Open Access

Dysregulation of KRAS signaling in pancreatic cancer is not associated with KRAS mutations and outcome

  • Authors:
    • Radmila Lemstrova
    • Veronika Brynychova
    • David J. Hughes
    • Viktor Hlavac
    • Pavel Dvorak
    • Joanne E. Doherty
    • Helena A. Murray
    • Martin Crockard
    • Martin Oliverius
    • Jan Hlavsa
    • Eva Honsova
    • Jan Mazanec
    • Zdenek Kala
    • Martin Lovecek
    • Roman Havlik
    • Jiri Ehrmann
    • Ondrej Strouhal
    • Pavel Soucek
    • Bohuslav Melichar
    • Beatrice Mohelnikova-Duchonova
  • View Affiliations

  • Published online on: September 14, 2017     https://doi.org/10.3892/ol.2017.6946
  • Pages: 5980-5988
  • Copyright: © Lemstrova et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pancreatic ductal adenocarcinoma (PDAC) is a tumor with a poor prognosis, and no targeted therapy is curre­ntly available. The aim of the present study was to investigate the prognostic significance of the expression of V‑Ki‑ras2 Κirsten rat sarcoma viral oncogene homolog (KRAS), downstream signaling pathway genes and the association with clinical characteristics in PDAC patients undergoing radical surgery. Tumors and adjacent non‑neoplastic pancreatic tissues were examined in 45 patients with histologically verified PDAC. KRAS and B‑Raf proto‑oncogene, serine/threonine kinase (BRAF) gene mutation analysis was performed using the KRAS/BRAF/phosphatidylinositol‑4,5‑bisphosphate 3‑kinase catalytic subunit α array. The transcript profile of 52 KRAS downstream signaling pathway genes was assessed using quantitative‑polymerase chain reaction. KRAS mutation was detected in 80% of cases. The genes of four signaling pathways downstream of KRAS, including the phosphoinositide 3‑kinase/3‑phosphoinositide‑dependent protein kinase 1/V‑akt murine thymoma viral oncogene homolog 1, RAL guanine nucleotide exchange factor, Ras and Rab interactor 1/ABL proto‑oncogene‑1, non‑receptor tyrosine kinase, and RAF proto‑oncogene serine/threonine‑protein kinase/mitogen‑activated protein kinase pathways, exhibited differential expression in PDAC compared with that in the adjacent normal tissues. However, no significant differences in expression were evident between patients with KRAS‑mutated and wild‑type tumors. The expression of KRAS downstream signaling pathways genes did not correlate with angioinvasion, perineural invasion, grade or presence of lymph node metastasis. Additionally, the presence of KRAS mutations was not associated with overall survival. Among the KRAS downstream effective signaling pathways molecules investigated, only v‑raf‑1 murine leukemia viral oncogene homolog 1 expression was predictive of prognosis. Overall, KRAS mutation is present in the majority of cases of PDAC, but is not associated with changes in the expression of KRAS downstream signaling pathways and the clinical outcome. This may partly explain the failure of KRAS‑targeted therapies in PDAC.

Introduction

Pancreatic ductal adenocarcinoma (PDAC; Online Mendelian Inheritance in Man no. 260350) ranks fourth in the leading causes of cancer-associated mortality in Western countries (1). Despite diagnostic and therapeutic advances, the prognosis of PDAC remains poor. Only 20% of patients present with potentially resectable disease at the time of diagnosis, while due to the high propensity for tumor recurrence, the 5-year overall survival (OS) rate in patients undergoing surgery with radical intent is usually <25%. Although a number of prospective clinical trials have demonstrated that adjuvant systemic therapy improves the patient outcome following surgery, adjuvant chemotherapy appears to be effective only in a minority of patients, and the majority of the patients ultimately succumb to the disease. The prognosis of metastatic patients is extremely poor, with a median OS time of <1 year (2). Consequently, novel regimens of adjuvant treatment are being investigated and there is currently no definitive standard of adjuvant therapy.

In PDAC, mutations in the V-Ki-ras2 Κirsten rat sarcoma viral oncogene homolog (KRAS) gene occur in 75–90% of cases, representing the most frequent, as well as the earliest, genetic alteration. KRAS mutations, specifically in codons 12 and 13, lead to constitutive activation of downstream signaling pathways that are important for tumor initiation, development and spread (3,4). KRAS signaling is highly complex and dynamic, with various downstream effector pathways interconnected at different levels by cross-signaling and feedback loops (5). The four major KRAS downstream pathways reported in PDAC are RAF proto-oncogene serine/threonine-protein kinase (RAF)/mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/3-phosphoinositide-dependent protein kinase 1 (PDK1)/ABL proto-oncogene-1, non-receptor tyrosine kinase (ABL), RAL guanine nucleotide exchange factor, and Ras and Rab interactor 1 (RIN1)/ABL pathways (Fig. 1) (610). This multiplicity of downstream pathways may partly explain the failure of existing efforts to target epidermal growth factor receptor, KRAS or serine/threonine-protein kinase B-raf (BRAF) using specific inhibitors, underlining the complexity of genetic changes and the resistance of the cancer cells.

