Elsevier

European Journal of Cancer

Volume 54, February 2016, Pages 75-83
European Journal of Cancer

Review
Targeting KRAS for diagnosis, prognosis, and treatment of pancreatic cancer: Hopes and realities

https://doi.org/10.1016/j.ejca.2015.11.012Get rights and content

Abstract

Mutation of the KRAS oncogene in pancreatic cancer is responsible for permanent activation of the P21 RAS protein and the cascade of signalling pathways. Consequently, multiple cellular processes, such as transformation, proliferation, invasion, and survival are activated. The aim of this review was to present all potential clinical applications of targeting KRAS in terms of diagnosis and management of pancreatic adenocarcinoma. Quantitative polymerase chain reaction technology provides reliable assessment of KRAS mutations, both in tissues and from fine-needle aspiration biopsies. Numerous studies report that the combination of endoscopic ultrasound-guided cytopathology and a KRAS mutation assay can improve the positive and differential diagnosis of pancreatic cancer, differentiating between benign versus malignant solid pancreatic cancer, and reducing false-negative results compared to cytopathology alone. In addition, the presence of a KRAS mutation is frequently associated with a worse prognosis, both in cases of advanced and resected tumours. However, the KRAS mutation assay is not as efficient at predicting a response to both anti-epidermal growth factor receptor treatments and/or chemotherapy. Targeting of KRAS to treat pancreatic adenocarcinoma has been applied at different stages of RAS molecular intracellular processes: at the transcription level with antisense or interference RNA, at the posttranslational level with inhibitors of farnesyl transferase or anti-RAS vaccination peptides, and to target multiple signalling pathways using inhibitors of mitogen-activated protein kinase, phosphoinositide 3-kinase, AKT, mammalian target of rapamycin, RAF. Despite some encouraging results at pre-clinical and phase I stages, no significant clinical benefits have been observed. Combinatory approaches with standard chemotherapy will be welcome.

Introduction

Pancreatic cancer remains one of the most deadly types of cancer: the 5-year survival rate after diagnosis is <3.5% [1]. The sole curative treatment is surgical resection, which is, unfortunately, applicable in no more than 15% of cases. Single-agent gemcitabine, FOLFIRINOX, and nab-paclitaxel–gemcitabine protocols, although not dramatically improving survival beyond 11 months, have demonstrated significant clinical benefits and have become the standard chemotherapy for advanced and metastatic pancreatic ductal adenocarcinoma (PDAC) [1], [2], [3], [4], [5]. Our understanding of pancreatic carcinoma has increased greatly from experimental models of genetic/epigenetic alterations and molecular expression, from analyses of pre-cancerous and cancerous tissues, from the use of molecular amplification, and from large-scale transcriptome analyses.

INK4a/ARF, TP53, DPC4/Smad4 tumour-suppressor pathways are genetically inactivated in the majority of pancreatic carcinomas (associated with losses of heterozygosity of, respectively, 9p21, 17p, and 18q), whereas oncogenic KRAS is activated [6], [7], [8]. At a late stage of tumour development, there is an increase in telomerase activity, over-expression of growth factors and/or their receptors (Epidermal growth factor [EGF], nerve growth factor, gastrin), and of pro-angiogenic factors (Vascular endothelial growth factor [VEGF], Fibroblast growth factor [FGF], Platelet derived growth factor [PDGF]), and increased invasive factors (metalloproteinases, tissue plasminogen activators). The microenvironment plays also a key role in the invasive and metastatic process of pancreatic carcinoma, with a strong relationship between cancerous cells and pancreatic stellate cells, as well as the extracellular matrix. This microenvironment strongly participates in tumour fibrosis, hypoxia, and hypovascularisation, resulting in inaccessibility of drugs [8], [9]. However, the activating point mutation of the KRAS oncogene on codon 12 (Exon 2) remains the major event (70–95% of PDAC cases: 71% of pancreatic cancer specimens in the COSMICS database harbour KRAS mutations) [10].

