Skip to main content
Intended for healthcare professionals
Free access
Research article
First published online February 6, 2013

ERK1/2 activity contributes to gemcitabine resistance in pancreatic cancer cells

Abstract

Objective

To test the hypothesis that chemoresistance in pancreatic cancer is mediated via extracellular signal-regulated protein kinase (ERK) 1/2 overactivity.

Methods

The human pancreatic cancer cell lines BxPC3, PANC-1 and a stably gemcitabine-resistant subline, PANC1GemRes, were treated with combinations of gemcitabine and the ERK1/2 inhibitor, U0126. Phosphorylated (p)ERK1/2 was examined by Western blotting; cell proliferation and apoptosis were quantified. A nude mouse xenograft model was established with each cell line, and the therapeutic efficacy of gemcitabine and U0126 alone or in combination was examined.

Results

Gemcitabine treatment visibly increased pERK1/2 levels in BxPC-3 and PANC-1 cells. PANC-1GemRes constitutively produced high levels of pERK1/2. U0126 treatment reversed the gemcitabine-associated increase in cell proliferation and reduction in apoptosis, in all three cell lines. Combination treatment with U0126 and gemcitabine inhibited tumour growth and promoted apoptosis in xenograft tumours derived from all three cell lines.

Conclusions

ERK1/2 activity may protect pancreatic cancer cells from chemotherapy-induced apoptosis. The combined use of an ERK1/2 inhibitor (such as U0126) together with gemcitabine may result in synergistic therapeutic effects at tolerable gemcitabine doses.

Introduction

Pancreatic cancer has the worst prognosis of all major cancers, with a 5-year overall survival rate of ∼5%.1 The current standard treatment for advanced pancreatic cancer is gemcitabine, which is a cytotoxic nucleoside analogue. The benefits of gemcitabine are modest, with a tumour response rate of 12% and a median survival time of 5 months.2 Data from a small number of studies suggest that a combination of targeted therapy and gemcitabine has statistically significant benefits.3 The extracellular signal-regulated protein kinases (ERK) 1 and 2 are potential targets for combination therapy.4
The ERK1/2 signalling pathway is involved in the control of diverse cellular processes such as proliferation, survival, differentiation and motility.4 This pathway is often upregulated in human tumours5,6 and represents an attractive target for the development of anticancer drugs.710 The inhibitor of mitogen-activated protein kinase kinase 1/2 (MAP2K1/2), U0126, has been shown to inhibit ERK1/2 phosphorylation effectively,11 and to enhance radiosensitivity of rhabdomyosarcoma cells.12 ERK1/2 has been shown to be activated in pancreatic carcinoma,5 but little is known regarding its role in chemoresistance.
This study investigated the effects of ERK1/2 inhibition by U0126 and gemcitabine, alone and in combination, on a pancreatic carcinoma model in vitro and in vivo.

Materials and methods

Cell culture

Human pancreatic adenocarcinoma PANC-1 and BxPC-3 cells were obtained from American Type Culture Collection (Shanghai, China) and maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum (Life Technologies, Gaithersburg, MD). Cells were incubated in 75-cm2 culture flasks in a humidified atmosphere at 37°C with 5% carbon dioxide in air, and passaged on reaching 60–80% confluence.

Development of PANC-1GemRes

The pancreatic adenocarcinoma cell line PANC-1 was used to develop a gemcitabine-resistant cell line, PANC-1GemRes, as described previously.13 Briefly, PANC-1 cells were serially subcultured through incrementally increasing gemcitabine concentrations, starting at the 50% inhibitory drug concentration (IC50), over 3 months. Persistence of stable resistance was confirmed by subculture in gemcitabine-free DMEM containing 10% fetal bovine serum; PANC-1GemRes retained the capacity for proliferation when returned to medium containing 5 µM gemcitabine.

Determination of gemcitabine IC50

The IC50 of gemcitabine was determined in each cell line. BxPC-3, PANC-1 and PANC-1GemRes cells were seeded at 1 × 104 per well in 96-well plates, in 0.1 ml of culture medium and cultured overnight. The next day, the medium was supplemented with 0–100 µM gemcitabine (Sigma, St Louis, MO, USA) for 48 h at 37°C. The IC50 was determined using 2,3-bis(2-methoxy-4-nitro-5-sulphophenyl)-2H-tetrazolium-5-carboxanilide (XTT) assay (Roche Molecular Biochemicals, Indianapolis, IN, USA) according to the manufacturer’s instructions. Cell proliferation was quantified by measuring the absorbance at 450 nm, and the IC50 for each of the cell lines was estimated from the semilogarithmic dose–response curves, by linear interpolation.

