Abstract
Advances in analytical methods, imaging techniques and an increased understanding of the influence of pharmacogenetic factors have added to our knowledge of the pharmacology of many chemotherapeutic agents. Extending the use of these approaches to pharmacodynamic end-points, together with the application of population-based modelling techniques, offers the potential to develop truly individualized therapy in the future.
Keywords: Cancer, pharmacology, modelling, pharmacogenetics
The concept that major improvements in cancer treatment could come about simply by increasing our knowledge of the systemic pharmacology of cytotoxic drugs has not been borne out by many of the studies performed in the last 20 years. Data on plasma concentrations of drug and metabolites have allowed the successful development of a number of novel agents and has been useful in guiding dosage regimens and the design of novel pro-drugs. However, the goal of using clinical pharmacology to individualize therapy in order to optimize the treatment of each tumour in every patient has not been achieved. Clinical pharmacology studies continue to generate more detailed data on classical cytotoxic drugs. However, to date only for carboplatin [1, 2] and possibly for methotrexate [3] and etoposide [4] is there any rationale for individualized dosing based solely on a knowledge of systemic pharmacology. In this review, I will focus on the methodological and conceptual developments that have augmented our understanding of ‘classical cytotoxic’ drugs, beyond simple measurements of total drug or metabolites in plasma. These developments include new technologies, integration of genetic and other biological determinants of pharmacology and novel statistical and data analysis approaches.
For the purposes of this discussion, classical cytotoxic chemotherapy is defined as treatment involving agents that have a nonspecific effect on cell division or cause cell death by means of damaging DNA, directly or indirectly. Many of these drugs were applied clinically before any detailed knowledge of their mechanism of action was available. To this extent, classical agents are distinct from more recently developed, targeted approaches, where drugs are designed specifically to interact with a defined oncogenic target [5, 6]. Although lessons learnt from the pharmacology of ‘classical’ cytotoxic drugs may have some relevance for more specific agents, these do not relate to any specific pharmacological properties.
Recent advances in clinical pharmacology can be broadly divided up into four main areas. These are:
Systemic pharmacology, based on measurements of drug or metabolites in blood. Advances in this area include improvements in analytical methodology and advances in pharmacokinetic modelling of data.
Pharmacodynamic end-points. This is a broad area taking in measurements of drug action in blood and possibly in the tumour tissue itself. Other advances in this area include development of novel imaging techniques for drug, metabolites and surrogate markers.
Pharmacogenetic and gene regulatory determinants of response and toxicity have been investigated for a number of drugs. Many more polymorphisms in enzymes and transporter proteins relevant to cytotoxic drugs have now been identified and there have been developments in the techniques applied to identify such genetic variants and to assign genotypes. The influence of epigenetics on response of tumours to drug treatment has also been increasingly recognized.
The mathematical techniques applied to pharmacokinetic modelling have recently been extended to incorporate models of pharmacodynamic response. Future developments in this area are likely to include elements of the other three areas mentioned above.
I will deal with each of these areas in turn, but it is important to recognize the extensive areas of overlap between them and the potential for integration of each of these developments.
Systemic pharmacology, analytical methodology and pharmacokinetic modelling
The last 15 years has seen significant developments in the analytical methods applied in cancer pharmacology [7, 8]. Most notable in these developments has been the advent of affordable, bench-top mass spectrometry as a method of detection, combined with liquid chromatography (LC-MS) [9]. Such instrumentation is now available to the nonspecialist with minimal training. The technique of LC-MS makes possible the determination of nanomolar or picomolar concentrations of drugs in body fluids (e.g. vincristine [10], actinomycin-D [11], cyclophosphamide [12, 13]). This increase in sensitivity permits the detection of species present at low concentrations, such as the active 4-hydroxy metabolites of cyclophosphamide [12] or 5-fluorouracil (5-FU) formed from capecitabine [14]. There is also the potential to use microlitre volumes of plasma in assay methods, which is of particular importance in studies in children with cancer (e.g. for etoposide [15]). Finally, the greater sensitivity and specificity of LC-MS over other LC detection methods means that drug and metabolites can be analysed in cells or tissues other than whole plasma, for example unbound etoposide [15] or cisplatin [16] in plasma water, methotrexate in cerebrospinal fluid [17] or fludarabine in leukaemic cells [18].
A related development is the combination of time-of-flight (TOF) mass spectrometers, which can be combined with a variety of sample separation and presentation methods to provide high-resolution mass spectra from biological samples. Such TOF instruments have applications in areas including metabolomic [19] and proteomic profiling [20]. Profiling of proteins and other biochemicals may be useful in characterizing tumours [21], detecting tumour material in peripheral blood or characterizing the response of a patient to a particular drug, by comparing profiles in tumour or blood before and after treatment [22].
Both the metabolism and mechanisms of action of anticancer drugs are often complex. Using pharmacokinetic modelling to gain insights into the action of these drugs is complicated by the low concentrations and unstable nature of important metabolites and by the fact that the relevant biological site is often removed from the most convenient site to sample (i.e. plasma). Many drugs are processed intracellularly and, although accessible cell populations such as peripheral blood lymphocytes (PBL) or erythrocytes [23] may serve as a surrogate, metabolism in tumour cells may differ from that in normal tissues. Indeed, many drugs rely on such differences to achieve selectivity towards tumour cells [24]. Population pharmacokinetic techniques have been employed to generate sophisticated models which provide information on the systemic and tumour pharmacology of many drugs used in cancer [25–28]. Incorporation of pharmacogenetic covariates (see below) and other biochemical information has increased the predictive power of these models in the individualization of cancer treatment [29].