This mutation occurs early in pancreatic carcinogenesis as attested by its presence in common pre-neoplastic lesions, such as PanIN (pancreatic intraductal neoplasia) and intraductal mucinous papillary pancreatic neoplasia [8], [12]. The single-nucleotide mutation induces a replacement of the GGT sequence (encoding for glycine) by the GAT sequence (aspartic acid – G12D – c35 G > A), GTT (valine – G12V – c35 G > T), CGT (arginine – G12R – C34 G > C), or GCT (alanine – G12A – c35 G > C). A point mutation can also occur, but less frequently, on codon 13 (G13D) or 61 (Q61L or Q61H) [7], [8], [10].

The KRAS gene encodes for the protein P21 RAS, which is a small Guanosine triphosphatase (GTPase) that acts as a molecular switch by coupling cell-membrane growth factor receptors to intracellular signalling pathways and transcription factors to control various cellular processes. P21 RAS is localised in the inner surface cell-membrane and interacts with more than 20 effector proteins. The RAS protein requires membrane association for its biological activity. This membrane association results in modification of lipids by a farnesyl isoprenoid. The point mutation of KRAS impairs intrinsic GTPase activity of RAS and prevents GTPase activating proteins to promote conversion of Guanosine triphosphate (GTP) (active) to Guanosine diphosphate (GDP) (inactive). P21 RAS is thus permanently bound to GTP and activates downstream signalling pathways, such as phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) or RAF/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinases (independently of upstream growth factor receptor activation) [11]. Following this activation, nuclear transcription factors are also activated (such as ELK/JUN/MYC) with stimulation of cell proliferation, transformation, adhesion, and survival (posttranslational regulation and signalling of RAS are summarised in Fig. 1) [11], [12].

Studying KRAS mutations has improved our understanding of the processes involved in transformation, uncontrolled proliferation, and invasion of pancreatic cancer cells, as well as the development of transgenic animal models. Nowadays, these models are at the centre of all pathophysiological studies, especially since the creation of the KC KRASG12D mouse model [13]. These mice reliably reproduce the human pathology with the presence of various PanIN lesions in 100% of animals. This confirms the initiating role of a KRAS mutation in pancreatic carcinogenesis, but also the importance of multistep genetic mutations in pancreatic carcinogenesis. Although KC mice develop PDAC in only 10% of cases, when inactivation of TP53, Smad4 and Ink4A/Arf is added after crossbreeding of transgenic mice, significant cancerous lesions are obtained [14], [15], [16], [17]. By using these models, the role of the signalling pathway, stroma, and microenvironment can be studied, as well as the co-factors, such as inflammation [18], [19]. These murine models offer also a wide range of applications in the domain of diagnosis and therapy. Biomarkers can be tested in blood and imaging such as positron emission tomography scan, computed tomography or ultrasound can be applied [13], [20], [21], [22], [23], [24]. These models displayed not only primary tumours with an organised tumour microenvironment but also metastasis. All these lesions cannot be reproduced by means of transplantable models. In consequence, pre-clinical test and proof of concept can be conducted for future antitumour agents [22], [23].

Activation of the RAS pathway is also important in the carcinogenesis of various types of tumours [25], [26], [27], [28]. Because the RAS-mediated signalling pathway links downstream growth factors (especially epidermal growth factor receptor [EGFR]), studies conducted in colon and lung cancer have found that the KRAS mutation and EGFR expression can be indicators for a poor prognosis [25], [26], [27]. Moreover, the absence of a KRAS mutation is correlated with the therapeutic response of colon carcinoma to anti-EGFR antibodies and the therapeutic response of lung cancer to anti-EGFR/HER molecules, with or without chemotherapy [28], [29], [30]. Taking into account the high frequency of KRAS mutations in PDAC and the major role of this mutation in proliferation and progression of pancreatic cancer, numerous studies have been conducted over the past 20 years to investigate if targeting the KRAS mutation can be applied clinically to either diagnose or provide a prognosis or treatment for PDAC. The aim of this review was to synthesise all the published ‘positive’ and ‘negative’ studies in this field to gain a straightforward picture of the main issues in terms of the possible applications of targeting the KRAS mutation in current clinical practice.

