclusion of its effect. CONCLUSIONS: RH1 was well tolerated with predictable and manageable toxicity. The MTD of 1430 µg/m(2)/day is the dose recommended for phase II trials. The biomarkers of DNA cross linking, DTD activity and NQO1 status have been validated and clinically developed.


Subject(s)
Aziridines/therapeutic use , Benzoquinones/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/drug therapy , Adult , Aged , Aziridines/pharmacokinetics , Benzoquinones/pharmacokinetics , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/genetics , Neoplasms/enzymology , Neoplasms/pathology , Polymorphism, Genetic/genetics , Retrospective Studies , Tissue Distribution , Treatment Outcome
9.
Sci Transl Med ; 2(40): 40ra50, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20630857

ABSTRACT

DNA-damaging agents are widely used in cancer treatment despite their lack of tumor specificity. Human NQO2 (quinone oxidoreductase-2) is an atypical oxidoreductase because no endogenous electron donor has been identified to date. The enzyme converts CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide], in the presence of the synthetic nicotinamide cofactor analog EP0152R, to a cytotoxic bifunctional alkylating agent. NQO2 activity in hepatocellular tumor tissue is higher than that in other cancer types by a factor of 6 and higher than that in bone marrow by a factor of 20. Structural data from x-ray crystallography and nuclear magnetic resonance spectroscopy allowed us to construct a model of CB1954 and EP0152R binding to NQO2, which suggested an optimal infusion schedule for a phase I trial combining the two agents. Thirty-two patients were treated, and diarrhea and serum transaminase concentrations defined a maximum tolerated dose for the drug combination. There was a clear pharmacokinetic interaction, with EP0152R inducing a marked increase in clearance of CB1954, in keeping with model predictions. We detected DNA interstrand cross-links caused by nitroreduced CB1954 in tumor biopsies from treated patients, demonstrating that the activated prodrug exerts its cytotoxic properties through DNA base alkylation.


Subject(s)
Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Neoplasms/drug therapy , Prodrugs/therapeutic use , Quinone Reductases/metabolism , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Aziridines/adverse effects , Aziridines/chemistry , Aziridines/pharmacokinetics , Cell Death/drug effects , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray , DNA, Neoplasm/metabolism , Enzyme Activation/drug effects , Female , Humans , Male , Middle Aged , Models, Molecular , Prodrugs/chemistry , Prodrugs/pharmacology , Quinone Reductases/chemistry
10.
BMC Cancer ; 9: 301, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19712451

ABSTRACT

BACKGROUND: The nitroreductase/5-(azaridin-1-yl)-2,4-dinitrobenzamide (NTR/CB1954) enzyme/prodrug system is considered as a promising candidate for anti-cancer strategies by gene-directed enzyme prodrug therapy (GDEPT) and has recently entered clinical trials. It requires the genetic modification of tumor cells to express the E. coli enzyme nitroreductase that bioactivates the prodrug CB1954 to a powerful cytotoxin. This metabolite causes apoptotic cell death by DNA interstrand crosslinking. Enhancing the enzymatic NTR activity for CB1954 should improve the therapeutical potential of this enzyme-prodrug combination in cancer gene therapy. METHODS: We performed de novo synthesis of the bacterial nitroreductase gene adapting codon usage to mammalian preferences. The synthetic gene was investigated for its expression efficacy and ability to sensitize mammalian cells to CB1954 using western blotting analysis and cytotoxicity assays. RESULTS: In our study, we detected cytoplasmic protein aggregates by expressing GFP-tagged NTR in COS-7 cells, suggesting an impaired translation by divergent codon usage between prokaryotes and eukaryotes. Therefore, we generated a synthetic variant of the nitroreductase gene, called ntro, adapted for high-level expression in mammalian cells. A total of 144 silent base substitutions were made within the bacterial ntr gene to change its codon usage to mammalian preferences. The codon-optimized ntro either tagged to gfp or c-myc showed higher expression levels in mammalian cell lines. Furthermore, the ntro rendered several cell lines ten times more sensitive to the prodrug CB1954 and also resulted in an improved bystander effect. CONCLUSION: Our results show that codon optimization overcomes expression limitations of the bacterial ntr gene in mammalian cells, thereby improving the NTR/CB1954 system at translational level for cancer gene therapy in humans.