A number of models have been developed in an attempt to predict the concentrations of active metabolites in tumour cells [26, 30]. These models have been developed with knowledge from in vitro experiments and animal tumour models [31, 32]. However, extrapolating to human tumours inevitably involves a number of assumptions [33]. Population approaches have also been successfully applied to the modelling of haematological toxicities (see below). The incorporation of data from novel sophisticated imaging methods (see below) will, it is to be hoped, improve the utility of these models.
Pharmacodynamic end-points
In cancer chemotherapy, pharmacodynamic end-points have always provided challenges and hurdles in understanding the pharmacology of cytotoxic drugs. The end-point that matters is, ultimately, the survival of the patient. Intermediate end-points, such as shrinkage or disappearance of the tumour, may be predictive of long-term survival. Classification of complete or partial responses is dependent on appropriate imaging or other tumour detection methods. In routine clinical practice, the assignment of a pharmacodynamic end-point to the action of a single drug is often complicated by the use of multiple agents in a single regimen.
Developments in detection methods for tumours, such as the ability of polymerase chain reaction to detect the Bcr-Abl gene in chronic myelogenous leukaemia (CML) [34, 35], have redefined the most relevant end-point from complete haematological remission to a so-called molecular remission. That development, in turn, has become meaningful only with the development of imatinib as a novel effective treatment for CML [6]. The development of imatinib, which inhibits the tyrosine kinase activity of the Bcr-Abl protein, is an example of a novel cancer treatment directed towards a known biochemical target selective to tumour cells. As the prototype for other such targeted therapies, imatinib is in contrast to conventional cytotoxic drugs that, at best, exploit the higher proliferation rates of tumour cells over normal tissues.
Investigations of pharmacodynamic end-point with conventional agents have relied on detection and quantification of drug—target interactions. For many of these drugs, the target is DNA and pharmacodynamic methods have been developed to measure DNA adducts, DNA damage or incorporation of nucleoside analogues into DNA [36]. Thus, a number of techniques have been developed to measure platinum DNA adducts, in the case of cisplatin, carboplatin and other analogues [37–39]. These techniques have been mainly applied to DNA from surrogate tissues such as PBL. The covalent binding of alkylating agents to DNA results in DNA cross-links or strand breaks. These lesions in DNA may be detected indirectly using the COMET assay [40]. So far, most studies have found little or no relationship between the degree of adduct formation and either pharmacokinetic end-points such as the area under the plasma concentration time curve (AUC), or clinical outcome (toxicity or antitumour effect). As with DNA reactive drugs, the incorporation of nucleotide analogues into DNA has been quantified in surrogate tissues such as PBL or erythrocytes. Such measurements has been possible for 6-mercaptopurine [23], gemcitabine [41, 42], fludarabine [18] and cytarabine [43].
Measurements in PBL have some worth in demonstrating that the drug has been appropriately activated and is able to interact with the target. However, there is no information regarding the effects of the drug in the tissue of interest, i.e. the tumour. Advances in imaging techniques have permitted some modest insights into the pharmacology of antitumour agents in tumours in patients [44]. The main advances in imaging have been the widening availability of positron emission tomography (PET). The diagnostic use of PET to grade and determine the response of solid tumours is now established practice in many countries [45, 46]. Research applications of PET are still limited by the short half-life of positron-emitting isotopes and the scarcity of cyclotron facilities. Nevertheless, a number of interesting studies have been performed. Labelling of the methylating group of temozolomide with 11C (half-life 20 min) has shown that temozolomide methylates DNA in brain tumours and also shows the time course of repair of this lesion [47]. Thymidine labelled with 11C can be used to demonstrate that tumours increase thymidine salvage when thymidylate synthase is inhibited, suggesting a number of approaches for augmenting the activity of these drugs [48]. Since fluorine has one of the more stable positron-emitting isotopes, labelling a compound with 18F (half-life 110 min), either at an existing fluorine or as a fluorine analogue, has been used in a number of studies. The major diagnostic use of PET uses 18fluoro-deoxyglucose [46]. Drugs such as 5-fluorouracil have been studied using PET [49, 50]. The complex data derived from these experiments require intensive mathematical models to distinguish vascular background and the contribution of metabolites from the signal for parent drug [51]. This highlights one of the current drawbacks with PET, that parent drug and any metabolite retaining the positron-emitting label will be detected indiscriminately.
Fluorine chemistry provides the opportunity for another potentially useful imaging technique. Magnetic resonance spectroscopy (MRS) imaging provides structural identity information, as well as some quantitative data on compounds which have a fluorine group [52]. This approach has been applied to 5-fluorouracil [53, 54], but is limited to tumours near to the body surface, due to the limited range of the MRS signal. Phosphorus is another possible nucleus which may be suitable for in vivo MRS imaging and has been applied to the study of ifosfamide and cyclophosphamide metabolism on urine samples [55] and in tumours. So far, results with MRS have been rather limited, due to the narrow range of compounds for which the technique is appropriate and limits on sensitivity and range of the available scanners.
Genetics and regulation of drug-metabolizing enzymes, transporter proteins and cytotoxic response genes
Advances in this broad category depend on an increased understanding of the role of drug metabolism in the activation and inactivation of cytotoxic drugs, the role of transport proteins in determining hepatic uptake and biliary and renal excretion and the role of DNA repair and other proteins in modulating the response to cytotoxic drug action.