Section snippets

KRAS mutation assay to improve positive and differential diagnosis of PDAC

Nowadays due to its invasiveness, ERCP is focused on therapeutic approaches of biliary and pancreatic disease. More than 15 years of endoscopic ultrasound (EUS) experience now allows safe guided fine-needle aspiration biopsies (FNA) of solid pancreatic lesions for cytopathological analysis [31]. EUS-guided FNA (EUS-FNA) is, thus, now an effective technique to diagnose and assess the staging of PDAC [31]. However, its accuracy to diagnose malignancy varies widely, with a sensitivity ranging from

KRAS mutation assay to assess a prognosis for PDAC

Several groups, including ours, have investigated whether the presence or not of a KRAS mutation can influence the prognosis of PDAC, especially in advanced tumours that are only investigated using EUS-FNA. All studies that have included ≥50 patients (range: 50–272) are summarised in Table 2. The numbers of KRAS mutations found in samples (biopsies or resected specimens) varied between 41 and 75%, and last, populations included either resected or non-resectable (locally advanced and/or

Therapeutic approaches to targeting KRAS

Several strategies have been proposed for targeting the RAS protein. The main strategies and molecular targets are shown in Fig. 1. The RAS-membrane association is induced by farnesyl transferase, which attaches a C15 farnesyl isoprenoid lipid to the cysteine of the RAS-terminal CAXX motif; however, the use of farnesyl transferase inhibitors (such as tipifarnib or lonafarnib) showed no clinical benefit [70], [71], [72]. This is because several RAS isoforms, such as NRAS, do not rely on

Conclusion

The oncogene KRAS point mutation is the major molecular event in PDAC. Nowadays, qPCR technology enables reliable assessment of KRAS mutations, both from tissues and from FNA biopsies. Numerous studies report that the cytopathology and KRAS mutation assay used on EUS-FNA material can improve the positive and differential diagnosis of PDAC. In this way, benign versus malignant solid masses can be more easily distinguished, thus avoiding unnecessary pancreatectomies. Conversely, in cases of

Conflict of interest statement

The authors have no conflict of interest to declare.

References (90)

  • T.J. Savides et al.

    EUS-guided FNA diagnostic yield of malignancy in solid pancreatic masses: a benchmark for quality performance measurement

    Gastrointest Endosc

    (2007)
  • K.J. Livak

    Allelic discrimination using fluorogenic probes and 5' nuclease assay

    Genet Anal

    (1999)
  • A. Didelot et al.

    Competitive allele specific TaqMan PCR for KRAS, BRAF and EGFR mutation detection in clinical formalin fixed paraffin embedded samples

    Exp Mol Pathol

    (2012)
  • L. Fuccio et al.

    The role of K-ras gene mutation analysis in EUS-guided FNA cytology specimens for the differential diagnosis of pancreatic solid masses: a meta-analysis of prospective studies

    Gastrointest Endosc

    (2013)
  • M. Tada et al.

    Quantitative analysis of K-ras gene mutation in pancreatic tissue obtained by endoscopic ultrasonography-guided fine needle aspiration: clinical utility for diagnosis of pancreatic tumor

    Am J Gastroenterol

    (2002)
  • K. Takahashi et al.

    Differential diagnosis of pancreatic cancer and focal pancreatitis by using EUS-guided FNA

    Gastrointest Endosc

    (2005)
  • T. Ogura et al.

    Clinical impact of K-ras mutation analysis in EUS-guided FNA specimens from pancreatic masses

    Gastrointest Endosc

    (2012)
  • A. Khalid et al.