The aim of the present study was to assess the association between gene expression from the four major KRAS-effective pathways in PDAC and the clinical features of the patients, and to evaluate the potential predictive and prognostic significance.

Materials and methods

Patients

A cohort of 45 consecutive patients with PDAC who underwent surgery with curative intent was recruited from two oncology centers in the Czech Republic (Institute of Clinical and Experimental Medicine, Prague; and University Hospital, Masaryk University, Brno, Czech Republic) between August 2008 and January 2012. Inclusion criteria were: i) Adult operable patients with suspected pancreatic carcinoma based on clinical imaging methods; ii) patients who provided informed consent; and iii) pancreatic carcinoma diagnosis was verified by collaborating pathologist. None of the patients had received prior chemotherapy. Characteristics of the patient cohort are summarized in Table I. The tissue specimen collection and processing, and the data retrieval were as described previously (11).

Table I.

Characteristics of the patient cohort.

Table I.

Characteristics of the patient cohort.

Variables20
Mean age (range), years63.9 (46–80)
Sex, n (%)
  Male20 (44.4)
  Female25 (55.6)
Histological grade, n (%)
  G1+G2 (well to moderate)30 (66.7)
  G3+G4 (poor)15 (33.3)
Primary tumor extent of invasion, n (%)
  pT1 tumor limited to the pancreas ≤2 cm1 (2.2)
  pT2 tumor limited to the pancreas >2 cm5 (11.1)
  pT3 tumor extending beyond the pancreas39 (86.7)
Regional lymph nodes, n (%)
  pN017 (37.8)
  pN128 (62.2)
KRAS mutations in codons 12 and 13a, n (%)
  Wild-type (GGTGGC)9 (20.0)
  G12V (GTTGGC)10 (22.2)
  G12D (GATGGC)15 (33.3)
  G12R (CGTGGC)6 (13.3)
  Other (G13D, Q61R, Q61H)3 (6.7)
  Not assessed2 (4.4)
BRAF mutationsa, n (%)
  Wild-type (GTG)43 (95.6)
  V600E (GAG)0 (0.0)
  Not assessed2 (4.4)
Patient status at the data cut off, n (%)
  Deceased37 (82.2)
  Alive8 (17.8)

a Base changes are underlined. BRAF, B-Raf proto-oncogene, serine/threonine kinase; KRAS, KRAS proto-oncogene, GTPase.

All patients signed an informed consent form, in accordance with the requirements for ethical approval, which was provided by the Institutional Review Boards of the Institute of Clinical and Experimental Medicine and University Hospital, Masaryk University, Brno.

Isolation of nucleic acids and cDNA synthesis

Tissue samples were homogenized and total RNA and DNA was isolated as previously described (12,13). cDNA was synthesized using 0.5 µg total RNA and characterized as previously described (14). cDNA was then pre-amplified by TaqMan® PreAmp Master mix to enrich the specific targets for gene expression analysis using TaqMan Gene Expression assays (Life Technologies; Thermo Fisher Scientific, Inc., Waltham, MA, USA) (Table II). The cDNA pre-amplification was performed with 5 µl cDNA using 14 pre-amplification cycles (10 min at 95°C and 14 cycles of 15 sec at 95°C), and the pre-amplification uniformity of cDNA was checked according to the procedure recommended by the manufacturer (Thermo Fisher Scientific, Inc.).

Table II.

List of TaqMan gene expression assays used in the study.

Table II.

List of TaqMan gene expression assays used in the study.