It has long been known that CYP enzymes are important in the activation and inactivation of drugs such as cyclophosphamide, docetaxel and etoposide. However, attempts to individualize therapy based on phenotypic enzyme activity, usually based on the elimination of a marker compound, have met with only limited success [56]. The role of genotype in a number of well-characterized polymorphisms in CYP2B6, CYP2C8 and CYP3A4 and 3A5 is now being investigated in the context of the pharmacology and clinical effect of cyclophosphamide [57, 58] and paclitaxel [29], with some intriguing preliminary results. Similarly, a critical role for UGT enzymes in the inactivation of camptothecins has suggested that dose individualization of irinotecan could be achieved by determining the number of TATA repeats in the regulatory region of the UGT1A1 gene [59], or by genotyping for polymorphisms in UGT1A7 or 1A9 [60].
These pharmacogenetic investigations have built on the examples of polymorphisms in dihydropyrimidine dehydrogenase (DPD) [61] and in thiopurine methyltransferase (TPMT) [62], influencing the pharmacology of 5-fluorouracil and 6-mercaptopurine, respectively. The inactivating genetic variants in DPD and TPMT are relatively rare and attention is now focusing on more frequent variants, with influences on pharmacology that are likely to be more subtle than complete ablation of a metabolic pathway. The role of single nucleotide polymorphisms (SNPs) in other genes which are involved in the systemic and cellular pharmacology of drugs such as nucleotide analogues [63], antifolates [64], bioreductive agents [65] and substrates for various drug transports [66, 67] is now under investigation. Many of the SNPs are in regulatory regions of the genes involved and the regulation of the corresponding enzymes and transporter proteins is an area of increasing interest and activity.
Regulation of other genes whose protein products influence response to treatment is another area where there have been some exciting recent advances. The repair of DNA methylation by the enzyme methylguanine methyltransferase (MGMT) has long been associated with resistance to temozolomide. Tumours with high expression of MGMT are relatively resistant to temozolomide in cell lines, xenografts and in clinical studies. Several recent studies have reported that silencing of the MGMT gene by promoter methylation in tumours is associated with a more favourable response to temozolomide treatment [68, 69]. Resistance to temozolomide may also result from a deficiency in one of the mismatch repair proteins, which have been shown to have an important role in the recognition and processing of alkylated DNA lesions [70, 71]. Loss of mismatch repair function is linked to failure to undergo apoptosis and silencing of MLH1 is due to promoter methylation [71]. Thus, reversal of MLH1 promoter methylation may improve the response to temozolomide and to other drugs, such as cisplatin or carboplatin, which require an intact mismatch repair pathway to cause a cytotoxic effect. Drugs such as decitabine, which inactivate DNA methyltransferase enzymes, can restore the sensitivity of tumour cells previously resistant to platinum drugs [72]. Clinical studies of decitabine in combination as a demethylating agent are ongoing [73]. Obviously, there is a contradiction here, in that methylation of MGMT is a good thing in blocking a DNA repair pathway, whereas methylation of MLH1 is a bad thing as that pathway is required for an antitumour effect. The exact role of gene methylation and the role of other epigenetic factors such as histone acetylation require further research before they can be exploited clinically to full effect.
Integrated modelling of pharmacokinetic, pharmacodynamic, pharmacogenetic and imaging data
Pharmacodynamic modelling in cancer pharmacology is handicapped by the dissociation, both temporal and physical, between drug presence in a measurable compartment (often plasma) and the pharmacological effect. As noted above, major toxicities such as myelosuppression or diarrhoea occur hours or days after detectable drug has been eliminated from plasma. Likewise, any measurable antitumour effect will not be seen for days or weeks after drug administration. To identify any relationship between plasma concentrations of drug and either toxicity or antitumour effect, investigators have resorted to summary measures of drug exposure (e.g. AUC or time above threshold concentration). For instance, the pharmacological effects of carboplatin have been closely correlated with AUC for ultrafiltrable platinum [2]. Paclitaxel haematological toxicity and antitumour effect in ovarian cancer have been associated with the time for which plasma concentrations exceed 0.05 µm[74].
More recently, physiological models of haematopoiesis or tumour growth have been combined with pharmacokinetic data to generate holistic models of drug action, which may be predictive of pharmacological effects for a number of drugs [75, 76]. Whether these models can be extended to incorporate the combined effects of a number of agents used in combination is not yet clear. Likewise, the incorporation of genotype as a covariate in population models has been reported for irinotecan [77] and paclitaxel [29]. These models may be useful in identifying genotype influence on the phenotype of drug metabolism. A further step would be to combine all of the above information, perhaps together with data from PET or magnetic resonance tumour imaging.
While the logical extension of combining all of the above modelling approaches to achieve a single integrated model of drug action may appear attractive, it should be recognized that the uncertainty involved in each of the components may be so large as to make such a model meaningless. Intersubject and interoccasion variability in the magnitude and nature of the influences on drug effect mean that any model developed in a particular population may lack wider application outside that patient group. Nevertheless, important information concerning the mechanism of action of a drug and the influence of important factors affecting drug action may be gleaned by such an approach. With the advent of microarray-based approaches, information on genes and proteins that influence drug action will be increasingly available, as will information on those genes and proteins which are in turn affected by the drug itself. Managing all of these data represents an increase in the order of magnitude of data that could be explained by a particular pharmacological model. Such a task would require the development of ever more sophisticated statistical and mathematical techniques, alongside a corresponding increase in computing power.
