Elsevier

European Journal of Pharmacology

Volume 741, 15 October 2014, Pages 8-16
European Journal of Pharmacology

Review
Gemcitabine: Metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer

https://doi.org/10.1016/j.ejphar.2014.07.041Get rights and content

Abstract

Gemcitabine is the first-line treatment for pancreatic adenocarcinoma, but is increasingly used to treat breast, bladder, and non-small cell lung cancers. Despite such broad use, intrinsic and acquired chemoresistance is common. In general, the underlying mechanisms of chemoresistance are poorly understood. Here, current knowledge of gemcitabine metabolism, mechanisms of action, sensitivity and chemoresistance reported over the past two decades are reviewed; and we also offer new perspectives to improve gemcitabine efficacy with particular reference to the treatment of pancreatic cancer.

Introduction

Gemcitabine is a nucleoside analog that has been used as a chemotherapeutic agent for more than 15 years. During this time gemcitabine has become the standard treatment choice for locally advanced and metastatic pancreatic cancer, for which there have been few treatment advances. Pancreatic cancer is one of the most devastating human cancers, with a median survival rate of less than 5 months after diagnosis and with under a 5% five-year survival rate even if brought under remission (Bilimoria et al., 2007). This poor prognosis is partly explained because of poor penetration of drugs into the dense and under vascularized tumour stroma (Neesse et al., 2011), a high degree of acquired chemoresistance by tumour cells and also because surgical intervention is not possible in almost 80% of patients (Vincent et al., 2011).

Gemcitabine is also used to treat a variety of solid tumors including breast, ovarian, and non-small cell lung cancer, especially when in combination with the platinum-based drugs cisplatin and carboplatin (Nagourney et al., 2008, Pfisterer et al., 2006, Reck et al., 2009). Despite its relatively broad and common use, mechanisms related to gemcitabine resistance are not well understood. A number of different cellular pathways, transcriptional factors and nucleotide metabolism enzymes have been linked to gemcitabine resistance and sensitivity.

Here the known molecular mechanisms commonly associated with gemcitabine pharmacokinetics and pharmacodynamics are reviewed with a special focus on pancreatic cancer. A better understanding of gemcitabine pharmacology is essential for developing improved treatments and ultimately for increasing patient survival in pancreatic cancer.

Section snippets

Uptake and metabolism

Gemcitabine (2′,2′-difluoro-2′-deoxycytidine; dFdC) is a deoxycytidine analog with multiple modes of action inside the cell. The basics of gemcitabine metabolism are illustrated in Fig. 1. As a prodrug, dFdC must be metabolized to the active triphosphate form of gemcitabine (2′,2′-difluoro-2′-deoxycytidine triphosphate; dFdCTP). Cellular uptake of gemcitabine is mediated by a family of integral membrane proteins termed human nucleoside transporters (hNTs), which overcome the inherent barrier to

Mechanisms of action

The most important mechanism of action of gemcitabine is inhibition of DNA synthesis (Huang et al., 1991). When dFdCTP is incorporated into DNA, a single deoxynucleotide is incorporated afterwards, preventing chain elongation (Gandhi et al., 1996). This non-terminal position of gemcitabine makes DNA polymerases unable to proceed, in a process known as ‘masked chain-termination′, which also inhibits removal of gemcitabine by DNA repair enzymes (Huang et al., 1991). This process is shown in Fig. 2

Gemcitabine chemoresistance and sensitivity

Like many other drugs used in cancer chemotherapy, resistance to gemcitabine may be intrinsic, or acquired by individual patient during treatment cycles. Several mechanisms have been reported in the literature and some of these seem well established (Table 1). However, as many of these processes are complex, it can be difficult to understand precise roles and, indeed, consequences in chemoresistance. This is particularly relevant for signalling pathways involved in apoptosis, growth,

Perspectives

Gemcitabine is still the standard chemotherapeutic agent of choice to treat pancreatic cancer; with the best patient clinical outcome, and this is unlikely to change in the coming years. However, few improvements have been made in gemcitabine-based chemotherapies for pancreatic and other cancers (e.g. breast, ovarian and non-small cell lung cancer), although combination therapies are likely to emerge as realistic treatment choices, especially for pancreatic cancer. A recent phase III study of

