Elsevier

Biochemical Pharmacology

Volume 81, Issue 1, 1 January 2011, Pages 24-31
Biochemical Pharmacology

Organ-specific carboxylesterase profiling identifies the small intestine and kidney as major contributors of activation of the anticancer prodrug CPT-11

https://doi.org/10.1016/j.bcp.2010.09.001Get rights and content

Abstract

The activation of the anticancer prodrug CPT-11, to its active metabolite SN-38, is primarily mediated by carboxylesterases (CE). In humans, three CEs have been identified, of which human liver CE (hCE1; CES1) and human intestinal CE (hiCE; CES2) demonstrate significant ability to hydrolyze the drug. However, while the kinetic parameters of CPT-11 hydrolysis have been measured, the actual contribution of each enzyme to activate the drug in biological samples has not been addressed. Hence, we have used a combination of specific CE inhibition and conventional chromatographic techniques to determine the amounts, and hydrolytic activity, of CEs present within human liver, kidney, intestinal and lung specimens. These studies confirm that hiCE demonstrates the most efficient kinetic parameters for CPT-11 activation, however, due to the high levels of hCE1 that are expressed in liver, the latter enzyme can contribute up to 50% of the total of drug hydrolysis in this tissue. Conversely, in human duodenum, jejunum, ileum and kidney, where hCE1 expression is very low, greater than 99% of the conversion of CPT-11 to SN-38 was mediated by hiCE. Furthermore, analysis of lung microsomal extracts indicated that CPT-11 activation was more proficient in samples obtained from smokers. Overall, our studies demonstrate that hCE1 plays a significant role in CPT-11 hydrolysis even though it is up to 100-fold less efficient at drug activation than hiCE, and that drug activation in the intestine and kidney are likely major contributors to SN-38 production in vivo.

Introduction

The anticancer drug CPT-11 (irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin), is a prodrug that is activated by esterases to yield SN-38 (7-ethyl-10-hydroxycamptothecin), a potent topoisomerase I poison [1]. The majority of biochemical studies have demonstrated that this is achieved by carboxylesterases (CE) [2], [3], [4], [5], [6], however butyrylcholinesterases (BChE) can also effect this process, albeit with poor efficiency [7], [8], [9], [10]. In humans, three CEs have so far been identified. The human liver CE, hCE1 (CES1), is predominantly expressed in the liver and demonstrates a preference for small, non-bulky substrates [11], [12], [13]. The human intestinal CE, hiCE (CES2), is expressed in the gut and the liver, and can hydrolyze much larger, more complex molecules. This is likely due to flexible domains present within the active site of the enzyme that allows for accommodation of these esters [14], [15]. The human brain CE, hBr3 (CES3), is believed to be expressed in the epithelia that form part of the blood brain barrier [16], although this protein has not been exhaustively tested for its substrate specificity [17]. However, all of these enzymes have been compared for their ability to activate CPT-11 [4], [5], [15], [17].

Results from these studies indicate that the hiCE is 64- to 100-fold more efficient than hCE1 at CPT-11 hydrolysis, with hBr3 being 20-fold poorer than the latter enzyme. Hence, due to the poor kinetic parameters for hBr3 with the drug (∼2000-fold less efficient than hiCE), and its very limited pattern of expression, it is unlikely that this CE significantly contributes to drug activation in vivo. Based upon this biochemical and enzyme kinetic evidence, we and others have assumed that hiCE would be the major esterase responsible for CPT-11 hydrolysis in cancer patients [4], [5], [17]. We hypothesized therefore that using selective hiCE inhibitors [18], [19], it would be possible to determine the amount of this enzyme present in biological samples using a simple substrate such as o-NPA. Simply, the difference in the enzyme activity assays in the presence and absence of the inhibitor should represent the amount of hiCE in the preparation. This could then be used as a measure of the ability of the sample to hydrolyze CPT-11. Such an approach would obviate the need for expensive and time consuming assays (HPLC with fluorescence detection) to monitor drug hydrolysis.

The studies described here sought to validate this approach by examining the ability of selective hiCE inhibitors [18], [19] to prevent the conversion of CPT-11 to SN-38 in a series of human microsomal samples. However, we were unable to dramatically reduce drug activation in these specimens using these specific inhibitors, suggesting that other proteins within the extracts could mediate the hydrolysis of CPT-11. Therefore, we have used a combination of chromatography and biochemical assays using CE inhibitors (both specific and non-specific), to determine the contribution of other enzymes towards CPT-11 activation. These studies demonstrate that hCE1, while demonstrating poor kinetic parameters for this substrate, can significantly contribute to drug hydrolysis. Furthermore, our studies identify the kidney as a source of CPT-11 activation and demonstrate that drug hydrolysis is more proficient in lung tissue isolated from smokers.