References
- 1.Calvert AH, Newell DR, Gumbrell LA, O'Reilly S, Burnell M, Boxall FE, Siddik ZH, Judson IR, Gore ME, Wiltshaw E. Carboplatin dosage: prospective evaluation of a simple formula based on renal function. J Clin Oncol. 1989;7:1748–56. doi: 10.1200/JCO.1989.7.11.1748. [DOI] [PubMed] [Google Scholar]
- 2.Jodrell DI, Egorin MJ, Canetta RM, Langenberg P, Goldbloom EP, Burroughs JN, Goodlow JL, Tan S, Wiltshaw E. Relationships between carboplatin exposure and tumor response and toxicity in patients with ovarian cancer. J Clin Oncol. 1992;10:520–8. doi: 10.1200/JCO.1992.10.4.520. [DOI] [PubMed] [Google Scholar]
- 3.Evans WE, Relling MV, Rodman JH, Crom WR, Boyett JM, Ching-Hon P. Conventional compared with individualised chemotherapy for childhood acute lymphoblastic leukemia. N Engl J Med. 1998;338:499–505. doi: 10.1056/NEJM199802193380803. [DOI] [PubMed] [Google Scholar]
- 4.Joel SP, Ellis P, O'Byrne K, Papamichael D, Hall M, Penson R, Nicholls S, O'Donnell C, Constantinou A, Woodhull J, Nicholson M, Smith I, Talbot D, Slevin M. Therapeutic monitoring of continuous infusion etoposide in small-cell lung cancer. J Clin Oncol. 1996;14:1903–12. doi: 10.1200/JCO.1996.14.6.1903. [DOI] [PubMed] [Google Scholar]
- 5.Cobleigh MA, Bogel CL, Tripathy D, Tobert NJ, Scholl S, Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ. Mulitnational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after cheotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–48. doi: 10.1200/JCO.1999.17.9.2639. [DOI] [PubMed] [Google Scholar]
- 6.Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers C. Efficacy and safety of a specific inhibitor of the Bcr-Abl tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–7. doi: 10.1056/NEJM200104053441401. [DOI] [PubMed] [Google Scholar]
- 7.Gelpi E. Biomedical and biochemical applications of liquid-chromatography mass-spectrometry. J Chromatogr. 1995;703:59–80. doi: 10.1016/0021-9673(94)01287-o. [DOI] [PubMed] [Google Scholar]
- 8.Geoghegan KF, Kelly MA. Biochemical applications of mass spectrometry in pharmaceutical drug discovery. Mass Spectrom Rev. 2005;24:347–66. doi: 10.1002/mas.20019. [DOI] [PubMed] [Google Scholar]
- 9.Blair IA, Tilve A. Analysis of anticancer drugs and their metabolites by mass spectrometry. Curr Drug Metab. 2002;3:463–80. doi: 10.2174/1389200023337234. [DOI] [PubMed] [Google Scholar]
- 10.Guo P, Wang XM, Zhou F, Gallo JM. Determination of vincristine in mouse plasma and brain tissues by liquid chromatography-electrospray mass spectrometry. J Chromatogr B-Anal Technol Biomed Life Sci. 2004;809:273–8. doi: 10.1016/j.jchromb.2004.06.025. [DOI] [PubMed] [Google Scholar]
- 11.Veal GJ, Cole M, Errington J, Parry A, Hale J, Pearson ADJ, Howe K, Chisholm JC, Beane C, Brennan B, Waters F, Glaser A, Hemsworth S, McDowel H, Wright Y, Pritchard-Jones K, Pinkerton R, Jenner G, Nicholson J, Elsworth AM, Boddy AV. Pharmacokinetics of dactinomycin in a pediatric patient population: a United Kingdom Children'Cancer Study Group study. Clin Cancer Res. 2005;11:5893–9. doi: 10.1158/1078-0432.CCR-04-2546. [DOI] [PubMed] [Google Scholar]
- 12.Huitema ADR, Tibben MM, Kerbusch T, Kettenes-van den Bosch JJ, Rodenhuis S, Beijnen JH. High performance liquid chromatographic determination of the stabilized cyclophosphamide metabolite 4-hydroxycyclophosphamide in plasma and red blood cells. J Liquid Chromatogr Related Technol. 2000;23:1725–44. [Google Scholar]
- 13.Kalhorn TF, Ren S, Howald WN, Lawrence RF, Slattery JT. Analysis of cyclophosphamide and five metabolites from human plasma using liquid chromatography-mass spectrometry and gas chromatography-nitrogen-phosphorus detection. J Chromatogr B. 1999;732:287–98. doi: 10.1016/s0378-4347(99)00300-x. [DOI] [PubMed] [Google Scholar]
- 14.Guichard SM, Mayer I, Jodrell DI. Simultaneous determination of capecitabine and its metabolites by HPLC and mass spectrometry for preclinical and clinical studies. J Chromatogr B. 2005;826:232–7. doi: 10.1016/j.jchromb.2005.09.010. [DOI] [PubMed] [Google Scholar]
- 15.Pang SK, Zheng NY, Felix CA, Scavuzzo J, Boston R, Blair IA. Simultaneous determination of etoposide and its catechol metabolite in the plasma of pediatric patients by liquid chromatography/tandem mass spectrometry. J Mass Spectrom. 2001;36:771–81. doi: 10.1002/jms.173. [DOI] [PubMed] [Google Scholar]
