Elsevier

The Lancet Oncology

Volume 6, Issue 10, October 2005, Pages 780-789
The Lancet Oncology

Review
Modulation of cytochrome P450 activity: implications for cancer therapy

https://doi.org/10.1016/S1470-2045(05)70388-0Get rights and content

Summary

Although metabolism mediated by cytochrome P450 isoenzymes is known to play a major part in the biotransformation of anticancer agents in vivo, few clinical studies have investigated activity of cytochrome P450s and therapeutic outcome in people with cancer. Variability between individuals in the pharmacokinetics of cancer chemotherapy has important consequences in terms of therapeutic efficacy and safety. We discuss here the effect of drug metabolism mediated by cytochrome P450 on therapeutic outcome. As examples, the biotransformation pathways of cyclophosphamide, ifosfamide, tamoxifen, docetaxel, paclitaxel, and irinotecan are discussed. Since most anticancer agents are transformed by enzymes, better knowledge of their metabolic pathways could help improve treatment outcome and safety. Furthermore, a more complete understanding of the metabolism of anticancer agents through phenotyping and genotyping approaches will facilitate the prediction of interactions between drugs. More clinical evidence is needed on the metabolic transformation and drug interactions with these agents to improve cancer therapeutics.

Introduction

Anticancer agents have a wide variation between individuals in response, owing partly to pharmacokinetic variability. The most common strategy used to restrict pharmacokinetic variability is to relate doses of drugs to the patient's body surface area (BSA). This approach is based on the assumption that each individual is capable of metabolising drugs with the same efficacy. However, BSA-based dosing strategies do not reduce variability between individuals in the pharmacokinetics of most chemotherapeutic agents.1 Therefore, other factors must contribute to drug disposition as well as therapeutic efficacy and toxic effects.

One of the sources of variation in drug effects is differences between individuals in inherited elements of drug response.2 Genetics is estimated to account for 20–95% of variability in therapeutic response and toxic effects. Research in pharmacogenomics aims to elucidate the genomic determinants of drug disposition and effect.3 Genetics affects various factors influencing drug disposition, including drug absorption, metabolism, distribution, and excretion.3 Several reviews on pharmacogenomics and cancer therapy have been published, but have focused mostly on genetic variability in phase II reaction enzymes such as UDP glucuronosyltransferase, N-acetyltransferases, and thiopurine methyltransferase.2 Here, we focus on drug metabolism, specifically the role of metabolism mediated by cytochrome P450s, on cancer therapy and the endogenous and exogenous factors that influence the activity of these enzymes.

The cytochrome P450 enzymes catalyse the oxidation of many structurally unrelated compounds of endogenous and exogenous origin. They also have important roles in the extent and duration of drug effects, by catabolising drugs to inactive metabolites or by bioactivating prodrugs to their active forms. Several anticancer agents are metabolised by cytochrome P450s (table).

Section snippets

Bioactivation and resistance

The most important site of metabolism mediated by cytochrome P450 is the liver, where the enzymes are ubiquitously expressed (figure 1). For CYP3A-mediated metabolism of drugs administered orally, the gastrointestinal tract is also potentially important. Enzymatic biotransformation within various other organs, such as the brain, lung, and kidneys, has been documented in experimental systems, but evidence of clinically relevant metabolism at these sites is lacking. There is also evidence that

Factors affecting activity of cytochrome P450s

Different factors can contribute to the variable activity of cytochrome P450s. The expression of these enzymes can be affected by endogenous factors such as an individual's age, sex, concomitant disease (such as cancer, hepatic dysfunction, or renal impairment), and genetic polymorphisms.8, 9, 10, 11

Anticancer agents metabolised by cytochrome P450

Experiments with one or more of the various liver preparations available or with recombinant cytochrome P450 isoforms can identify the specific enzymes involved in the metabolism of each of the anticancer agents that are substrates for the enzyme system (table). In general, knowledge of which isoforms have roles in the transformation of an anticancer agent facilitates the prediction of interactions with other drugs. However, these in-vitro systems have limitations.32

The clinical importance of

Conclusions

A most important difficulty in cancer pharmacology is the prediction of outcome of therapy, in terms of both tumour response and toxic effects. In general, identification of specific factors that might affect cytochrome P450 activity, the pharmacokinetics, and the pharmacodynamic profile of drugs in healthy individuals is difficult. Measurement of cytochrome P450 activity in individual patients with cancer is even more complex, given their heterogeneous backgrounds and the complex physiological

Search strategy and selection criteria

Data for this review were identified by searches in PubMed and references cited in relevant articles. Search terms included: “pharmacogenetics”, “pharmacogenomics”, “drug metabolism”, “oncology”, “chemotherapy”, and “cytochrome P450”. Only papers published in English were included.

References (77)

  • M Ciotti et al.

    Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP glucuronosyltransferases encoded at the UGT1 locus

    Biochem Biophys Res Commun

    (1999)
  • SD Baker et al.

    Role of body surface area in dosing of investigational anticancer agents in adults, 1991–2001

    J Natl Cancer Inst

    (2002)
  • WE Evans et al.

    Pharmacogenomics: translating functional genomics into rational therapeutics

    Science

    (1999)
  • WE Evans et al.

    Pharmacogenomics: drug disposition, drug targets, and side effects

    N Engl J Med

    (2003)
  • LH Patterson et al.

    Tumour cytochrome P450 and drug activation

    Curr Pharm Des

    (2002)
  • MC McFadyen et al.

    Cytochrome P450 CYP1B1 protein expression: a novel mechanism of anticancer drug resistance

    Biochem Pharmacol

    (2001)
  • WA Denny

    Tumor-activated prodrugs: a new approach to cancer therapy

    Cancer Invest

    (2004)
  • GU Dachs et al.

    From bench to bedside for gene-directed enzyme prodrug therapy of cancer

    Anticancer Drugs

    (2005)
  • MM Cotreau et al.

    The influence of age and sex on the clearance of cytochrome P450 3A substrates

    Clin Pharmacokinet

    (2005)
  • V Pichette et al.

    Drug metabolism in chronic renal failure

    Curr Drug Metab

    (2003)
  • RH Elbekai et al.

    The effect of liver cirrhosis on the regulation and expression of drug metabolizing enzymes

    Curr Drug Metab

    (2004)
  • MT Kinirons et al.

    Drug metabolism and ageing

    Br J Clin Pharmacol

    (2004)
  • Food and Drug Administration. FDA guidance for industry: in vivo bioequivalence studies based on population and individual bioequivalence approaches

    (1997)
  • C Durnas et al.

    Hepatic drug metabolism and aging

    Clin Pharmacokinet

    (1990)
  • EA Sotaniemi et al.

    Age and cytochrome P450-linked drug metabolism in humans: an analysis of 226 subjects with equal histopathologic conditions

    Clin Pharmacol Ther

    (1997)
  • Z Bebia et al.

    Bioequivalence revisited: influence of age and sex on CYP enzymes

    Clin Pharmacol Ther

    (2004)
  • KT Kivisto et al.

    The role of human cytochrome P450 enzymes in the metabolism of anticancer agents: implications for drug interactions

    Br J Clin Pharmacol

    (1995)
  • LP Rivory et al.

    Hepatic cytochrome P450 3A drug metabolism is reduced in cancer patients who have an acute-phase response

    Br J Cancer

    (2002)
  • TC Dowling et al.

    Characterization of hepatic cytochrome p4503A activity in patients with end-stage renal disease

    Clin Pharmacol Ther

    (2003)
  • SD Baker et al.

    Factors affecting cytochrome P-450 3A activity in cancer patients

    Clin Cancer Res

    (2004)
  • CM Ulrich et al.

    Cancer pharmacogenetics: polymorphisms, pathways and beyond

    Nat Rev Cancer

    (2003)
  • I Johansson et al.

    Inherited amplification of an active gene in the cytochrome P450 CYP2D locus as a cause of ultrarapid metabolism of debrisoquine

    Proc Natl Acad Sci USA

    (1993)
  • KA Phillips et al.

    Potential role of pharmacogenomics in reducing adverse drug reactions: a systematic review

    JAMA

    (2001)
  • H Swaisland et al.

    The effect of the induction and inhibition of CYP3A4 on the pharmacokinetics of single oral doses of ZD1839 (‘Iressa’), a selective epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), in healthy male volunteers

    Proc Am Soc Clin Oncol

    (2002)
  • VJ Wacher et al.

    Overlapping substrate specificities and tissue distribution of cytochrome P450 3A and P-glycoprotein: implications for drug delivery and activity in cancer chemotherapy

    Mol Carcinog

    (1995)
  • JH Lin et al.

    Role of P-glycoprotein in pharmacokinetics: clinical implications

    Clin Pharmacokinet

    (2003)
  • K Venkatakrishnan et al.

    Human drug metabolism and the cytochromes P450: application and relevance of in vitro models

    J Clin Pharmacol

    (2001)
  • AV Boddy et al.

    Metabolism and pharmacokinetics of oxazaphosphorines

    Clin Pharmacokinet

    (2000)
  • Cited by (117)

    • New insights on the marine cytochrome P450 enzymes and their biotechnological importance

      2020, International Journal of Biological Macromolecules
      Citation Excerpt :

      This results in conversion of the prodrug to the corresponding cytotoxin locally in the tumor tissue. Some of these gene-directed enzyme prodrug therapy systems are currently in phase III studies [44]. Oxycodone has replaced morphine as the first opioid of choice, and it is provided as oral immediate-release, oral extended-release and intravenous formulations [45].

    View all citing articles on Scopus
    View full text