Section snippets

Chemicals, CPT-11 and chromatographic media

Reagents for biochemical assays and HPLC were provided by Sigma–Aldrich (St. Louis, MO). CPT-11 was provided as a kind gift by Dr. J.P. McGovren. Sephacryl S-200 high resolution resin was purchased from Amersham Biosciences Inc. (Piscataway, NJ).

Enzymes, antibodies and inhibitors

Pure recombinant hiCE and hCE1, used as markers for western analyses and controls for biochemical assays, were prepared as previously described [20]. Anti-rabbit hiCE- and hCE1-specific antibodies were generated by repeated injection of animals with

Use of selective CE inhibitors to monitor enzyme levels in microsomal samples

Since human tissues can contain up to three different CEs (hiCE, hCE1 and hBr3), of which the former is the most efficient at activating CPT-11, we hypothesized that determining substrate hydrolysis in the presence or absence of selective hiCE inhibitors, might allow us to generate accurate estimates of the amounts of this protein in biological samples. The advantage of such an approach would be that the assay could be performed using a substrate other than CPT-11 (e.g. o-NPA), thereby making

Discussion

We and others have previously reported that hiCE is the most efficient carboxylesterase at activating CPT-11 in humans [4], [5], [17], [20]. This has been based entirely upon in vitro studies using either CEs purified from human liver specimens, or recombinant protein expressed in mammalian or insect cells. Based upon the poor kinetic parameters demonstrated by hCE1 (∼25-fold higher Km and 90-fold lower kcat/Km than hiCE [4], [5], [17]), it has been assumed that this enzyme is unlikely to

Acknowledgements

We thank Dr. J.P. McGovren (Pfizer) for the gift of CPT-11. This work was supported in part by NIH Grants CA108775, an NIH Cancer Center Core Grant CA21765, and by the American Lebanese Syrian Associated Charities and St. Jude Children's Research Hospital (SJCRH).

References (27)

  • A.A. Guemei et al.

    Human plasma carboxylesterase and butyrylcholinesterase enzyme activity: correlations with SN-38 pharmacokinetics during a prolonged infusion of irinotecan

    Cancer Chemother Pharmacol

    (2001)
  • C.L. Morton et al.

    CPT-11 is a potent inhibitor of acetylcholinesterase but is rapidly catalyzed to SN-38 by butyrylcholinesterase

    Cancer Res

    (1999)
  • H.M. Dodds et al.

    The mechanism for the inhibition of acetylcholinesterases by irinotecan (CPT-11)

    Mol Pharmacol

    (1999)
  • Cited by (83)

    • Involvement of esterases in the pulmonary metabolism of beclomethasone dipropionate and the potential influence of cannabis use

      2022, Chemico-Biological Interactions
      Citation Excerpt :

      The results were confirmed by the selective inhibition study, where known CES1 inhibitors (PMSF, benzil, and betulinic acid) failed to prevent BDP from hydrolysis by HLuS9 (Fig. 4). CES1 was initially hypothesized as a main contributor in this study because esterases in the lung were believed to be responsible for BDP activation [9–11] and several published studies showed expression of CES1 in the lung tissue [12,14,42]. Under our assay conditions, CES1 exhibited efficient metabolic activity toward BDP with an estimated intrinsic clearance of 0.726 ml/min/mg S9 protein.

    • Novel potent bifunctional carboxylesterase inhibitors based on a polyfluoroalkyl-2-imino-1,3-dione scaffold

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Compounds with a 1,2-dione scaffold (Fig. 1) have been identified as the most important structures for CES inhibition. These compounds have Ki values in the nanomolar range and exert potent and selective inhibitory effects toward hCES1 or hCES2 depending on the orientation of the dione oxygens [35,39,44,45]. While the 1,2-dione moiety is very beneficial for CES inhibition [39,40], such compounds do not exhibit untoward inhibitory effects on human acetylcholinesterase (AChE, EC 3.1.1.7) or butyrylcholinesterase (BChE, EC 3.1.1.8) [2,33].

    • Irinotecan-mediated diarrhea is mainly correlated with intestinal exposure to SN-38: Critical role of gut Ugt

      2020, Toxicology and Applied Pharmacology
      Citation Excerpt :

      The disposition of irinotecan is complex involving several enzymes and transporters. It is well-known that irinotecan is converted into SN-38 catalyzed by carboxylesterases and other esterases (Hatfield et al., 2011; Bahar et al., 2012) and SN-38 can be conjugated into SN-38 glucuronide (SN-38G) catalyzed by uridine diphosphate glucuronosyltransferases (UGTs/Ugts) (Iyer et al., 1998). For the metabolism of irinotecan, human carboxylesterases 2 is more efficient in than other carboxylesterases (Humerickhouse et al., 2000).

    View all citing articles on Scopus
    View full text