Cell proliferation and apoptosis analysis

Cells (BxPC-3, PANC-1 or PANC-1GemRes) were seeded at 1 × 104 per well in 96-well plates in 0.1 ml of culture medium and allowed to grow overnight. The next day, the medium was supplemented with IC50 gemcitabine for 48 h at 37°C in the presence or absence of 25 µM U0126 (Sigma, St Louis, MO, USA) (added 1 h before gemcitabine), or treated with 25 µM U0126 alone. Cell proliferation was quantified by XTT assay as described above. Parallel cultures were immediately fixed in 37% formaldehyde at room temperature for 30 min and allowed to dry overnight. Apoptosis was determined using the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labelling (TUNEL) method (Roche in situ Apoptosis Detection System; Roche, Mannheim, Germany), according to the manufacturer's instructions. Cells were examined by light microscopy; apoptosis was quantified as the mean number of stained cells as a percentage of total cells in five random high-power fields (×100 magnification).

Western blotting

Cells (BxPC-3, PANC-1 or PANC-1GemRes) were seeded at 1 × 104 per well in 96-well plates, in 0.1 ml of culture medium, and allowed to grow overnight. The next day, the medium was supplemented with IC50 gemcitabine for 48 h at 37°C. Cells were rinsed once with cold phosphate buffered saline (PBS; 20 mM, pH 7.6) and lysed in radioimmunoprecipitation assay buffer (50 mM Tris-HCl, pH 8.0, 150 mM sodium chloride, 1% nonyl phenoxypolyethoxylethanol, 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulphate [SDS]), containing 0.3 mg/l protease inhibitor (Roche), for 15 min on ice. Cell lysates were centrifuged at 28 340 g for 15 min, and 25 µg of protein for each sample was resolved by 10% SDS–polyacrylamide gel electrophoresis under reduced conditions. Proteins were transferred to nitrocellulose membranes (Millipore, Billerica, MA, USA) and blocked with TBST (10 mM Tris-HCl, pH 7.4, 100 mM sodium chloride, 0.1% Tween-20) containing 5% skim milk for 30 min at 4°C. Membranes were incubated with mouse antihuman phosphorylated (p)ERK1/2 monoclonal antibody (1 : 200 dilution) or mouse antihuman β-actin monoclonal antibody (1 : 500 dilution; both Santa Cruz Biotechnology; Santa Cruz, CA, USA) overnight at 4°C. Membranes were washed four times with TBST at room temperature for 5 min each wash, then incubated with horseradish peroxidase-conjugated rabbit antimouse immunoglobulin G (1 : 5000 dilution; Zymed, San Francisco, CA, USA) for 60 min at room temperature. Membranes were washed four times with TBST at room temperature (5 min each wash) and immunoreactive protein bands were visualized using enhanced chemiluminescence (ECL detection system; Pierce, Rockford, IL, USA), followed by exposure to AGFA X-ray blue film (AGFA-Gevaert, Hortsel, Belgium).

Mouse tumour model

Male athymic nu/nu mice (aged 5 weeks; weight 18–20 g; specific pathogen free) were obtained from the Laboratory Centre of Shandong University. Mice were housed in microisolator cages, with autoclaved bedding in a specific pathogen-free facility, with a 12-h light/dark cycle. Animals received water and food ad libitum and were observed for signs of tumour growth, activity, feeding and pain, in accordance with the guidelines of the Standing Committee on Animals, Shandong University. The study protocol was approved by the Ethics Committee of Jinan Central Hospital of Shandong University.
Xenograft tumours were established by subcutaneous injection of 1 × 106 BxPC-3, PANC-1 or PANC1GemRes cells in 20 µl of PBS (pH 7.6). When tumours were 50–100 mm3 in diameter (after 4 weeks) the mice were divided into four groups (n = 8 per group), to receive the following treatments by intraperitoneal injection, three times per week for 4 weeks (all injections were of identical volume): 250 µg/kg U0126 (in dimethyl sulphoxide; DMSO); 100 mg/kg gemcitabine (in 1% DMSO); 250 µg/kg U0126 and 100 mg/kg gemcitabine (in DMSO); and control group (1% DMSO). Tumour growth was monitored with calipers three times per week during the treatment period; tumour volume was calculated according to the formula (length × width2)/2. On completion of treatment, mice were sacrificed and the tumours were excised.
Ttissue samples were immediately snap-frozen in liquid nitrogen and stored at −80°C until used for analysis. Apoptosis was quantified in frozen sections of tumour tissue (4-µm thick) using TUNEL assay (Roche in situ Apoptosis Detection System), according to the manufacturer’s instructions, as previously described.14 Sections were examined by light microscopy and the apoptosis index was determined by counting ≥1000 cells in 5 randomly selected high-power fields (×200 magnification).