Subject(s)
Gene Expression , Genes, Synthetic , Nitroreductases/genetics , Protein Engineering , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Base Sequence , Biotransformation , Cell Line, Tumor , Cell Proliferation/drug effects , Codon , Humans , Molecular Sequence Data , Nitroreductases/metabolism , Prodrugs/pharmacokinetics
11.
J Urol ; 180(1): 116-20, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18485407

ABSTRACT

PURPOSE: We studied the safety, tolerability and pharmacokinetics of a single immediate post-transurethral resection intravesical instillation of apaziquone for patients with nonmuscle invasive bladder cancer. MATERIALS AND METHODS: Patients with cTa-T1, G1-G2 urothelial cell carcinoma of the bladder underwent transurethral resection of bladder tumor(s) followed by a single intravesical instillation of apaziquone 4 mg/40 ml for 1 hour within 6 hours of transurethral bladder tumor resection. Adverse events and safety parameters were assessed on days 8 and 15 after transurethral bladder tumor resection. Blood samples were drawn before and during the instillation for pharmacokinetic analyses. The first 10 patients with pTa-T1, G1-G2 nonmuscle invasive bladder cancer were also evaluated by cystoscopy 3 months after treatment to determine mucosal healing. RESULTS: Of 20 patients receiving apaziquone 13 (65%) reported 35 adverse events, mostly grade 1 to 2. Eight patients (40%) reported 13 adverse events related to treatment, in particular dysuria, hematuria, bladder spasm, abdominal pain, asthenia and postoperative urinary retention. Three grade 3 and 1 grade 4 event(s) occurred, but these were considered unrelated to treatment. No other significant clinical changes were observed. Apaziquone and the active metabolite EO5a were not detected with pharmacokinetic analyses at any point of time. After 3 months no evidence of impaired mucosal healing was observed. CONCLUSIONS: A single immediate post-transurethral bladder tumor resection instillation of apaziquone was well tolerated with an expected good safety profile. Apaziquone and its metabolite EO5a were not detected systemically with pharmacokinetic analyses. These results have lead to further study of a single immediate instillation of apaziquone.


Subject(s)
Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/surgery , Indolequinones/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/surgery , Administration, Intravesical , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Aziridines/administration & dosage , Aziridines/adverse effects , Aziridines/pharmacokinetics , Carcinoma, Transitional Cell/pathology , Combined Modality Therapy , Female , Humans , Indolequinones/administration & dosage , Indolequinones/adverse effects , Indolequinones/pharmacokinetics , Male , Middle Aged , Neoplasm Invasiveness , Prospective Studies , Time Factors , Urinary Bladder Neoplasms/pathology
12.
Prostate Cancer Prostatic Dis ; 11(2): 187-93, 2008.
Article in English | MEDLINE | ID: mdl-17726452

ABSTRACT

The nitroreductase (NR)/CB1954 enzyme prodrug system has given promising results in pre-clinical studies and is currently being assessed in phase I and II clinical trials in prostate cancer. Enhanced cell killing by apparent immune-mediated mechanisms has been shown in pancreatic and colorectal cancer models, by co-expressing murine granulocyte macrophage colony-stimulating factor (GM-CSF) with NR in a single replication deficient adenoviral vector. This consists of the CMV immediate early promotor driving expression of NR, with an internal ribosome entry site (IRES) and the gene for murine GM-CSF (mGM-CSF). To examine if similar enhancement of tumour cell killing could be produced in prostate cancer, the TRAMP model was chosen. Results illustrate that the combination of suicide gene therapy using NR and CB1954, with cytokine stimulation with mGM-CSF gives an improved response compared with either modality alone. The mechanism of this improved response is however likely to be non-immune based as it lacks a memory effect.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Escherichia coli Proteins/metabolism , Genes, Transgenic, Suicide , Genetic Therapy , Genetic Vectors/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Nitroreductases/metabolism , Prodrugs/therapeutic use , Prostatic Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenoviridae/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Aziridines/pharmacokinetics , Aziridines/toxicity , Biotransformation , Combined Modality Therapy , Defective Viruses/genetics , Escherichia coli Proteins/genetics , Genes, Synthetic , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Nitroreductases/genetics , Prodrugs/pharmacokinetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Recombinant Proteins , Specific Pathogen-Free Organisms
13.
J Org Chem ; 72(24): 9391-4, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17958451

