Elsevier

Biochemical Pharmacology

Volume 81, Issue 7, 1 April 2011, Pages 950-956
Biochemical Pharmacology

Effect of its deaminated metabolite, 2′,2′-difluorodeoxyuridine, on the transport and toxicity of gemcitabine in HeLa cells

https://doi.org/10.1016/j.bcp.2011.01.016Get rights and content

Abstract

Gemcitabine is a pyrimidine analog effective against many solid tumors. Following intravenous administration, deaminases in the plasma rapidly convert the parent compound, gemcitabine, to its deaminated metabolite, 2′,2′-difluorodeoxyuridine (dFdU), resulting in an elimination half-life for gemcitabine of 8 min. The half-life of dFdU, however, is upwards of 14 h, yielding plasma concentrations that are frequently 10–20-fold higher than that of gemcitabine. The uptake of gemcitabine into tumor cells is facilitated by both concentrative (hCNT) and equilibrative (hENT) nucleoside transporters. Recently, it was observed that dFdU is a substrate for hCNT as well. The purpose of this study was to investigate the effects of dFdU on gemcitabine uptake and efflux via hENT1 and hENT2 in HeLa cells. Our results suggest that dFdU is a substrate for both hENT1 and hENT2 as well as a competitive inhibitor of gemcitabine transport at concentrations >100-fold lower than those typically achieved in plasma (IC50 = 0.45 and 1.2 μM for hENT1/2 and hENT2, respectively). However, inhibition of gemcitabine uptake is time-dependent, as dFdU limits gemcitabine uptake into HeLa cells by more than 80% during short (<20 s) incubation periods but increases net gemcitabine retention as incubation length increases. While dFdU enhances the accumulation of gemcitabine by up to 1.5-fold following a 60 min incubation, dFdU did not enhance gemcitabine cytotoxicity. In conclusion, this is the first report of an interaction between dFdU and gemcitabine suggesting that the deaminated metabolite may play an important role in the disposition of gemcitabine in tumor cells.

Graphical abstract

Rate of gemcitabine (dFdC) uptake via equilibrative nucleoside transporters (ENT) is decreased in the presence of its metabolite (dFdU), but intracellular retention is ultimately increased.

  1. Download : Download full-size image

Introduction

Gemcitabine (2′,2′-difluorodeoxycytidine, dFdC), is an analog of deoxycytidine with high activity against many types of solid tumors including pancreatic, cervical, ovarian, breast, bladder, and non-small cell lung cancers [1], [2], [3], [4]. As a hydrophilic nucleoside analog, gemcitabine utilizes nucleoside transporters to cross plasma membranes. Once inside the cell, this prodrug is quickly phosphorylated to its active di- and triphosphate moieties (dFdCDP, dFdCTP) [5]. dFdCDP and dFdCTP are then incorporated into nascent DNA and RNA strands, eventually leading to inhibition of DNA polymerases, chain termination and cessation of DNA replication [6].

In addition to phosphorylation, gemcitabine may also undergo intra- and extracellular deamination to the much less active form, 2′,2′-difluorodeoxyuridine (dFdU), via cytidine deaminase (CDA), which is present at high concentrations in many types of normal and malignant tissues as well as in the plasma [7], [8]. In fact, after intravenous administration, cytidine deaminases in the plasma rapidly convert gemcitabine to dFdU, resulting in a gemcitabine plasma elimination half-life of only 8 min [9]. The long half-life of dFdU (14 h), leads to plasma concentrations of the dFdU that are frequently 10–20-fold higher than those of the parent compound when measured shortly after gemcitabine administration [10], [11], [12]. Additionally, deamination of the monophosphate form of gemcitabine may also occur intracellularly via deoxycytidine monophosphate deaminase (dCMPD) to yield dFdUMP, which may then be further phosphorylated to dFdUDP and dFdUTP or dephosphorylated to dFdU [5]. As with gemcitabine, dFdU may also be directly phosphorylated intracellularly to its mono-, di-, and triphosphate metabolites by deoxycytidine kinase. However, the majority of dFdU nucleotides are thought to be formed by the breakdown of gemcitabine nucleotides as dFdU is predicted to have a low affinity for deoxycytidine kinase (dCK) [13], [14], [15].

Gemcitabine is transported into cells by both the concentrative (hCNT) and equilibrative (hENT) nucleoside transporters. Human concentrative nucleoside transporter 1 (hCNT1) is the most efficient transporter of gemcitabine, with a Km of around 18 μM, but the distribution of this protein throughout the body is limited when compared to the more ubiquitously expressed ENT family [16], [17]. hENT1 and hENT2 also facilitate gemcitabine uptake into cells, and clinical studies have demonstrated a correlation between the expression of these transporters and response to gemcitabine therapy [18], [19], [20].

Recently, it has been reported that dFdU is transported by both hCNT1 and hENT1 into hCNT1-transfected MDCK cells [21]. The extent of dFdU uptake was nearly the same as that of gemcitabine, suggesting that dFdU is a high affinity substrate for these transporters as well. Additionally, it was determined in the same study that dFdU undergoes biphasic efflux from HepG2 and A549 cells preloaded with gemcitabine, presumably via the same nucleoside transporters.

When administered alone, dFdU displays minimal cytotoxicity when compared with gemcitabine. Yet, with both the parent and metabolite competing for the same transporters, high plasma concentrations of dFdU may affect the efficacy of gemcitabine against solid tumors by decreasing its cellular uptake. Therefore, the objective of this study was to investigate the effects of dFdU on gemcitabine uptake and efflux via hENT1 and hENT2 in HeLa cells, a cell line which endogenously expresses these two nucleoside transporters. Based on our results, we have developed a mechanistic model whereby dFdU limits the uptake of gemcitabine into the cell, yet ultimately results in an increased intracellular sequestration of gemcitabine within cells.