- 16.Oe T, Tian Y, O'Dwyer PJ, Roberts DW, Malone MD, Bailey CJ, Blair IA. A validated liquid chromatography/tandem mass spectrometry assay for cis-amminedichloro (2-methylpyridine) platinum (II) in human plasma ultrafiltrate. Anal Chem. 2002;74:591–9. doi: 10.1021/ac010792v. [DOI] [PubMed] [Google Scholar]
- 17.Gelderblom H, Baker SD, Zhao M, Verweij J, Sparreboom A. Distribution of paclitaxel in plasma and cerebrospinal fluid. Anti-Cancer Drugs. 2003;14:365–8. doi: 10.1097/00001813-200306000-00007. [DOI] [PubMed] [Google Scholar]
- 18.Kalhorn TF, Ren AG, Slattery JT, McCune JS, Wang J. A highly sensitive high-performance liquid chromatography-mass spectrometry method for quantification of fludarabine triphosphate in leukemic cells. J Chromatogr B-Anal Technol Biomed Life Sci. 2005;820:243–50. doi: 10.1016/j.jchromb.2005.03.034. [DOI] [PubMed] [Google Scholar]
- 19.Fan TWM, Lane AN, Higashi RM. The promise of metabolomics in cancer molecular therapeutics. Curr Opin Mol Therapeutics. 2004;6:584–92. [PubMed] [Google Scholar]
- 20.Kolch W, Mischak H, Pitt AR. The molecular make-up of a tumour: proteomics in cancer research. Clin Sci. 2005;108:369–83. doi: 10.1042/CS20050006. [DOI] [PubMed] [Google Scholar]
- 21.Ippolito JE, Xu J, Jain SJ, Moulder K, Mennerick S, Crowley JR, Townsend RR, Gordon JI. An integrated functional genomics and metabolomics approach for defining poor prognosis in human neuroendocrine cancers. Proc Natl Acad Sci USA. 2005;102:9901–6. doi: 10.1073/pnas.0500756102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Turner SM, Hellerstein MK. Emerging applications of kinetic biomarkers in preclinical and clinical drug development. Curr Opin Drug Discov Devel. 2005;8:115–26. [PubMed] [Google Scholar]
- 23.Davies HA, Lennard L, Lilleyman JS. Variable mercaptopurine metabolism in children with leukaemia. a problem of non-compliance. BMJ. 1993;306:1239–40. doi: 10.1136/bmj.306.6887.1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Budman DR, Meropol NJ, Reigner B, Creaven PJ, Lichtman SM, Berghorn E, Behr J, Gordon RJ, Osterwalder B, Griffin T. Preliminary studies of a novel oral fluoropyrimidine carbamate: Capecitabine. J Clin Oncol. 1998;16:1795–802. doi: 10.1200/JCO.1998.16.5.1795. [DOI] [PubMed] [Google Scholar]
- 25.Karlsson MO, Anehall T, Friberg LE, Henningsson A, Kloft C, Sandstrom M, Xie R. Pharmacokinetic/pharmacodynamic modelling in oncological drug development. Basic Clin Pharmacol Toxicol. 2005;96:206–11. doi: 10.1111/j.1742-7843.2005.pto960310.x. [DOI] [PubMed] [Google Scholar]
- 26.Coustere C, Mentre F, Sommadossi J-P, Diasio RB, Steimer J-L. A mathematical model of the kinetics of 5-fluorouracil and its metabolites in cancer patients. Cancer Chemother Pharmacol. 1991;28:123–9. doi: 10.1007/BF00689701. [DOI] [PubMed] [Google Scholar]
- 27.Gieschke R, Reigner B, Blesch KS, Steimer JL. Population pharmacokinetic analysis of the major metabolites of capecitabine. J Pharmacokinet Pharmacodynamics. 2002;29:25–47. doi: 10.1023/a:1015716617967. [DOI] [PubMed] [Google Scholar]
- 28.Klein CE, Gupta E, Reid JM, Atherton PJ, Sloan JA, Pitot HC, Ratain MJ, Kastrissios H. Population pharmacokinetic model for irinotecan and two of its metabolites, SN-38 and SN-38 glucuronide. Clin Pharmacol Therapeutics. 2002;72:638–47. doi: 10.1067/mcp.2002.129502. [DOI] [PubMed] [Google Scholar]
- 29.Henningsson A, Marsh S, Loos WJ, Karlsson MO, Garsa A, Mross K, Mielke S, Vigano L, Locatelli A, Verweij J, Sparreboom A, McLeod HL. Association of CYP2C8, CYP3A4, CYP3A5 and ABCB1 polymorphisms with the pharmacokinetics of paclitaxel. Clin Cancer Res. 2005;11:8097–104. doi: 10.1158/1078-0432.CCR-05-1152. [DOI] [PubMed] [Google Scholar]
- 30.Chatelut E, Roche H, Plusquellec Y, Peyrille F, Debiasi J, Pujol A, Canal P, Houin G. Pharmacokinetic modelling of plasma and cerebrospinal fluid methotrexate after high-dose intravenous infusion in children. J Pharm Sci. 1991. pp. 730–4. [DOI] [PubMed]
- 31.Simeoni M, Magni P, Cammia C, De Nicolao G, Croci V, Pesenti E, Germani M, Poggesi I, Rocchetti M. Predictive pharmacokinetic—pharmacodynamic modeling of tumor growth kinetics in xenograft models after administration of anticancer agents. Cancer Res. 2004;64:1094–101. doi: 10.1158/0008-5472.can-03-2524. [DOI] [PubMed] [Google Scholar]