Statistical analyses

All experiments were performed a minimum of three times. Data were presented as mean ± SD and compared using Student’s t-test. A P-value <0.05 was considered to be statistically significant. Statistical analyses were performed using SPSS® software, version 11.0 (SPSS Inc., Chicago, IL, USA) for Windows®.

Results

Data regarding the gemcitabine IC50 for PANC-1, BxPC-3 and PANC-1GemRes cells are shown in Table 1. The gemcitabine IC50 of PANC-1GemRes cells was significantly greater than that of the parental cell line, PANC-1 (P < 0.05). Treatment with U0126 significantly reduced the gemcitabine IC50 in PANC-1 and PANC-1GemRes cells (P < 0.05; Table 1), but had no effect in BxPC-3 cells.
Table 1. Effect of the extracellular signal-regulated protein kinase 1/2 inhibitor U0126 on the 50% inhibitory drug concentration (IC50) of gemcitabine in the human pancreatic adenocarcinoma cell lines BxPC-3 and PANC-1, and in a gemcitabine-resistant PANC-1 derived cell line, PANC-1GemRes.
Cell line Gemcitabine Gemcitabine  + U0126
BxPC-3 14.0 ± 2.1 nM 7.9 ± 1.2 nM
PANC-1 49.6 ± 5.4 nM 19.7 ± 4.3 nMb
PANC-1GemRes 12.4 ± 1.8 µMa 2.4 ± 0.5 µMb
Data presented as mean ± SD.
a
P < 0.05 vs PANC-1 cells, bP < 0.05 vs gemcitabine alone; Student’s t-test.
Western blot analysis of pERK1/2 in PANC-1, BxPC-3 and PANC-1GemRes cells, after treatment with gemcitabine, is presented in Figure 1. BxPC-3 cells appeared to have no constitutive pERK1/2 (absence of protein at 0 h). Gemcitabine treatment resulted in visibly increased pERK1/2 levels in BxPC-3 and PANC-1 cells at 6 and 12 h after treatment, respectively. PANC-1GemRes cells constitutively produced relatively high levels of pERK1/2 that were unchanged by gemcitabine treatment.
Figure 1. Western blot analysis of phosphorylated signal regulated extracellular kinase (pERK)1/2 and β-actin in human pancreatic adenocarcinoma cell lines BxPC-3 and PANC-1, and a gemcitabine-resistant PANC-1 derived cell line, PANC-1GemRes. Cells were treated with their 50% inhibitory concentration of gemcitabine (14 nM, 49.6 nM and 12.4 µM, respectively) for 48 h.
Treatment with U0126 alone had no effect on apoptosis in PANC-1, BxPC-3 or PANC-1GemRes cells. Combined treatment with U0126 and gemcitabine resulted in significantly higher levels of apoptosis than gemcitabine alone, in all three cell lines (P < 0.01 for each cell line; Table 2)
Table 2. Effect of treatment with U0126 and/or gemcitabine on apoptosisa in the human pancreatic adenocarcinoma cell lines BxPC-3 and PANC-1, and a gemcitabine-resistant PANC-1 derived cell line, PANC-1GemRes.
Treatment Cell line
BxPC-3 PANC-1 PANC-1GemRes
Control 1.12 ± 0.02 1.04 ± 0.03 1.16 ± 0.02
U0126 1.14 ± 0.02 1.13 ± 0.03 1.18 ± 0.04
Gemcitabine 13.5 ± 1.46* 5.14 ± 1.05 1.23 ± 0.06
U0126 + gemcitabine 37.4 ± 6.3*** 17.3 ± 2.54** 19.32 ± 2.46**
Data presented as mean ± SD.
a
mean percentage of apoptotic cells as a proportion of total cells in five random high power fields (× 100 magnification).
*
P < 0.05, **P < 0.01, ***P < 0.001 versus control; Student’s t-test.
All mice survived the treatment period. Tumours derived from all three cell lines were significantly smaller following combination U0126 and gemcitabine treatment compared with the tumours observed in control animals (P < 0.05 for each cell line; Table 3). Of the three cell lines, only BxPC-3-derived tumours were significantly smaller than controls, following gemcitabine treatment alone (P < 0.05; Table 3). U0126 treatment had no effect on the growth of tumours derived from any cell line.
Table 3. Effect of treatment with U0126 and/or gemcitabine on tumour volume and apoptosis in mouse xenograft models of pancreatic carcinoma, derived from human pancreatic adenocarcinoma cell lines BxPC-3 and PANC-1, and a gemcitabine-resistant PANC-1 derived cell line, PANC-1GemRes.
Parameter Cell line
BxPC-3 PANC-1 PANC-1GemRes
Tumour volume, mm3      
 Control 2540 ± 250 2350 ± 280 2120 ± 320
 U0126 2430 ± 235 2286 ± 315 2216 ± 280
 Gemcitabine 1760 ± 85* 2032 ± 178 2080 ± 260
 U0126 + gemcitabine 210 ± 35** 1100 ± 65* 1650 ± 120*
Apoptosisa      
 Control 1.02 ± 0.08 1.83 ± 0.12 1.14 ± 0.12
 U0126 1.08 ± 0.07 1.92 ± 0.12 1.24 ± 0.13
 Gemcitabine 4.46 ± 0.13 2.17 ± 0.15 2.15 ± 0.13
 U0126 + gemcitabine 14.30 ± 1.16*** 11.20 ± 2.30*** 9.70 ± 2.54**
Data presented as mean ± SD.
a
mean percentage of apoptotic cells as a proportion of total cells in five random high power fields (×200 magnification).
*
P < 0.05, **P < 0.01, ***P < 0.001 versus control; Student’s t-test.
In vivo, combination treatment with U0126 and gemcitabine resulted in significantly increased apoptosis compared with controls, in tumours derived from all three cell lines (P < 0.001 for each comparison; Table 3). Neither U0126 nor gemcitabine treatment alone had any effect on apoptosis in vivo.