ABSTRACT

Catalyzed by Rhodococcus erythropolis AJ270 (whole cell catalyst) under very mild conditions, a number of racemic trans-3-arylaziridine-2-carbonitriles and amides were efficiently transformed into enantiopure 2R,3S-3-arylaziridine-2-carboxamides. While the nitrile hydratase exhibits low selectivity against nitrile substrates, the amidase is highly enantioselective toward 2S,3R-3-arylaziridine-2-carboxamides. Upon the treatment with catalytic hydrogenation, amine, or water in the presence of one equivalent of TFA, the resulting aziridine-2-carboxamides underwent highly efficient and stereospecific ring-opening reactions to produce enantiopure alpha-amino-, alpha,beta-diamino-, and alpha-amino-beta-hydroxy-propanamide derivatives in high yields.


Subject(s)
Amides/pharmacokinetics , Aziridines/pharmacokinetics , Hydro-Lyases/pharmacokinetics , Nitriles/pharmacokinetics , Rhodococcus/enzymology , Amides/chemistry , Amines/chemistry , Aziridines/chemistry , Biotransformation , Catalysis , Hydrogenation , Models, Chemical , Nitriles/chemistry , Stereoisomerism , Trifluoroacetic Acid/chemistry , Water/chemistry
14.
Toxicology ; 240(1-2): 70-85, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17804142

ABSTRACT

5-(Aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954), a promising anti-tumour compound, is associated with clinical hepatotoxicity. We have previously demonstrated that human liver preparations are capable of endogenous 2- and 4-nitroreduction of CB 1954 to generate highly potent cytotoxins. The present study initially examined the in vitro metabolism of CB 1954 in S9 preparations of several non-clinical species and strains. The CD-1 nu/nu mouse and Sprague-Dawley rat were subsequently chosen for further assessment of in vivo metabolism and hepatotoxicity of CB 1954, as well as the mechanisms that may be involved. Animals were administered the maximum tolerated dose (MTD). At 562 micromol/kg, the mouse exhibited transaminase elevation and centrilobular hepatocyte injury. Moreover, thiol adducts as well as hepatic glutathione depletion paralleled temporally by maximal nitroreduction were observed. The rat had a much lower MTD of 40 micromol/kg and showed signs of gastro-intestinal disturbances. In contrast to mouse, peri-portal damage and biliary changes were observed in rat without any alterations in plasma biomarkers or hepatic glutathione levels. Immunohistochemical analysis did not reveal any correlation between the location of injury and expression of cytochrome P450 reductase and NAD(P)H quinone oxidoreductase 1, two enzymes implicated in the bioactivation of this drug. In conclusion, the present study showed that following administration of CB 1954 at the respective MTDs, hepatotoxicity was observed in both mouse and rat. However, the degree of sensitivity to the drug and the mechanisms of toxicity involved appear to be widely different between CD-1 nu/nu mice and Sprague-Dawley rats.


Subject(s)
Antineoplastic Agents , Aziridines , Chemical and Drug Induced Liver Injury/etiology , Microsomes, Liver/metabolism , Prodrugs , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Aziridines/blood , Aziridines/pharmacokinetics , Aziridines/toxicity , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dogs , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , Humans , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/drug effects , Microsomes, Liver/pathology , NAD(P)H Dehydrogenase (Quinone)/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Rats , Rats, Sprague-Dawley , Species Specificity
15.
J Urol ; 176(4 Pt 1): 1344-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16952628

ABSTRACT

PURPOSE: The quinone based bioreductive drug apaziquone (EO9) failed to demonstrate efficacy in previous phase II studies following intravenous administration. We determined the dose of apaziquone that can be safely administered intravesically and explored its activity for superficial bladder transitional cell carcinoma. MATERIALS AND METHODS: Six patients with multifocal, Ta/T1 and G1/G2 transitional cell carcinoma of the bladder received escalating doses of apaziquone formulated as EOquintrade mark (0.5 mg/40 ml up to 16 mg/40 ml) weekly for 6 weeks. A further 6 patients received weekly apaziquone at the highest nontoxic dose established. Pharmacokinetic parameters were determined in urine and blood, and the pharmacodynamic markers NQO1 (reduced nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase-1) and glucose transporter 1 were also characterized. Efficacy was determined against a marker lesion. RESULTS: Local toxicity (grades 2 and 3 dysuria, and hematuria) was observed at doses of 8 mg/40 ml and above but 4 mg/40 ml was well tolerated with no systemic or local side effects. Apaziquone in urine increased linearly with the dose but no apaziquone was detected in plasma. In 8 of 12 patients complete macroscopic and histological disappearance of the marker lesion occurred. A correlation between response and NQO1 and/or glucose transporter 1 expression could not be established. CONCLUSIONS: Intravesical administration of 4 mg/40 ml apaziquone was well tolerated and had ablative activity against superficial bladder cancer marker lesions.