Gene abbreviationGene nameAssay ID
AKT1V-akt murine thymoma viral oncogene homolog 1Hs00178289_m1
AKT2V-akt murine thymoma viral oncogene homolog 2Hs01086102_m1
ARAFV-raf murine sarcoma viral oncogene homolog 1Hs00176427_m1
BRAFV-raf murine sarcoma viral oncogene homolog Β1Hs00269944_m1
GRB2Growth factor receptor-bound protein 2Hs00257910_s1
GSK3BGlycogen synthase kinase 3-βHs00275656_m1
KRASV-ki-ras2 Κirsten rat sarcoma viral oncogene homologHs00364284_g1
MAP2K1Mitogen-activated protein kinase kinase 1Hs00983247_g1
MAP2K2Mitogen-activated protein kinase kinase 2Hs04194606_gH
MAP2K7Mitogen-activated protein kinase kinase 7Hs00178198_m1
MAP3K1Mitogen-activated protein kinase kinase kinase 1Hs00394890_m1
MAP3K2Mitogen-activated protein kinase kinase kinase 2Hs01109981_m1
MAP3K4Mitogen-activated protein kinase kinase kinase 4Hs00245958_m1
MAP3K7Mitogen-activated protein kinase kinase kinase 7Hs01105682_m1
MAPK1Mitogen-activated protein kinase 1Hs01046830_m1
MAPK10Mitogen-activated protein kinase 10Hs00373455_m1
MAPK14Mitogen-activated protein kinase 14Hs01051152_m1
MAPK3Mitogen-activated protein kinase 3Hs00946872_m1
MAPK7Mitogen-activated protein kinase 7Hs00611114_g1
MAPK8Mitogen-activated protein kinase 8Hs00177083_m1
MAPK9Mitogen-activated protein kinase 9Hs00177102_m1
MKNK1Mitogen-activated protein kinase-interacting serine/threonine kinase 1Hs00374376_m1
MKNK2Mitogen-activated protein kinase-interacting serine/threonine kinase 2Hs01046586_g1
MTORMechanistic target of rapamycinHs00234508_m1
PAK1p21 protein-activated kinase 1Hs00176815_m1
PDPK1 3-phosphoinositide-dependent protein kinase 1Hs00176884_m1
PIK3CA Phosphatidylinositol 3-kinase, catalytic, αHs00907966_m1
PIK3CG Phosphatidylinositol 3-kinase, catalytic, γHs00277090_m1
PLK3Polo-like kinase 3Hs00177725_m1
PRKACAProtein kinase, camp-dependent, catalytic, αHs00427274_m1
PRKCAProtein kinase c, αHs00925195_m1
PTENPhosphatase and tensin homologHs02621230_s1
PTK2Protein-tyrosine kinase, cytoplasmicHs01056457_m1
PTK2BProtein-tyrosine kinase 2, βHs01559708_m1
RAC1Ras-related C3 botulinum toxin substrate 1Hs01025984_m1
RAF1V-raf-1 murine leukemia viral oncogene homolog 1Hs00234119_m1
RALAV-ral simian leukemia viral oncogene homolog AHs01564991_g1
RALBP1RalA-binding protein 1Hs01034988_g1
RALGDSRal guanine nucleotide dissociation stimulatorHs00325141_m1
RAP1ARas-related protein 1AHs01092205_g1
RASA1Ras p21 protein activator 1Hs00963555_m1
RASA2Ras p21 protein activator 2Hs01003325_m1
RHOARas homolog gene family, member AHs00357608_m1
RIN1Ras and rab interactor 1Hs00182870_m1
RPS6KA2Ribosomal protein S6 kinase, 90-kd, 2Hs00179731_m1
RPS6KA4Ribosomal protein S6 kinase, 90-kd, 4Hs00177670_m1
RPS6KA5Ribosomal protein S6 kinase, 90-kd, 5Hs01046594_m1
SHC1SHC transforming proteinHs01050699_g1
SOS1Son of sevenless, Drosophila, homolog 1Hs00362316_m1
SOS2Son of sevenless, Drosophila, homolog 2Hs00412876_g1
SRCV-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogeneHs01082238_g1
STAT3Signal transducer and activator of transcription 3Hs01047580_m1
ELF1aE74-like factor 1Hs00152844_m1
EIF2B1aEukaryotic translation initiation factor 2B, subunit 1Hs00426752_m1
MRPL19aMitochondrial ribosomal protein l19Hs00608519_m1
POP4 aProcessing of precursor 4, S. cerevisiae, homolog ofHs00198357_ml

a Reference genes.

Quantitative polymerase chain reaction (qPCR)

qPCR was performed using the ViiA7 Real-Time PCR system using TaqMan Gene Expression assays (Table I), with optimized primer and probe sets and TaqMan Gene Expression Master mix (Thermo Fisher Scientific, Inc.). Processing of precursor 4, S. cerevisiae, homolog of, mitochondrial ribosomal protein L19, E74-like factor 1 and eukaryotic translation initiation factor 2B subunit 1 were used as reference genes for studies of gene expression in human pancreatic carcinoma based on our previously published data (15). Determination of transcript levels was performed exactly as previously described (10) and the qPCR study adhered to the Minimum Information for Publication of Quantitative Real-Time PCR Experiments Guidelines (16).