Conclusion

Many important molecular targets and pathways related to gemcitabine chemoresistance and sensitivity are now established; however, their precise mechanisms of action are not completely understood in most cases. Different pathways and molecular interactions have been described in recent years and new roles for some relatively well-known chemoresistance markers (e.g. NF-κB) have been discovered. Gemcitabine still has clinical potential to treat a broad spectrum of human cancers, but the

Acknowledgments

Financial support was provided by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP – Grants 2009/01303-1 and 2011/04938-8) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES). We thank Dr PF Long from King׳s College London and Faculdade de Ciências Farmacêuticas, Universidade de São Paulo for his critical comments.

References (105)

  • R. Kong et al.

    Downregulation of nuclear factor-kappaB p65 subunit by small interfering RNA synergizes with gemcitabine to inhibit the growth of pancreatic cancer

    Cancer Lett.

    (2010)
  • J.L. Kummer et al.

    Apoptosis induced by withdrawal of trophic factors is mediated by p38 mitogen-activated protein kinase

    J. Biol. Chem.

    (1997)
  • C. Maasch et al.

    Polyetheylenimine-polyplexes of Spiegelmer NOX-A50 directed against intracellular high mobility group protein A1 (HMGA1) reduce tumor growth in vivo

    J. Biol. Chem.

    (2010)
  • C. Mazzon et al.

    Cytosolic and mitochondrial deoxyribonucleotidases: activity with substrate analogs, inhibitors and implications for therapy

    Biochem. Pharmacol.

    (2003)
  • J.C. Morote-Garcia et al.

    Hypoxia-inducible factor-dependent repression of equilibrative nucleoside transporter 2 attenuates mucosal inflammation during intestinal hypoxia

    Gastroenterology

    (2009)
  • R.A. Nagourney et al.

    Carboplatin plus gemcitabine repeating doublet therapy in recurrent breast cancer

    Clin. Breast Cancer

    (2008)
  • M. Nakashima et al.

    Phosphorylation status of heat shock protein 27 plays a key role in gemcitabine-induced apoptosis of pancreatic cancer cells

    Cancer Lett.

    (2011)
  • A. Portal et al.

    Sustained response with gemcitabine plus nab-paclitaxel after folfirinox failure in metastatic pancreatic cancer: report of an effective new strategy

    Clin. Res. Hepatol. Gastroenterol.

    (2014)
  • V.W. Ruiz van Haperen et al.

    Regulation of phosphorylation of deoxycytidine and 2′,2′-difluorodeoxycytidine (gemcitabine); effects of cytidine 5′-triphosphate and uridine 5′-triphosphate in relation to chemosensitivity for 2′,2′-difluorodeoxycytidine

    Biochem. Pharmacol.

    (1996)
  • J.C. Sarup et al.

    Regulation of purine deoxynucleoside phosphorylation by deoxycytidine kinase from human leukemic blast cells

    Biochem. Pharmacol.

    (1989)
  • G.L. Semenza

    Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy

    Trends Pharmacol. Sci.

    (2012)
  • A.P. Singh et al.

    CXCL12/CXCR4 protein signaling axis induces sonic hedgehog expression in pancreatic cancer cells via extracellular regulated kinase- and Akt kinase-mediated activation of nuclear factor κB: implications for bidirectional tumor-stromal interactions

    J. Biol. Chem.

    (2012)
  • A. Vincent et al.

    Pancreatic cancer

    Lancet

    (2011)
  • M. Watanabe et al.

    HMGA-targeted phosphorothioate DNA aptamers increase sensitivity to gemcitabine chemotherapy in human pancreatic cancer cell lines

    Cancer Lett.

    (2012)
  • P. Aksoy et al.

    Cytosolic 5’-nucleotidase III (NT5C3): gene sequence variation and functional genomics

    Pharmacogenet. Genomics

    (2009)
  • A. Arlt et al.

    Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death

    Oncogene

    (2003)
  • M. Bahra et al.

    Combination of hedgehog signaling blockage and chemotherapy leads to tumor reduction in pancreatic adenocarcinomas

    Pancreas

    (2012)
  • J.M. Bailey et al.