Subject(s)
Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Carcinoma, Transitional Cell/drug therapy , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Carcinoma, Transitional Cell/pathology , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Indolequinones/pharmacokinetics , Male , Middle Aged , Pilot Projects , Treatment Outcome , Urinary Bladder Neoplasms/pathology
16.
Anticancer Agents Med Chem ; 6(4): 281-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842231

ABSTRACT

Solid tumours contain regions of very low oxygen concentrations that are said to be hypoxic. Hypoxia is a natural phenotype of solid tumours resulting from an imperfect vascular network. There are a number of consequences associated with tumour hypoxia including: resistance to ionising radiation, resistance to chemotherapy and the magnification of mutated p53. In addition tissue hypoxia has been regarded as a key factor for tumour aggressiveness and metastasis by activation of signal transduction pathways and gene regulatory mechanisms. It is clear that hypoxia in solid tumours promotes a strong oncogenic phenotype and is a phenomenon that occurs in all solid tumours. As such this provides a significant target for drug discovery particularly for tumour-targeting agents. A range of chemical classes (N-oxides, quinones, nitro-aromatics) have been explored as bioreductive agents that target tumour hypoxia. The most advanced agent, tirapazamine, is in phase III clinical trials in combination with cis-platin. The aim of this review is to give a brief overview of the current molecules and strategies being explored for targeting tumour hypoxia.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Hypoxia/drug effects , Neoplasms/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Anthraquinones/pharmacokinetics , Anthraquinones/therapeutic use , Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Aziridines/therapeutic use , Benzoquinones/pharmacokinetics , Benzoquinones/therapeutic use , Clinical Trials, Phase III as Topic , Drug Screening Assays, Antitumor , Humans , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Indolequinones/pharmacokinetics , Indolequinones/therapeutic use , Neoplasms/blood supply , Neoplasms/metabolism , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Radiation-Sensitizing Agents/pharmacokinetics , Tirapazamine , Triazines/pharmacokinetics , Triazines/therapeutic use
17.
Cancer Chemother Pharmacol ; 58(4): 532-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16470409

ABSTRACT

OBJECTIVES: The objectives of this study were to characterize pharmacokinetics of N-2-chloroethylaziridine (CEA) in the rat model and assess the in vivo fraction of total clearance of phosphoramide mustard (PM) that furnished CEA to circulation. METHODS: The disposition of CEA was investigated following separate intravenous (iv) administrations of PM, synthetic CEA, and their combination to the Sprague-Dawley rats. In addition, in rats receiving prodrug cyclophosphamide (CP), plasma concentrations of CP and its metabolites, 4-hydroxycyclophosphamide (HOCP), PM, and CEA, were simultaneously quantified using GC/MS and stable isotope dilution techniques. RESULTS: Following iv administration of synthetic CEA, concentrations of CEA declined biexponentially with the mean terminal half-life and total body clearance of 47.5 min and 167 ml/min/kg, respectively. Urinary excretion of unchanged CEA was 0.164% of the administered dose. CEA was found to be the major circulating metabolite after iv administration of precursor PM to rats. The fraction of total clearance of PM that furnished CEA to circulation was estimated to be 100%, indicating virtually complete availability of the metabolite to circulation once formed. In rats administered with CP, PM exhibited the highest plasma and urinary concentrations compared to HOCP and CEA. CONCLUSIONS: For the first time, CEA was demonstrated to be an important in vivo metabolite of CP in the present study. In light of the poor permeability and in vivo stability of PM, the ultimate DNA alkylator, the findings obtained in this study suggested that CEA may contribute significantly to the overall antitumor activity of prodrug CP.


