A new nanomedicine of gemcitabine displays enhanced anticancer activity in sensitive and resistant leukemia types

https://doi.org/10.1016/j.jconrel.2007.08.018Get rights and content

Abstract

Gemcitabine is an anticancer nucleoside analogue active against various solid tumors. However, it possesses important drawbacks like a poor biological half-life and the induction of resistance. With the objective of overcoming the above drawbacks, we designed a new nanomedicine of gemcitabine and studied its anticancer efficacy against leukemia at preclinic. Gemcitabine has been covalently coupled with 1,1′,2-tris-nor-squalenic acid to obtain the new anticancer nanomedicine 4-(N)-Tris-nor-squalenoyl-gemcitabine (SQdFdC NA). The SQdFdC NA exhibited, in comparison to gemcitabine, 3.26- and 3.22-folds higher cytotoxicity respectively, in murine resistant leukemia L1210 10K cells and in human leukemia resistant cell line CEM/ARAC8C. Following intravenous treatment of murine aggressive metastatic leukemia L1210 wt bearing mice, the SQdFdC NA caused significant increase in survival time compared to gemcitabine and also led to long-term survivals, which was not the case after gemcitabine treatment. This was attributed to significantly higher deposition of SQdFdC NA in spleen and liver (P < 0.05), the major metastatic organs. In comparison to gemcitabine, SQdFdC NA displayed greater ability to induce S-phase arrest of the cancer cells followed by increased apoptotic induction. Interestingly, like gemcitabine, SQdFdC NA didn't induce appreciable differences in blood parameters even at doses higher than those used for anticancer evaluation. The preclinical data obtained in vitro and in vivo with SQdFdC NA demonstrate that this nanomedicine represents a new therapeutic system for the effective treatment of leukemia.

Introduction

Squalene (Fig. 1A) is an isoprenoid compound, precursor in the biosynthesis of cholesterol; it is widely distributed in nature. In humans, about 60% of the dietary squalene is absorbed [1]. It has been previously used as an excipient vehicle for various categories of drugs [2], [3] and may enhance the therapeutic efficacy of drugs such as cholesterol-lowering agents (e.g. Pravastatin), when co-administered [4]. Apart from its other properties such as antioxidant for skin and eye, an interesting therapeutic application of squalene is its use as an adjunct in the treatment of variety of cancers, even though the therapeutic improvement in vivo, when combined with anticancer drug was very low [5]. Squalene was also found to be able to inhibit the effect of the tumor-promoting agent 12-O-tetradecanoylphorbol-13-acetate in mouse-skin carcinogenesis [6]. On the other hand, prolonged administration of squalene did not demonstrate appreciable side-effects and toxic signs in animals [7]. Very recently, we have developed the concept of “squalenoylation” involving the chemical linkage of squalene with various nucleosides analogues which surprisingly, self-organized in water as nanoassemblies of 100–300 nm, irrespective of the nucleoside analogue used [8].

In the group of anticancer nucleoside analogues, gemcitabine is a potent compound demonstrated to be effective in the treatment of a variety of solid tumors including colon, lung, pacreatic, breast, bladder and ovarian cancers [9], [10]. However, gemcitabine undergoes rapid deamination into the inactive uracil derivative, hence resulting in a short half-life. Another concern is the observed induction of resistance in preclinical models both in vitro and in vivo which would be important in future clinical situations.

Since gemcitabine is a hydrophilic compound, it requires membrane proteins called nucleoside transporters for entering the cells. The main transporter types include hENT1, hENT2, hCNT1 and hCNT3 [11]. Intracellularly, a part of gemcitabine molecules may undergo deamination into the inactive uracil derivative (dFdU) [12]. In fact, gemcitabine needs to be phosphorylated into its active form, primarily into monophosphate mainly by deoxycytidine kinase and, to a lower extent, by other intracellular kinases [13], [14]. The monophosphate is further phosphorylated into di- and triphosphate forms. The triphosphate form (dFdCTP) of gemcitabine is the only active form which inhibits DNA synthesis. However, any modification in the above sequence of events would lead to resistance. For instance, in the case of leukemia, the cellular resistance to gemcitabine due to nucleoside transporter and dCK deficiency has been well documented [11], [15], [16]. Clearly, the drug resistance is an important clinical concern in the treatment with gemcitabine.

Control over the spread of cancer cells and metastasis is obviously another challenge in chemotherapy, since it accounts for 90% of lethality in cancer patients [17]. In many clinical situations, gemcitabine like other anticancer agents is not efficient in inhibiting metastasis, because it doesn't reach the metastasized tissues in sufficient concentrations. In leukemia, the abnormal cells generally spread to the tissues such as bone marrow, spleen and liver [18].