KRAS and BRAF mutation status

KRAS and BRAF gene mutation analysis was performed using the KRAS/BRAF/phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) (KBP) Array (EV3799A/B; Randox Laboratories Ltd., Crumlin, Northern Ireland) according to the manufacturer's instructions. The assay is based on a combination of multiplex PCR and biochip array hybridization for high discrimination between multiple wild-type and mutant DNA regions in the KRAS (mutations in codons 12, 13 and 61), BRAF (V600E mutation) and PIK3CA (mutations in codons 542, 545 and 1,047) genes. Providing there are enough copies of DNA present, ~1% of mutants can readily be detected in a background of wild-type genomic DNA. A unique primer set is designed for each mutation target (and control), which will hybridize to a complementary discrete test region (DTR) on the biochip array. Each DTR corresponds to a particular mutation target. One of the target-specific primers in each pair contains a biotin label, which on addition of streptavidin-horseradish peroxidase conjugate permits chemiluminescence detection of hybridized products on the biochip array. Dedicated software processes produced automatic results.

Statistical analysis

Differences in gene expression profiles between tumor and paired non-neoplastic control tissues and between wild-type and KRAS-mutated samples were evaluated using the RT2 Profiler PCR Assay Data Analysis v3.5 program (Qiagen GmbH, Hilden, Germany). This gene expression analysis suite performs fold-change calculations from raw quantification cycle values for reference and target genes based on the ΔΔCq method described by Livak and Schmittgen (17), and enables hierarchical clustering of gene expression profiles between the compared groups of patients and data. Differences in intratumoral gene expression levels between patients stratified by clinical data were evaluated by the Kruskal-Wallis test.

OS was defined as the time between the date of surgery and all-cause mortality. Surviving patients were censored at the last follow-up in December 2015. Patients were divided into two groups by the median intratumoral gene expression levels of individual genes and the survival functions were computed by the Kaplan-Meier method, with statistical significance evaluated by the Breslow test using SPSS v16.0 (SPSS, Inc., Chicago, IL, USA).

P<0.05 was considered to indicate a statistically significant difference. All P-values are departures from two-sided tests. The correction for multiple testing was applied according to the Bonferroni and the false discovery rate (FDR) methods.

Results

Study population

The study was performed on 45 patients with resected (R0 resection in >90% of cases) PDAC who had not received any prior neoadjuvant therapy. Overall, 80% (36/45) of patients harbored KRAS mutations in the DNA of the tumor tissue, while BRAF mutations were not found in any sample (Table II). The majority of patients (76%; n=34) received adjuvant chemotherapy consisting of nucleoside analogs (gemcitabine and/or 5-fluorouracil).

The median OS time was 23.7 months, with 18% of patients (n=8) alive at the time of data cut off (December 2015).

Transcript levels of KRAS signaling pathways genes in PDAC

Considering the pivotal role of KRAS oncogene in the integration and transduction of mitogenic and metabolic signals, the transcript levels of 52 genes covering four major pathways downstream of KRAS were measured (Table I). The KRAS pathway was significantly dysregulated in tumors compared with that in adjacent non-malignant pancreatic tissues (Fig. 2; Table III). Significant overexpression of genes of the PI3K/PDK1/AKT, RAL guanine nucleotide exchange factor, and RIN1/ABL [phosphatidylinositol 3-kinase, catalytic, α/γ (PIK3CA/G), p21 protein-activated kinase 1, V-ral simian leukemia viral oncogene homolog A, RalA-binding protein 1, Ras-related C3 botulinum toxin substrate 1, RIN1, protein-tyrosine kinase, cytoplasmic, and V-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogene] pathways were observed, leading to cytoskeletal remodeling, endocytosis, cell spreading and migration (Table III; Fig. 1). By contrast, genes of the RAF/MAPK pathway exhibited significantly lower expression in tumors compared with that in the paired adjacent non-malignant pancreatic tissues (particularly in genes ARAF, BRAF, V-RAF-1 murine leukemia viral oncogene homolog 1 (RAF1), mitogen-activated protein kinase kinase, mitogen-activated protein kinase 1, mitogen-activated protein kinase-interacting serine/threonine kinase 1/2 (MKNK1/2) and ribosomal protein S6 kinase, 90-kd, 2. All these results remained significant after FDR adjustment for multiple testing and the majority remained significant after Bonferroni correction (Table III; Fig. 1).

Table III.

Dysregulation of KRAS pathway genes in pancreatic ductal adenocarcinoma tumors in comparison to paired adjacent non-malignant tissues.