Section snippets

Chemicals

2′,2′-Difluorodeoxycytidine (dFdC; gemcitabine) and 2′,2′-difluorodeoxyuridine (dFdU) were synthesized by the Institute for Therapeutics, Discovery, and Development at the University of Minnesota, Minneapolis, MN. [5-3H]-gemcitabine (11 Ci/mmol) was obtained from Moravek chemicals (La Brea, CA). Uridine, dilazep, and nitrobenzyl-mercaptopurine riboside (NBMPR) were obtained from Sigma–Aldrich (Saint Louis, MO). 13C,15N2-dFdU was purchased from Toronto Research Chemicals (North York, Ontario,

Uptake of gemcitabine over short time periods

Due to rapid equilibration, experiments designed to examine nucleoside uptake via equilibrative transporters were performed over a very short timeframe to minimize intracellular metabolism and limit efflux of substrate and metabolites [25]. Therefore, initial experiments to characterize the effects of dFdU on gemcitabine uptake via hENTs in HeLa cells were conducted with incubation periods of <1 min, under the assumption that all detectable intracellular radiolabel was in the form of the parent

Discussion

Following intravenous administration of gemcitabine, dFdU reaches peak plasma levels within 5–15 min post-infusion. Unlike gemcitabine, concentrations of dFdU remain elevated for a prolonged period of time with an elimination half-life of up to 24 h [9]. Yet, despite the difference in their respective pharmacokinetic profiles, the contribution of dFdU to gemcitabine disposition is unknown. A nucleoside analog itself, dFdU has been observed to undergo influx via both hCNT1 and hENT1 in

References (35)

  • V. Heinermann

    Gemcitabine in metastatic breast cancer

    Expert Rev Anticancer Ther

    (2005)
  • P. Huang et al.

    Action of 2′,2′-difluorodeoxycytidine on DNA synthesis

    Cancer Res

    (1991)
  • D.H.W. Ho

    Distribution of kinase and deaminase of 1-beta-d-arabinofuranosylcytosine in tissues of man and mouse

    Cancer Res

    (1973)
  • J.L. Abbruzzese et al.

    A phase I clinical, plasma, and cellular pharmacology study of gemcitabine

    J Clin Oncol

    (2000)
  • E. Marangon et al.

    Simultaneous determination of gemcitabine and its main metabolite, dFdU, in plasma of patients with advanced non-small-cell lung cancer by high-performance liquid chromatography–tandem mass spectrometry

    J Mass Spectrom

    (2008)
  • X. Jiang et al.

    Population pharmacokinetics of gemcitabine and its metabolite in patients with cancer: effect of oxaliplatin and infusion rate

    Br J Clin Pharmacol

    (2007)
  • V. Heinemann et al.

    Cellular elimination of 2′,2′-difluorodeoxycytidine 5′-triphosphate: a mechanism of self potentiation

    Cancer Res

    (1992)
  • Cited by (20)

    • Comparative Proteomic Analysis Identifies Key Metabolic Regulators of Gemcitabine Resistance in Pancreatic Cancer

      2022, Molecular and Cellular Proteomics
      Citation Excerpt :

      DCTD, which deaminates dFdCMP to dFdUMP, was also upregulated. The resulting dFdUMP could be converted into dFdU and exported rapidly from cells by CNT (52, 53), reducing the pool of intracellular dFdU that can be metabolized to dFdUDP and dFdUTP, both of which can mediate cytotoxicity via incorporation into DNA and effects on dNTP pools (54, 55). The overall pattern of differential protein expression in MIA-GR8 cells suggest that multiple mechanisms operant in MIA-GR8 cells diminish dFdCMP, which would deplete the active Gem precursor pool.

    • Polymeric modification of gemcitabine via cyclic acetal linkage for enhanced anticancer potency with negligible side effects

      2020, Biomaterials
      Citation Excerpt :

      However, GEM is a water-soluble and low-molecular-weight drug, whereby it encounters rapid elimination from bloodstream leading to inefficient tumor accumulation as well as dispersion into normal tissues. Indeed, GEM is internalized by cells via nucleoside transporters (NTs) [9,43,44], and thus it accumulates in NT-enriched organs (e.g., gastrointestines) [45,46], and causes adverse effects in such organs. Moreover, myelosuppression caused by GEM can be a critical reason to stop the administration to the patients [10,11].

    • Half generations magnetic PAMAM dendrimers as an effective system for targeted gemcitabine delivery

      2016, International Journal of Pharmaceutics
      Citation Excerpt :

      Gemcitabine with a small molecular weight is a highly hydrophilic drug, with a short plasma half-life. Therefore, it is often administered at toxic high doses in clinical applications (Hodge et al., 2011). Furthermore, due to its hydrophilic nature, it cannot traverse cell membranes by passive diffusion and, therefore enters via nucleoside transporters that may lead to drug resistance.

    • A patient-derived subrenal capsule xenograft model can predict response to adjuvant therapy for cancers in the head of the pancreas

      2015, Pancreatology
      Citation Excerpt :

      One potential biomarker is expression of hENT1 [34], an equilibrative transporter of nucleosides into cancer cells. High expression is considered important for gemcitabine effectiveness, because its uptake into tumour cells is facilitated by both concentrative (hCNT) and equilibrative (hENT) nucleoside transporters [35]. However, the intracellular distribution of gemcitabine depends on both transport and metabolic processes, and the rate at which gemcitabine enters depends also on the presence of the deaminated metabolite which competes with gemcitabine and may be altering its metabolic fate [36].

    View all citing articles on Scopus
    1

    Current address: Dept. of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, USA.

    View full text