- 32.Xu L, Eiseman JL, Egorin MJ, D'Argenio DZ. Physiologically-based pharmacokinetics and molecular pharmacodynamics of 17-(allylamino) 17-demethoxygeldanamycin and its active metabolite in tumor-bearing mice. J Pharmacokinet Pharmacodyn. 2003;30:185–219. doi: 10.1023/a:1025542026488. [DOI] [PubMed] [Google Scholar]
- 33.Gallo JM, Vicini P, Orlansky A, Li SL, Zhou F, Ma JG, Puffer S, Bookman MA, Guo P. Pharmacokinetic model-predicted anticancer drug concentrations in human tumors. Clin Cancer Res. 2004;10:8048–58. doi: 10.1158/1078-0432.CCR-04-0822. [DOI] [PubMed] [Google Scholar]
- 34.Hochhaus A, Weisser A, La Rosee P, Emig M, Muller MC, Saussele S, Reiter A, Kuhn C, Berger U, Hehlmann R, Cross NCP. Detection and quantification of residual disease in chronic myelogenous leukemia. Leukemia. 2000;14:998–1005. doi: 10.1038/sj.leu.2401811. [DOI] [PubMed] [Google Scholar]
- 35.Radich JP, Kopecky KJ, Boldt DH, Head D, Slovak ML, Babu R, Kirk J, Lee A, Kessler P, Appelbaum F, Gehly G. Detection of Bcr-Abl fusion genes in adult acute lymphoblastic-leukemia by the polymerase chain-reaction. Leukemia. 1994;8:1688–95. [PubMed] [Google Scholar]
- 36.McGurk CJ, McHugh PJ, Tilby MJ, Grimaldi KA, Hartley JA. Measurement of covalent drug—DNA interactions at the nucleotide level in cells at pharmacologically relevant doses. Methods Enzymol. 2001. pp. 358–76. [DOI] [PubMed]
- 37.Hengstler JG, Fuchs J, Oesch F. DNA strand breaks and DNA cross-links in peripheral mononuclear blood cells of ovarian cancer patients during chemotherapy with cyclophosphamide/carboplatin. Cancer Res. 1992;52:5622–6. [PubMed] [Google Scholar]
- 38.Reed E, Ostchega Y, Steinberg SM, Yuspa SH, Young RC, Ozols RF, Poirier MC. Evaluation of platinum-DNA adduct levels relative to known prognostic variables in a cohort of ovarian cancer patients. Cancer Res. 1990;50:2256–60. [PubMed] [Google Scholar]
- 39.Peng B, Tilby MJ, English MW, Price L, Pearson ADJ, Boddy AV, Newell DR. Platinum-DNA adduct formation in leucocytes of children in relation to pharmacokinetics after cisplatin and carboplatin therapy. Br J Cancer. 1997;76:1466–73. doi: 10.1038/bjc.1997.579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hartley JM, Spanswick VJ, Gander M, Giacomini G, Whelan J, Souhami RL, Hartley JA. Measurement of DNA cross-linking in patients on ifosfamide therapy using the single cell gel electrophoresis (comet) assay. Clin Cancer Res. 1999;5:507–12. [PubMed] [Google Scholar]
- 41.Buesa JM, Losa R, Fernandez A, Sierra M, Esteban E, Diaz A, Lopez-Pousa A, Fra J. Phase I clinical trial of fixed-dose rate infusional gemcitabine and dacarbazine in the treatment of advanced soft tissue sarcoma, with assessment of gemcitabine triphosphate accumulation. Cancer. 2004;101:2261–9. doi: 10.1002/cncr.20612. [DOI] [PubMed] [Google Scholar]
- 42.Kroep JR, Giaccone G, Voorn DA, Smit EF, Beijnen JH, Rosing H, van Moorsel CJA, van Groeningen CJ, Postmus PE, Pinedo HM, Peters GJ. Gemcitabine and paclitaxel. Pharmacokinetic and pharmacodynamic interactions in patients with non-small-cell lung cancer. J Clin Oncol. 1999;17:2190–7. doi: 10.1200/JCO.1999.17.7.2190. [DOI] [PubMed] [Google Scholar]
- 43.Yamauchi T, Ueda T. A sensitive new method for clinically monitoring cytarabine concentrations at the DNA level in leukemic cells. Biochem Pharmacol. 2005;69:1795–803. doi: 10.1016/j.bcp.2005.03.013. [DOI] [PubMed] [Google Scholar]
- 44.Price P, Griffiths J. Tumour pharmacokinetics? — we do need to know. Lancet. 1994;343:1174–5. doi: 10.1016/s0140-6736(94)92395-7. [DOI] [PubMed] [Google Scholar]
- 45.Macmanus MP, Hicks RJ, Matthews JP, McKenzie A, Rischin D, Salminen EK, Ball DL. Positron emission tomography is superior to computed tomography scanning for response-assessment after radical radiotherapy or chemoreadiotherapy in patients with non-small-cell lung cancer. J Clin Oncol. 2003;21:1285–92. doi: 10.1200/JCO.2003.07.054. [DOI] [PubMed] [Google Scholar]
- 46.Juweid ME, Cheson BD. Positron-emission tomography and assessment of cancer Therapy. N Engl J Med. 2006;354:496–507. doi: 10.1056/NEJMra050276. [DOI] [PubMed] [Google Scholar]