Table III.

Dysregulation of KRAS pathway genes in pancreatic ductal adenocarcinoma tumors in comparison to paired adjacent non-malignant tissues.

Gene Fold-changea (tumor vs. non-malignant tissue)95% confidence intervala P-valuea
AKT10.73(0.63–0.83)<0.001b
ARAF0.72(0.63–0.81) <0.001b
BRAF0.84(0.74–0.93)0.001
GRB2c1.37(1.04–1.69) <0.001b
KRASc2.04(1.67–2.41) <0.001b
MAP2K20.64(0.46–0.82)<0.001b
MAP2K70.52(0.39–0.65) <0.001b
MAP3K10.85(0.75–0.95)0.010
MAP3K2c1.24(1.13–1.36) <0.001b
MAP3K7c1.28(1.14–1.41) <0.001b
MAPK10.77(0.59–0.94)<0.001b
MAPK14c1.27(1.14–1.40)<0.001b
MAPK3c1.71(1.26–2.15)<0.001b
MAPK7c1.20(1.01–1.38)0.006
MAPK80.81(0.74–0.88)<0.001b
MAPK90.47(0.38–0.55) <0.001b
MKNK10.31(0.25–0.38)<0.001b
MKNK20.35(0.26–0.44) <0.001b
PAK1c1.27(1.08–1.45)0.001
PDPK10.73(0.64–0.81) <0.001b
PIK3CAc1.46(1.25–1.68) <0.001b
PIK3CGc2.22(1.61–2.82) <0.001b
PLK3c1.56(1.23–1.88) <0.001b
PTENc1.29(1.05–1.53)0.006
PTK2Bc1.68(1.44–1.91)<0.001b
RAC1c1.65(1.34–1.96)<0.001b
RAF10.62(0.54–0.69) <0.001b
RALAc1.43(1.27–1.59) <0.001b
RALBP1c1.60(1.39–1.81) <0.001b
RAP1Ac1.18(1.07–1.29) <0.001b
RASA1c1.28(1.12–1.43) <0.001b
RASA2c1.87(1.51–2.23) <0.001b
RHOAc1.23(1.13–1.34) <0.001b
RIN1c1.39(1.10–1.67)0.002
RPS6KA20.65(0.49–0.81)0.001b
RPS6KA4c1.76(1.45–2.08) <0.001b
SHC1c1.24(1.09–1.38)0.001b
SOS1c1.32(1.14–1.50) <0.001b
SOS20.68(0.59–0.77)<0.001b
SRCc1.43(1.16–1.71)<0.001b

a Fold-change, 95% confidence interval and P-values were calculated using RT2 Profiler PCR Assay Data analysis v3.5 program

b result that passed Bonferroni's correction for 52 analyzed genes (cut off P=0.001)

c upregulated genes. There were 14 additional genes whose expression was not statistically significantly changed and that are therefore not listed in the table: AKT2, GSK3B, MAP2K1, MAP3K4, MAPK10, MTOR, PRKACA, PRKCA, PTEN, PTK2, RALGDS, RAP1A, RPS6KA and STAT3.

However, no association between KRAS downstream signaling pathway gene expression and tumor characteristics, including tumor size, grade, angioinvasion, lymph node metastasis or perineural invasion, passed the significance threshold of the Bonferroni test.

Impact of KRAS mutation status on transcript levels of target genes

From the 80% of tumor samples with KRAS mutations, the most common mutation, KRASG12D, was present in 33% (n=15) of the tumors. Only 1 tumor was found with a mutation in codon 13, and 2 cases with a mutation in codon 61 (Table II).

Patients divided by the KRAS mutation status significantly differed in terms of the gene expression of 5 of the 52 analyzed genes [BRAF, mitogen-activated protein kinase kinase kinase 4, mitogen-activated protein kinase 8, MKNK1 and son of sevenless, Drosophila, homolog 2 (SOS2; P<0.05; Table IV)], but none of these associations passed the threshold for the multiple testing correction. The expression profiles of the KRAS signaling pathway as a whole also did not significantly differ between KRAS wild-type and KRAS-mutated tumors (Fig. 3).

Table IV.

Downregulation of KRAS pathway genes in PDAC KRAS-mutated tumors compared with cases with wild-type KRAS.

Table IV.

Downregulation of KRAS pathway genes in PDAC KRAS-mutated tumors compared with cases with wild-type KRAS.

