Metabolic activation of temozolomide measured in vivo using positron emission tomography.

The purpose of this research was to quantitate and confirm the mechanism of in vivo metabolic activation of temozolomide. The secondary aims were to evaluate the tumor, normal tissue, and plasma pharmacokinetics of temozolomide in vivo, and to determine whether such pharmacokinetics resulted in tumor targeting. [(11)C]temozolomide kinetics were studied in men using positron emission tomography (PET). It has been postulated that temozolomide undergoes decarboxylation and ring opening in the 3-4 position to produce the highly reactive methyldiazonium ion that alkylates DNA. To investigate this, a dual radiolabeling strategy, with [(11)C]temozolomide separately radiolabelled in the 3-N-methyl and 4-carbonyl positions, was used. We hypothesized that (11)C in the C-4 position of [4-(11)C-carbonyl]temozolomide would be converted to [(11)C]CO(2) if the postulated mechanism of metabolic conversion was true resulting in lower [(11)C]temozolomide tumor exposure. Paired studies were performed with both forms of [(11)C]temozolomide in 6 patients with gliomas. Another PET scan with (11)C-radiolabelled bicarbonate was performed and used to account for the metabolites of temozolomide using a data-led analytical approach. Plasma was analyzed for [(11)C]temozolomide and [(11)C]metabolites throughout the scan duration. Exhaled air was also sampled throughout the scan for [(11)C]CO(2). The percentage ring opening of temozolomide over 90 min was also calculated to evaluate whether there was a differential in metabolic breakdown among plasma, normal tissue, and tumor. There was rapid systemic clearance of both radiolabelled forms of [(11)C]temozolomide over 90 min (0.2 liter/min/m(2)), with [(11)C]CO(2) being the primary elimination product. Plasma [(11)C]CO(2) was present in all of the studies with [4-(11)C-carbonyl]temozolomide and in half the studies with [3-N-(11)C-methyl]temozolomide. The mean contributions to total plasma activity by [(11)C]CO(2) at 10 and 90 min were 12% and 28% with [4-(11)C-carbonyl]temozolomide, and 1% and 4% with [3-N-(11)C-methyl]temozolomide, respectively. There was a 5-fold increase in exhaled [(11)C]CO(2) sampled with [4-(11)C-carbonyl]temozolomide compared with [3-N-(11)C-methyl]temozolomide (P < 0.05). A decrease in tissue exposure [area under the curve between 0 and 90 min (AUC(0-90 min))] to [(11)C]temozolomide was also observed with [4-(11)C-carbonyl] temozolomide compared with [3-N-(11)C-methyl]temozolomide. Of potential therapeutic advantage was the higher [(11)C]radiotracer and [(11)C]temozolomide exposure (AUC(0-90 min)) in tumors compared with normal tissue. [(11)C]temozolomide ring opening over 90 min was less in plasma (20.9%; P < 0.05) compared with tumor (26.8%), gray matter (29.7%), and white matter (30.1%), with no differences (P > 0.05) between tumor and normal tissues. The significantly higher amounts of [(11)C]CO(2) sampled in plasma and exhaled air, in addition to the lower normal tissue and tumor [(11)C]temozolomide AUC(0-90 min) observed with [4-(11)C-carbonyl]temozolomide, confirmed the postulated mechanism of metabolic activation of temozolomide. A higher tumor [(11)C]temozolomide AUC(0-90 min) in tumors compared with normal tissue and the tissue-directed metabolic activation of temozolomide may confer potential therapeutic advantage in the activity of this agent. This is the first report of a clinical PET study used to quantify and confirm the in vivo mechanism of metabolic activation of a drug.

[1]  A. Gescher,et al.  Comparison of the cytotoxicity in vitro of temozolomide and dacarbazine, prodrugs of 3-methyl-(triazen-1-yl)imidazole-4-carboxamide , 2004, Cancer Chemotherapy and Pharmacology.

[2]  S. Osman,et al.  Extraction of 5-fluorouracil by tumor and liver: a noninvasive positron emission tomography study of patients with gastrointestinal cancer. , 2001, Cancer research.