In this view, we investigated in the present communication, the anticancer activity of the 4-(N)-Tris-nor-squalenoyl-gemcitabine nanoassemblies (SQdFdC NA) in vitro on resistant murine and human leukemia cells as well as in vivo on murine aggressive metastatic leukemia L1210 wt. Although the main indication of gemcitabine in the clinic is solid tumors, the initial preclinical evaluation of gemcitabine was done on L1210 murine leukemia [19]. However, in vivo, high doses of gemcitabine were required probably due to the deamination process and subsequently the shorter half-life, contributing to its poor activity in controlling the extensive leukemic metastasis. It was, therefore, of interest to investigate whether the squalenoyl conjugation improved or not the anticancer activity of gemcitabine on leukemia in vivo. Tissue distribution studies have been carried out using similar treatment schedule. The cell cycle parameters such as S-phase and apoptosis have also been evaluated in vivo in L1210 wt leukemia ascitic bearing mice. Finally, the blood parameters have been evaluated after treating the animals with gemcitabine or SQdFdC NA at doses higher than those used for the treatment of the L1210 wt leukemia bearing mice.

Section snippets

Materials and methods

Gemcitabine hydrochloride was purchased from Sequoia Research Products Ltd (UK). Squalene and Dextrose were purchased from Sigma-Aldrich Chemical Co., France.

Characterization of 4-(N)-Tris-nor-squalenoyl-gemcitabine nanoassemblies (SQdFdC NA)

4-(N)-Tris-nor-squalenoyl-gemcitabine (SQdFdC) (Fig. 1B) was synthesized by coupling the 1,1′,2-tris-nor-squalenic acid covalently to the amino group of the heterocyclic ring of gemcitabine. The synthesized SQdFdC molecules behave as amphiphiles which resulted in the spontaneous formation of SQdFdC nanoassemblies when dispersed in water. Their mean diameter was measured to be 130 nm (± 9 nm) with a unimodal size distribution and a zeta potential of − 20 mV.

In vitro cytotoxicity of gemcitabine and SQdFdC nanoassemblies

The anticancer activity of SQdFdC NA was

Discussion

Gemcitabine is a hydrophilic molecule and its cellular uptake occurs by means of active transport through nucleoside transporters, among which hENT type is one of the most important [22]. Thus, the cells with deficiency in hENT1 transporters represent an interesting model to study resistance to gemcitabine. We found that, in such CEM/ARAC8C cells, the SQdFdC NA still displayed a significant anticancer activity. This suggested that these nanoassemblies of SQdFdC should penetrate into the

Conclusion

The squalenoylation of gemcitabine which leads to the formation of ultrasmall nanoassemblies of ca. 130 nm represents a new approach to address gemcitabine to cancer cells. Though, the SQdFdC NA underwent higher accumulation in liver, it didn't considerably modify the liver enzyme levels, suggesting that it doesn't contribute to hepatotoxicity. The preclinical results described here, both in vitro and in vivo, demonstrate the efficacy of this nanomedicine in the treatment of leukemia (both

Acknowledgements

The financial support of the “Agence Nationale de la Recherche” (ANR, grant SYLIANU) and of the CNRS (grant “Ingénieur de valorisation”) is acknowledged, as is a post-doctoral fellowship to LHR from the Univ. Paris-Sud. Dr. B. Rousseau is acknowledged for providing the radiolabeled SQdFdC.

References (30)

  • H. Kamimura et al.

    Studies on distribution, excretion and subacute toxicity of squalene in dogs

    Fukuoka Igaku Zasshi

    (1989)
  • P. Couvreur et al.

    Squalenoyl nanomedicines as potential therapeutics

    Nano Lett.

    (2006)
  • L.W. Hertel et al.

    Evaluation of the antitumor activity of gemcitabine (2′,2′-difluoro-2′-deoxycytidine)

    Cancer Res.

    (1990)
  • W. Plunkett et al.

    Preclinical characteristics of gemcitabine

    Anticancer Drugs

    (1995)
  • J.R. Mackey et al.

    Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines

    Cancer Res.

    (1998)
  • Cited by (117)

    • Plasma membrane lipid bilayer is druggable: Selective delivery of gemcitabine-squalene nano-medicine to cancer cells

      2023, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      In vivo, the SQGem NPs have been shown to be much more effective than free gemcitabine on solid tumors grafted subcutaneously as well as on murine models of aggressive metastatic leukemia [11]. In cell culture, SQGem NPs exhibited >3-folds higher cytotoxicity both in murine resistant leukemia L1210 10 K and in human leukemia resistant CEM/ARAC8C cells in comparison to the free drug [12]. Cytotoxic effects of nanoparticles composed of cis-platin conjugated to squalene (SQ-CDDP) were assessed in human colonic cells and in mouse models of intestinal cancers [13].

    • Carrier-free nanomedicines for cancer treatment

      2022, Progress in Materials Science
    • Recent advances in prodrug-based nanoparticle therapeutics

      2021, European Journal of Pharmaceutics and Biopharmaceutics
    View all citing articles on Scopus
    View full text