- 47.Saleem A, Brown GD, Brady F, Aboagye EO, Osman S, Luthra SK, Ranicar ASO, Brock CS, Stevens MFG, Newlands E, Jones T, Price P. Metabolic activation of temozolomide measured in vivo using positron emission tomography. Cancer Res. 2003;63:2409–15. [PubMed] [Google Scholar]
- 48.Wells P, Aboagye E, Gunn RN, Osman S, Boddy AV, Taylor GA, Rafi I, Hughes AN, Calvert AH, Price PM, Newell DR. 2-[C-11]thymidine positron emission tomography as an indicator of thymidylate synthase inhibition in patients treated with AG337. J Natl Cancer Inst. 2003;95:675–82. doi: 10.1093/jnci/95.9.675. [DOI] [PubMed] [Google Scholar]
- 49.Harte RJA, Matthews JC, O'eilly SM, Tilsley DWO, Osman S, Brown G, Luthra SJ, Brady F, Jones T, Price PM. Tumor, normal tissue, and plasma pharmacokinetic studies of fluorouracil biomodulation with N-phosphonacetyl-L-aspartate, folinic acid, and interferon alfa. J Clin Oncol. 1999;17:1580–8. doi: 10.1200/JCO.1999.17.5.1580. [DOI] [PubMed] [Google Scholar]
- 50.Saleem A, Yap J, Osman S, Brady F, Suttle B, Lucas SV, Jones T, Price PM, Aboagye EO. Modulation of fluorouracil tissue pharmacokinetics by eniluracil: in-vivo imaging of drug action. Lancet. 2000;355(9221):2125–31. doi: 10.1016/s0140-6736(00)02380-1. [DOI] [PubMed] [Google Scholar]
- 51.Aboagye EO, Saleem A, Cunningham VJ, Osman S, Price PM. Extraction of 5-fluorouracil by tumor and liver: a noninvasive positron emission tomography study of patients with gastrointestinal cancer. Cancer Res. 2001;61:4937–41. [PubMed] [Google Scholar]
- 52.van Laarhoven HWM, Punt CJA, Kamm YJL, Heerschap A. Monitoring fluoropyrimidine metabolism in solid tumors with in vivo F-19 magnetic resonance spectroscopy. Crit Rev Oncol Hematol. 2005;56:321–43. doi: 10.1016/j.critrevonc.2005.03.009. [DOI] [PubMed] [Google Scholar]
- 53.Presant CA, Wolf W, Waluch V, Wiseman C, Kennedy P, Blayney D, Brechner RR. Association of intratumoral pharmacokinetics of fluorouracil with clinical-response. Lancet. 1994;343(8907):1184–7. doi: 10.1016/s0140-6736(94)92399-x. [DOI] [PubMed] [Google Scholar]
- 54.Findlay MPN, Leach MO, Cunningham D, Collins DJ, Payne GS, Glaholm J, Mansi JL, McCready VR. The noninvasive monitoring of low-dose, infusional 5-fluorouracil and its modulation by interferon-alpha using in-vivo F-19 magnetic-resonance spectroscopy in patients with colorectal-cancer — a pilot-study. Ann Oncol. 1993;4:597–602. doi: 10.1093/oxfordjournals.annonc.a058595. [DOI] [PubMed] [Google Scholar]
- 55.Joqueviel C, Gilard V, Martino R, MaletMartino M, Niemeyer U. Urinary stability of carboxycyclophosphamide and carboxyifosfamide, two major metabolites of the anticancer drugs cyclophosphamide and ifosfamide. Cancer Chemother Pharmacol. 1997;40:391–9. doi: 10.1007/s002800050676. [DOI] [PubMed] [Google Scholar]
- 56.Goh BC, Lee SC, Wang LZ, Fan L, Guo JY, Lamba J, Schuetz E, Lim R, Lim HL, Ong AB, Lee HS. Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol. 2002;20:3683–90. doi: 10.1200/JCO.2002.01.025. [DOI] [PubMed] [Google Scholar]
- 57.Petros WP, Hopkins PJ, Spruill S, Broadwater G, Vredenburgh JJ, Colvin OM, Peters WP, Jones RB, Hall J, Marks JR. Associations between drug metabolism genotype, chemotherapy pharmacokinetics, and overall survival in patients with breast cancer. J Clin Oncol. 2005;23:6117–25. doi: 10.1200/JCO.2005.06.075. [DOI] [PubMed] [Google Scholar]
- 58.DeMichele A, Aplenc R, Botbyl J, Colligan T, Wray L, Klein-Cabral M, Foulkes A, Gimotty P, Glick J, Weber B, Stadtmauer E, Rebbeck TR. Drug-metabolizing enzyme polymorphisms predict clinical outcome in a node-positive breast cancer cohort. J Clin Oncol. 2005;23:5552–9. doi: 10.1200/JCO.2005.06.208. [DOI] [PubMed] [Google Scholar]
- 59.Rouits E, Boisdron-Celle M, Dumont A, Guerin O, Morel A, Gamelin E. Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity. A molecular and clinical study of 75 patients. Clin Cancer Res. 2004;10:5151–9. doi: 10.1158/1078-0432.CCR-03-0548. [DOI] [PubMed] [Google Scholar]
- 60.Carlini LE, Meropol NJ, Bever J, Andria ML, Hill T, Gold P, Rogatko A, Wang H, Blanchard RL. UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res. 2005;11:1226–36. [PubMed] [Google Scholar]