Gene Fold-changea (tumor vs. non-tumor)95% confidence intervala P-valuea
BRAF0.84(0.72–0.95)0.021
MAP3K40.79(0.67–0.91)0.035
MAPK80.84(0.71–0.97)0.027
MKNK10.72(0.45–0.99)0.033
SOS20.77(0.59–0.94)0.003

a Fold-change, 95% confidence interval and P-values were calculated using RT2 Profiler PCR Assay Data analysis v3.5 program.

KRAS mutation status had no significant effect on the OS time of the PDAC patients. KRAS wild-type patients experienced a median OS time of 22.3 months, and patients with KRAS mutation experienced a median OS time of 21.0 months (P=0.182).

There was also no association between KRAS mRNA transcript levels and OS time. In contrast to the rest of pathway, RAF1 showed a significant association with the OS time of the PDAC patients. Patients with RAF1 expression levels lower than the median experienced longer OS times than patients with higher RAF1 expression levels (P=0.030) (Fig. 4). However, this association did not pass Bonferroni correction for multiple testing.

Discussion

Mutation analysis of the present cohort of patients with operable PDAC aligns with that of prior studies reporting the presence of KRAS mutation in the majority of PDAC cases (18,19). Additionally, the genes of four KRAS downstream signaling pathways, including the PI3K/PDK1/AKT, RAL guanine nucleotide exchange factor, RIN1/ABL and RAF/MAPK pathways, exhibited differential expression in PDAC compared with that of the adjacent normal tissues, although no significant differences were observed in the expression of these genes between patients with KRAS-mutated and wild-type tumors. The expression profiles of KRAS downstream signaling pathways were not associated with pathological characteristics that reflect tumor biology, including angioinvasion, perineural invasion, grade or presence of lymph node metastasis.

Similar to earlier studies (2022), the present data indicated that in this cohort of patients (with early-stage disease and following radical surgery) the presence of a KRAS mutation had no effect on the OS time of the patients, although there was limited power to determine associations indicating more minor effects due to the limited size of the patient cohort. Moreover, with the exception of RAF1, no impact was observed of the expression profile of the KRAS downstream major effective signaling pathways on OS. These findings may explain why all previous efforts targeting KRAS failed to improve the patient outcome.

Despite sustained efforts in preclinical and clinical research, PDAC remains a malignancy with an almost uniformly fatal prognosis (23). In contrast to other solid tumors, there has been no major progress in the systemic therapy of PDAC during the last decade. In particular, there is currently no targeted agent with clinically significant activity against this tumor.

Although molecular biomarkers play a crucial role in the management of numerous solid tumors (24), there are currently no useful biomarkers for treatment selection in PDAC. In recent years, a number of negative trials of targeted therapy have been conducted in PDAC (25,26). Consequently, there is an urgent requirement to improve the understanding of PDAC pathogenesis and biology in order to identify novel therapeutic approaches and to define subgroups of patients for tailored therapies. It has been demonstrated that KRAS mutations represent the driver mutations in the majority of PDAC cases. KRAS-targeted agents can be classified into several categories according to the mechanism of action, namely small-molecule RAS-binding ligands, inhibitors of KRAS membrane anchorage, inhibitors that bind to RAS-binding domains of RAS-effector proteins and inhibitors of KRAS expression (27). However, attempts to therapeutically target KRAS or the downstream pathways have all thus far failed in clinical trials (2832).

In conclusion, as expected, KRAS was mutated in the majority of PDAC cases. The genes of the KRAS downstream signaling pathways, including the PI3K/PDK1/AKT, RAL guanine nucleotide exchange factor, RIN1/ABL and RAF/MAPK pathways, were differentially expressed in PDAC compared with those in adjacent non-neoplastic tissues. However, neither the presence of KRAS mutation nor the extent of KRAS signaling dysregulation was associated with OS time. Among the KRAS downstream signaling pathway genes investigated, only RAF1 expression was predictive of outcome. It is possible that the analysis of post-transcriptional and epigenetic factors associated with KRAS signaling may shed more light onto the molecular biology of PDAC.