Subject(s)
Aziridines/pharmacokinetics , Phosphoramide Mustards/metabolism , Animals , Area Under Curve , Aziridines/blood , Gas Chromatography-Mass Spectrometry , Half-Life , Humans , Male , Metabolic Clearance Rate , Phosphoramide Mustards/pharmacokinetics , Protein Binding , Rats , Rats, Sprague-Dawley
18.
Clin Cancer Res ; 11(7): 2695-701, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15814651

ABSTRACT

PURPOSE: The purpose of our study was to investigate the cellular accumulation, DNA cross-linking ability, and cellular toxicity of RH1 (2,5-diaziridinyl-3-[hydroxymethyl[-6-methyl-1,4-benzoquinone), a novel DNA alkylating agent currently in clinical trials. In addition, the in vivo efficacy of RH1 formulated in different vehicles was also compared. EXPERIMENTAL DESIGN: RH1 is activated by the two-electron reducing enzyme NQO1 [NADPH:quinone oxidoreductase] forming a potent cytotoxic agent that cross-links DNA. We have used whole blood, cell lines, and primary explanted tumor cultures to measure both the cellular accumulation, DNA cross-linking, and cytotoxicity of RH1. Furthermore, the pharmacokinetic and pharmacodynamic characteristics of RH1 formulated in different vehicles were measured in vivo using the validated comet-X assay in mice bearing human tumor xenografts. RESULTS: Accumulation of RH1 was shown to be both time and concentration dependent, reaching a maximum after 2 hours and correlated well with DNA cross-linking measurements. DNA cross-linking in vitro could be detected at low (1-10 nmol/L) concentrations after as little as 2 hours exposure. In primary tumor cultures, RH1 induces much higher levels of DNA cross-links at lower doses than either mitomycin C or cisplatin. In vivo efficacy testing using polyvinyl pyrrolidone, saline, or cyclodextrin as vehicles showed DNA cross-links readily detectable in all tissues examined and was enhanced when given in cyclodextrin compared with polyvinyl pyrrolidone or saline. CONCLUSIONS: RH1 represents a potent bioreductive anticancer drug, which may prove effective in the treatment of cancers, particularly those that overexpress NQO1. DNA cross-linking can be reliably measured in tissue using the validated comet-X assay.


Subject(s)
Aziridines/pharmacology , Benzoquinones/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Aziridines/blood , Aziridines/pharmacokinetics , Benzoquinones/blood , Benzoquinones/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Comet Assay/methods , Cross-Linking Reagents/pharmacology , DNA/chemistry , DNA/genetics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Nude , Treatment Outcome , Tritium
20.
J Clin Oncol ; 22(9): 1546-52, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15051757

ABSTRACT

PURPOSE: Virus-directed enzyme prodrug therapy depends on selective delivery of virus encoding a prodrug-activating enzyme to tumor, followed by systemic treatment with prodrug to achieve high levels of the activated cytotoxic at the intended site of action. The use of the bacterial enzyme nitroreductase to activate CB1954 (5-(aziridin-1-yl)-2,4-dinitrobenzamide) to a short lived, highly toxic DNA cross-linking agent has been demonstrated in tumor xenografts. In this study, we report the first clinical trial investigating the feasibility, safety, and transgene expression of a replication-defective adenovirus encoding nitroreductase (CTL102) in patients with liver tumors. PATIENTS AND METHODS: Patients with resectable primary or secondary (colorectal) liver cancer received a single dose of CTL102 delivered by direct intratumoral inoculation 3 to 8 days before surgical resection. RESULTS: Eighteen patients were treated with escalating doses of CTL102 (range, 10(8)-5 x 10(11) virus particles). The vector was well tolerated with minimal side effects, had a short half-life in the circulation, and stimulated a robust antibody response. Dose-related increases in tumoral nitroreductase expression measured by immunohistochemical analysis have been observed. CONCLUSION: Direct intratumoral inoculation of CTL102 to patients with primary and secondary liver cancer is feasible and well tolerated. The high level of nitroreductase expression observed at 1 to 5 x 10(11) virus particles mandates further studies in patients with inoperable tumors who will receive CTL102 and CB1954.


Subject(s)
Adenoviridae/genetics , Aziridines/metabolism , Aziridines/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/secondary , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/pharmacology , Gene Expression Regulation , Genetic Therapy/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Nitroreductases/genetics , Nitroreductases/pharmacology , Prodrugs/administration & dosage , Prodrugs/therapeutic use , Transgenes , Adenoviridae/pathogenicity , Aged , Aziridines/pharmacokinetics , DNA Adducts , Female , Flavoproteins , Gene Transfer Techniques , Genetic Vectors , Half-Life , Humans , Immunohistochemistry , Male , Middle Aged , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...