[3]  T Jones,et al.  Pharmacokinetic evaluation of N-[2-(dimethylamino)ethyl]acridine-4-carboxamide in patients by positron emission tomography. , 2001, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  S. Osman,et al.  Modulation of fluorouracil tissue pharmacokinetics by eniluracil: in-vivo imaging of drug action , 2000, The Lancet.

[5]  A general method to correct PET data for tissue metabolites using a dual-scan approach. , 2000, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[6]  S. Osman,et al.  On-line measurement of exhaled [11C]CO2 during PET. , 2000, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[7]  S. Osman,et al.  Tumor, normal tissue, and plasma pharmacokinetic studies of fluorouracil biomodulation with N-phosphonacetyl-L-aspartate, folinic acid, and interferon alfa. , 1999, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  L. Grochow,et al.  Absorption, metabolism, and excretion of 14C-temozolomide following oral administration to patients with advanced cancer. , 1999, Clinical cancer research : an official journal of the American Association for Cancer Research.

[9]  Julian C. Matthews,et al.  Pharmacokinetic assessment of novel anti-cancer drugs using spectral analysis and positron emission tomography: A feasibility study , 1998, Cancer Chemotherapy and Pharmacology.

[10]  E S Newlands,et al.  Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. , 1997, Cancer treatment reviews.

[11]  E. Newlands,et al.  The Charing Cross Hospital experience with temozolomide in patients with gliomas. , 1996, European journal of cancer.

[12]  M. Stevens,et al.  Antitumor imidazotetrazines. 32. Synthesis of novel imidazotetrazinones and related bicyclic heterocycles to probe the mode of action of the antitumor drug temozolomide. , 1995, Journal of medicinal chemistry.

[13]  N. Bleehen,et al.  Cancer Research Campaign phase II trial of temozolomide in metastatic melanoma. , 1995, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[14]  M. Stevens,et al.  NMR and molecular modeling investigation of the mechanism of activation of the antitumor drug temozolomide and its interaction with DNA. , 1994, Biochemistry.

[15]  Nucleophilic [18F]radiofluorination using microwave cavity: Application to [18F]FDG and aromatic [18F]fluoro amino acids synthesis , 1993 .

[16]  E. Newlands,et al.  Depletion of O6-alkylguanine-DNA alkyltransferase correlates with potentiation of temozolomide and CCNU toxicity in human tumour cells. , 1993, British Journal of Cancer.

[17]  T. Jones,et al.  Spectral Analysis of Dynamic PET Studies , 1993, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[18]  T J Spinks,et al.  Physical performance of a positron tomograph for brain imaging with retractable septa. , 1992, Physics in medicine and biology.

[19]  J B Bassingthwaighte,et al.  Contribution of labeled carbon dioxide to PET imaging of carbon-11-labeled compounds. , 1992, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[20]  E. Newlands,et al.  Phase I trial of temozolomide (CCRG 81045: M&B 39831: NSC 362856). , 1992, British Journal of Cancer.

[21]  N. Gibson,et al.  DNA sequence specificity of guanine N7-alkylations for a series of structurally related triazenes. , 1988, Carcinogenesis.

[22]  S P Langdon,et al.  Antitumor activity and pharmacokinetics in mice of 8-carbamoyl-3-methyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG 81045; M & B 39831), a novel drug with potential as an alternative to dacarbazine. , 1987, Cancer research.

[23]  M. Tisdale Antitumor imidazotetrazines--XV. Role of guanine O6 alkylation in the mechanism of cytotoxicity of imidazotetrazinones. , 1987, Biochemical pharmacology.

[24]  C. G. Newton,et al.  Antitumor imidazotetrazines. 1. Synthesis and chemistry of 8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d]-1,2,3,5-tetrazin-4(3 H)-one , a novel broad-spectrum antitumor agent. , 1984, Journal of medicinal chemistry.

[25]  R A Brooks,et al.  Theory of image reconstruction in computed tomography. , 1975, Radiology.