    Sonic hedgehog promotes desmoplasia in pancreatic cancer

    Clin. Cancer Res.

    (2008)
  • A.K. Berger et al.

    Successful treatment with nab-paclitaxel and gemcitabine after FOLFIRINOX failure in a patient with metastasized pancreatic adenocarcinoma

    Onkologie

    (2013)
  • A.M. Bergman et al.

    In vivo induction of resistance to gemcitabine results in increased expression of ribonucleotide reductase subunit M1 as the major determinant

    Cancer Res.

    (2005)
  • K.Y. Bilimoria et al.

    Validation of the 6th edition AJCC pancreatic cancer staging system: report from the national cancer database

    Cancer

    (2007)
  • C.L. Costantino et al.

    The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase

    Cancer Res.

    (2009)
  • T. Conroy et al.

    FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer

    N. Engl. J. Med.

    (2011)
  • V.L. Damaraju et al.

    Nucleoside anticancer drugs: the role of nucleoside transporters in resistance to cancer chemotherapy

    Oncogene

    (2003)
  • J.D. Davidson et al.

    An increase in the expression of ribonucleotide reductase large subunit 1 is associated with gemcitabine resistance in non-small cell lung cancer cell lines

    Cancer Res.

    (2004)
  • M.S. Duxbury et al.

    RNA interference targeting the M2 subunit of ribonucleotide reductase enhances pancreatic adenocarcinoma chemosensitivity to gemcitabine

    Oncogene

    (2004)
  • H. Eda et al.

    The antiproliferative activity of DMDC is modulated by inhibition of cytidine deaminase

    Cancer Res.

    (1998)
  • H.K. Eltzschig et al.

    HIF-1-dependent repression of equilibrative nucleoside transporter (ENT) in hypoxia

    J. Exp. Med.

    (2005)
  • G. Ferrandina et al.

    Expression of nucleoside transporters, deoxycitidine kinase, ribonucleotide reductase regulatory subunits, and gemcitabine catabolic enzymes in primary ovarian cancer

    Cancer Chemother. Pharmacol.

    (2010)
  • C.G. Ferreira et al.

    Chemotherapy triggers apoptosis in a caspase-8-dependent and mitochondria-controlled manner in the non-small cell lung cancer cell line NCI-H460

    Cancer Res.

    (2000)
  • N. Funamizu et al.

    Is the resistance of gemcitabine for pancreatic cancer settled only by overexpression of deoxycytidine kinase?

    Oncol. Rep.

    (2010)
  • A. Fusco et al.

    Roles of HMGA proteins in cancer

    Nat. Rev. Cancer

    (2007)
  • V. Gandhi et al.

    Excision of 2′,2′-difluorodeoxycytidine (gemcitabine) monophosphate residues from DNA

    Cancer Res.

    (1996)
  • E. Giovannetti et al.

    Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine

    Cancer Res.

    (2006)
  • S. Gourgou-Bourgade et al.

    Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial

    J. Clin. Oncol.

    (2013)
  • V. Heinemann et al.

    Comparison of the cellular pharmacokinetics and toxicity of 2′,2′-difluorodeoxycytidine and 1-beta-D-arabinofuranosylcytosine

    Cancer

    (1988)
  • V. Heinemann et al.

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

    Cancer Res.

    (1992)
  • V. Heinemann et al.

    Inhibition of ribonucleotide reduction in CCRF-CEM cells by 2′,2′-difluorodeoxycytidine

    Mol. Pharmacol.

    (1990)
  • L.S. Hodge et al.

    The deaminated metabolite of gemcitabine, 2′,2′-difluorodeoxyuridine, modulates the rate of gemcitabine transport and intracellular phosphorylation via deoxycytidine kinase

    Drug Metab. Dispos.

    (2011)
  • R. Hu et al.

    UMP/CMPK is not the critical enzyme in the metabolism of pyrimidine ribonucleotide and activation of deoxycytidine analogs in human RKO cells

    PLoS One

    (2011)
  • Cited by (408)

    • Liposomal nanostructures for Gemcitabine and Paclitaxel delivery in pancreatic cancer

      2023, European Journal of Pharmaceutics and Biopharmaceutics
    View all citing articles on Scopus
    View full text