- 61.Diasio RB, Beavers TL, Carpenter JT. Familial deficiency of dihydropyrimidine dehydrogenase — biochemical basis for familial pyrimidinemia and severe 5-fluorouracil-induced toxicity. J Clin Invest. 1988;81:47–51. doi: 10.1172/JCI113308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Krynetshi EY, Tai HL, Yates CR, Fessing CR, Loennechen T, Schuetz JD, Relling MV, Evans WE. Genetic-polymorphism of thiopurine S-methyltransferase — clinical importance and molecular mechanisms. Pharmacogenetics. 1996;6:279–90. doi: 10.1097/00008571-199608000-00001. [DOI] [PubMed] [Google Scholar]
- 63.Jakobsen A, Nielsen JN, Gyldenkerne N, Lindeberg J. Thymidylate synthase and methylenetetrahydrofolate reductase gene polymorphism in normal tissue as predictors of fluorouracil sensitivity. J Clin Oncol. 2005;23:1365–9. doi: 10.1200/JCO.2005.06.219. [DOI] [PubMed] [Google Scholar]
- 64.Aplenc R, Thompson J, Han P, La M, Zhao HQ, Lange B, Rebbeck T. Methylenetetrahydrofolate reductase polymorphisms and therapy response in pediatric acute lymphoblastic leukemia. Cancer Res. 2005;65:2482–7. doi: 10.1158/0008-5472.CAN-04-2606. [DOI] [PubMed] [Google Scholar]
- 65.Goetz MP, Toft D, Reid J, Ames M, Stensgard B, Safgren S, Adjei AA, Sloan J, Atherton P, Vasile V, Salazaar S, Adjei A, Croghan G, Erlichman C. Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer. J Clin Oncol. 2005;23:1078–87. doi: 10.1200/JCO.2005.09.119. [DOI] [PubMed] [Google Scholar]
- 66.Farker K, Merkel U, Wedding U, Hippius M, Hoffken K, Hoffmann A. C3435T polymorphism of MDR1 gene in colorectal cancer patients and the effect on pharmacokinetics of Irinotecan. Eur J Clin Pharmacol. 2005;61:705. [Google Scholar]
- 67.Plasschaert SLA, Groninger E, Boezen M, Kema I, de Vries EGE, Uges D, Veerinan AJP, Kamps WA, Vellenga E, de Graaf SS, de Bont E. Influence of functional polymorphisms of the MDR1 gene on vincristine pharmacokinetics in childhood acute lymphoblastic leukemia. Clin Pharmacol Therapeut. 2004;76:220–9. doi: 10.1016/j.clpt.2004.05.007. [DOI] [PubMed] [Google Scholar]
- 68.Hegi ME, Diserens A, Gorlia T, Hamou M, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JEC, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
- 69.Luyken C, Loeser S, Blaschke B, Kohler S, Kreuzmann A, Seghrouchni S, Reifenberger G, Sabel M. Prognostic value of MGMT methylation, TP53 mutation, and MDM2, EGFR, or CDK4 amplification in malignant glioma patients treated with adjuvant temozolomide chemotherapy. Neuro-Oncology. 2005;7:328. [Google Scholar]
- 70.Madhusudan S, Middleton MR. The emerging role of DNA repair proteins as predictive, prognostic and therapeutic targets in cancer. Cancer Treat Rev. 2005;31:603–17. doi: 10.1016/j.ctrv.2005.09.006. [DOI] [PubMed] [Google Scholar]
- 71.Maier S, Dahlstroem C, Haefliger C, Plum A, Piepenbrock C. Identifying DNA methylation biomarkers of cancer drug response. Am J Pharmacogenomics. 2005;5:223–32. doi: 10.2165/00129785-200505040-00003. [DOI] [PubMed] [Google Scholar]
- 72.Plumb JA, Strathdee G, Sludden J, Kaye SB, Brown R. Reversal of drug resistance in human tumor xenografts by 2″-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res. 2000;60:6039–44. [PubMed] [Google Scholar]
- 73.Samlowski WE, Leachman SA, Wade M, Cassidy P, Porter-Gill P, Busby L, Wheeler R, Boucher K, Fitzpatrick F, Jones DA, Karpf AR. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol. 2005;23:3897–905. doi: 10.1200/JCO.2005.06.118. [DOI] [PubMed] [Google Scholar]
- 74.Gianni L, Kearns C, Giani A, Capri G, Vigano L, Locatelli A, Bonadonna G, Egorin M. Nonlinear pharmacokinetic and metabolism of paclitaxel and its pharmacokinetic/pharmacodynamic relationships in humans. J Clin Oncol. 1995;13:180–90. doi: 10.1200/JCO.1995.13.1.180. [DOI] [PubMed] [Google Scholar]
- 75.Latz JE, Rusthoven JJ, Karlsson MO, Ghosh A, Johnson RD. Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol. 2006;57:427–35. doi: 10.1007/s00280-005-0035-2. [DOI] [PubMed] [Google Scholar]
- 76.Friberg LE, Henningsson A, Maas H, Nguyen L, Karlsson MO. Model of chemotherapy-induced myelosuppression with parameter consistency across drugs. J Clin Oncol. 2002;20:4713–21. doi: 10.1200/JCO.2002.02.140. [DOI] [PubMed] [Google Scholar]
- 77.Mathijssen RHJ, Marsh S, Karlsson MO, Xie RJ, Baker SD, Verweij J, Sparreboom A, McLeod HL. Irinotecan pathway genotype analysis to predict pharmacokinetics. Clin Cancer Res. 2003;9:3246–53. [PubMed] [Google Scholar]