Acknowledgements

This study was supported by projects of the Czech Science Foundation (grant no. P301/12/1734), the Ministry of Health of the Czech Republic (grant no. 16-28375A), the Ministry of Education Youth and Sports of the Czech Republic (no. LO1503) and the Czech Ministry of Education (nos. NPU I LO1304 and RVO 61989592).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, et al: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 364:1817–1825. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Morris JP IV, Wang SC and Hebrok M: KRAS, hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nat Rev Cancer. 10:683–695. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Pylayeva-Gupta Y, Grabocka E and Bar-Sagi D: RAS oncogenes: Weaving a tumorigenic web. Nat Rev Cancer. 11:761–774. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Eser S, Schnieke A, Schneider G and Saur D: Oncogenic KRAS signalling in pancreatic cancer. Br J Cancer. 111:817–822. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Eser S, Reiff N, Messer M, Seidler B, Gottschalk K, Dobler M, Hieber M, Arbeiter A, Klein S, Kong B, et al: Selective requirement of PI3K/PDK1 signaling for Kras oncogene-driven pancreatic cell plasticity and cancer. Cancer Cell. 23:406–420. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, Charles RP, Rabinovich BA, Hann B, Dankort D, et al: A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2:685–693. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Lim KH, Baines AT, Fiordalisi JJ, Shipitsin M, Feig LA, Cox AD, Der CJ and Counter CM: Activation of RalA is critical for Ras-induced tumorigenesis of human cells. Cancer Cell. 7:533–545. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Feldmann G, Mishra A, Hong SM, Bisht S, Strock CJ, Ball DW, Goggins M, Maitra A and Nelkin BD: Inhibiting the cyclin-dependent kinase CDK5 blocks pancreatic cancer formation and progression through the suppression of Ras-Ral signaling. Cancer Res. 70:4460–4469. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Dhaka A, Costa RM, Hu H, Irvin DK, Patel A, Kornblum HI, Silva AJ, O'Dell TJ and Colicelli J: The RAS effector RIN1 modulates the formation of aversive memories. J Neurosci. 23:748–757. 2003.PubMed/NCBI

11 

Mohelnikova-Duchonova B, Kocik M, Duchonova B, Brynychova V, Oliverius M, Hlavsa J, Honsova E, Mazanec J, Kala Z, Ojima I, et al: Hedgehog pathway overexpression in pancreatic cancer is abrogated by new-generation taxoid SB-T-1216. Pharmacogenomics J. Aug 30–2016.(Epub ahead of print). PubMed/NCBI

12 

Mohelnikova-Duchonova B, Brynychova V, Hlavac V, Kocik M, Oliverius M, Hlavsa J, Honsova E, Mazanec J, Kala Z, Melichar B and Soucek P: The association between the expression of solute carrier transporters and the prognosis of pancreatic cancer. Cancer Chemother Pharmacol. 72:669–682. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Mohelnikova-Duchonova B, Brynychova V, Oliverius M, Honsova E, Kala Z, Muckova K and Soucek P: Differences in transcript levels of ABC transporters between pancreatic adenocarcinoma and nonneoplastic tissues. Pancreas. 42:707–716. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Soucek P, Anzenbacher P, Skoumalova I and Dvorak M: Expression of cytochrome P450 genes in CD34+ hematopoietic stem and progenitor cells. Stem Cells. 23:1417–1422. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Mohelnikova-Duchonova B, Oliverius M, Honsova E and Soucek P: Evaluation of reference genes and normalization strategy for quantitative real-time PCR in human pancreatic carcinoma. Dis Markers. 32:203–210. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, Nolan T, Pfaffl MW, Shipley GL, et al: The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 55:611–622. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Caldas C, Hahn SA, Hruban RH, Redston MS, Yeo CJ and Kern SE: Detection of K-ras mutations inthe stool of patients with pancreatic adenocarcinoma and pancreatic ductal hyperplasia. Cancer Res. 54:3568–3573. 1994.PubMed/NCBI

19 

Bournet B, Buscail C, Muscari F, Cordelier P and Buscail L: Targeting KRAS for diagnosis, prognosis, and treatment of pancreatic cancer: Hopes and realities. Eur J Cancer. 54:75–83. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Zhou L, Baba Y, Kitano Y, Miyake K, Zhang X, Yamamura K, Kosumi K, Kaida T, Arima K, Taki K, et al: KRAS, BRAF, and PIK3CA mutations, and patient prognosis in 126 pancreatic cancers: Pyrosequencing technology and literature review. Med Oncol. 33:322016. View Article : Google Scholar : PubMed/NCBI

21 

Oliveira-Cunha M, Hadfield KD, Siriwardena AK and Newman W: EGFR and KRAS mutational analysis and their correlation to survival in pancreatic and periampullary cancer. Pancreas. 41:428–434. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Schultz NA, Roslind A, Christensen IJ, Horn T, Høgdall E, Pedersen LN, Kruhøffer M, Burcharth F, Wøjdemann M and Johansen JS: Frequencies and prognostic role of KRAS and BRAF mutations in patients with localized pancreatic and ampullary adenocarcinomas. Pancreas. 41:759–766. 2012.PubMed/NCBI