    EUS-FNA mutational analysis in differentiating autoimmune pancreatitis and pancreatic cancer

    Pancreatology

    (2011)
  • J. Franko et al.

    Loss of heterozygosity predicts poor survival after resection of pancreatic adenocarcinoma

    J Gastrointest Surg

    (2008)
  • H. Chen et al.

    K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer

    Eur J Surg Oncol

    (2010)
  • J.B. Bachet et al.

    S100A2 is a predictive biomarker of adjuvant therapy benefit in pancreatic adenocarcinoma

    Eur J Cancer

    (2013)
  • S. Mosolits et al.

    Therapeutic vaccination in patients with gastrointestinal malignancies. A review of immunological and clinical results

    Ann Oncol

    (2005)
  • A. Ackley et al.

    An algorithm for generating small RNAs capable of epigenetically modulating transcriptional gene silencing and activation in human cells

    Mol Ther Nucleic Acids

    (2013)
  • J.R. Infante et al.

    Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial

    Lancet Oncol

    (2012)
  • J.R. Infante et al.

    A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas

    Eur J Cancer

    (2014)
  • C. Montagut et al.

    Targeting the RAF-MEK-ERK pathway in cancer therapy

    Cancer Lett

    (2009)
  • D.P. Ryan et al.

    Pancreatic adenocarcinoma

    N Engl J Med

    (2014)
  • H.A. Burris et al.

    Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial

    J Clin Oncol

    (1997)
  • T. Conroy et al.

    FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer

    N Engl J Med

    (2011)
  • D.D. Von Hoff et al.

    Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine

    N Engl J Med

    (2013)
  • M. Ducreux et al.

    Treatment of advanced pancreatic cancer

    Semin Oncol

    (2007)
  • S.A. Hahn et al.

    Recent discoveries in cancer genetics of exocrine pancreatic neoplasia

    Digestion

    (1998)
  • Y. Delpu et al.

    Genetic and epigenetic alterations in pancreatic carcinogenesis

    Curr Genomics

    (2011)
  • S.A. Forbes et al.

    COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in Cancer

    Nucleic Acids Res

    (2011)
  • A.D. Cox et al.

    Targeting RAS Membrane Association: back to the future for anti-RAS drug discovery?

    Clin Cancer Res

    (2015)
  • A.J. Aguirre et al.

    Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma

    Genes Dev

    (2003)
  • N. Bardeesy et al.

    Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse

    Proc Natl Acad Sci U S A

    (2006)
  • C.V. Rao et al.

    Inhibition of pancreatic intraepithelial neoplasia progression to carcinoma by nitric oxide-releasing aspirin in p48(Cre/+)-LSL-Kras(G12D/+) mice

    Neoplasia

    (2012)
  • M.A. Pysz et al.

    Vascular endothelial growth factor receptor type 2-targeted contrast-enhanced US of pancreatic cancer neovasculature in a genetically engineered mouse model: potential for earlier detection

    Radiology

    (2015)
  • A. Neesse et al.

    SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice

    Gut

    (2014)
  • B. Alagesan et al.

    Combined MEK and PI3K inhibition in a mouse model of pancreatic cancer

    Clin Cancer Res

    (2015)
  • L. Ligat et al.

    Pancreatic preneoplastic lesions plasma signatures and biomarkers based on proteome profiling of mouse models

    Br J Cancer

    (2015 Oct 29)
  • A. Russo et al.

    Prognostic and predictive factors in colorectal cancers: Kirsten ras in CRC (RASCAL) and TP53CRC collaborative studies

    Ann Oncol

    (2005)
  • A. Lièvre et al.

    KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab

    J Clin Oncol

    (2008)
  • A. Lièvre et al.

    KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer

    Cancer Res

    (2006)
  • Cited by (149)

    View all citing articles on Scopus
    View full text