23 

Lemstrova R, Melichar B and Mohelnikova-Duchonova B: Therapeutic potential of taxanes in the treatment of metastatic pancreatic cancer. Cancer Chemother Pharmacol. 78:1101–1111. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Melichar B: Laboratory medicine and medical oncology: The tale of two Cinderellas. Clin Chem Lab Med. 51:99–112. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Fuchs CS, Azevedo S, Okusaka T, Van Laethem JL, Lipton LR, Riess H, Szczylik C, Moore MJ, Peeters M and Bodoky G: A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: The GAMMA trial. Ann Oncol. 26:921–927. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Deplanque G, Demarchi M, Hebbar M, Flynn P, Melichar B, Atkins J, Nowara E, Moyé L, Piquemal D, Ritter D, et al: A randomized, placebo-controlled phase III trial of masitinib plus gemcitabine in the treatment of advanced pancreatic cancer. Ann Oncol. 26:1194–1200. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Chuang HC, Huang PH, Kulp SK and Chen CS: Pharmacological strategies to target oncogenic KRAS signaling in pancreatic cancer. Pharmacol Res. 117:370–376. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Chung V, McDonough S, Philip PA, Cardin D, Wang-Gillam A, Hui L, Tejani MA, Seery TE, Dy IA, Al Baghdadi T, et al: Effect of Selumetinib and MK-2206 vs oxaliplatin and fluorouracil in patients with metastatic pancreatic cancer after prior therapy: SWOG S1115 study randomized clinical trial. JAMA Oncol. 3:516–522. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Riely GJ, Johnson ML, Medina C, Rizvi NA, Miller VA, Kris MG, Pietanza MC, Azzoli CG, Krug LM, Pao W and Ginsberg MS: A phase II trial of Salirasib in patients with lung adenocarcinomas with KRAS mutations. J Thoracic Oncol. 6:1435–1437. 2011. View Article : Google Scholar

30 

Karp JE, Vener TI, Raponi M, Ritchie EK, Smith BD, Gore SD, Morris LE, Feldman EJ, Greer JM, Malek S, et al: Multi-institutional phase 2 clinical and pharmacogenomic trial of tipifarnib plus etoposide for elderly adults with newly diagnosed acute myelogenous leukemia. Blood. 119:55–63. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Rao S, Cunningham D, de Gramont A, Scheithauer W, Smakal M, Humblet Y, Kourteva G, Iveson T, Andre T, Dostalova J, et al: Phase III double-blind placebo-controlled study of farnesyl transferase inhibitor R115777 in patients with refractory advanced colorectal cancer. J Clin Oncol. 22:3950–3957. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Van CE, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, Schoffski P, Post S, Verslype C, Neumann H, et al: Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 22:1430–1438. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lemstrova R, Brynychova V, Hughes DJ, Hlavac V, Dvorak P, Doherty JE, Murray HA, Crockard M, Oliverius M, Hlavsa J, Hlavsa J, et al: Dysregulation of KRAS signaling in pancreatic cancer is not associated with KRAS mutations and outcome. Oncol Lett 14: 5980-5988, 2017
APA
Lemstrova, R., Brynychova, V., Hughes, D.J., Hlavac, V., Dvorak, P., Doherty, J.E. ... Mohelnikova-Duchonova, B. (2017). Dysregulation of KRAS signaling in pancreatic cancer is not associated with KRAS mutations and outcome. Oncology Letters, 14, 5980-5988. https://doi.org/10.3892/ol.2017.6946
MLA
Lemstrova, R., Brynychova, V., Hughes, D. J., Hlavac, V., Dvorak, P., Doherty, J. E., Murray, H. A., Crockard, M., Oliverius, M., Hlavsa, J., Honsova, E., Mazanec, J., Kala, Z., Lovecek, M., Havlik, R., Ehrmann, J., Strouhal, O., Soucek, P., Melichar, B., Mohelnikova-Duchonova, B."Dysregulation of KRAS signaling in pancreatic cancer is not associated with KRAS mutations and outcome". Oncology Letters 14.5 (2017): 5980-5988.
Chicago
Lemstrova, R., Brynychova, V., Hughes, D. J., Hlavac, V., Dvorak, P., Doherty, J. E., Murray, H. A., Crockard, M., Oliverius, M., Hlavsa, J., Honsova, E., Mazanec, J., Kala, Z., Lovecek, M., Havlik, R., Ehrmann, J., Strouhal, O., Soucek, P., Melichar, B., Mohelnikova-Duchonova, B."Dysregulation of KRAS signaling in pancreatic cancer is not associated with KRAS mutations and outcome". Oncology Letters 14, no. 5 (2017): 5980-5988. https://doi.org/10.3892/ol.2017.6946