Discussion

Human pancreatic cancer is characterized by high intrinsic (de novo) and acquired (therapy induced) resistance to traditional chemo- and radiation therapy.15 Understanding the underlying mechanisms of drug resistance in pancreatic cancer is critical in order for new and effective treatments to be developed for this deadly disease. Gemcitabine is the standard chemotherapeutic agent for pancreatic cancer patients, but its clinical impact is limited by the high degree of inherent and acquired resistance.15
Studies have found ERK1/2 to be involved in tumour chemoresistance,16,17 and increased ERK1/2 activity has been shown to correlate with inherent gemcitabine resistance in intrahepatic cholangiocarcinoma cells.18 Levels of pERK1/2 appeared to be higher in PANC-1GemRes cells than in the gemcitabine-sensitive PANC-1 parental cell line in the present study, suggesting that gemcitabine resistance may be mediated via pERK1/2.
Inhibition of ERK1/2 activity by U0126 promoted gemcitabine-induced apoptosis in human pancreatic adenocarcinoma cell lines and reversed acquired gemcitabine resistance in PANC1GemRes in the present study. BxPC-3 cells, which were shown not to produce pERK1/2 constitutively in the present study, were sensitive to gemcitabine-induced apoptosis in vitro. These data suggest that inherent ERK1/2 activity may play an important role in gemcitabine resistance.
It has been suggested that the ERK1/2 pathway mediates apoptosis induced by various stimuli, including cisplatin.19 This is supported by the finding that Nic-T24 cells (which have low ERK1/2 activity) tend to remain in G0/G1 phase rather than undergo apoptosis, in response to antitumour agents.17
In conclusion, the findings of the present study suggest that high ERK1/2 activity protects tumour cells from chemotherapy-induced apoptosis, in pancreatic cancer. The combined use of an ERK1/2 inhibitor (such as U0126) and gemcitabine may result in synergistic therapeutic effects at tolerable gemcitabine doses. Further studies are required to elucidate the mechanism by which ERK1/2 inhibition enhances chemotherapeutic effects. The significance of pERK1/2 as a therapeutic marker in clinical specimens remains to be investigated.

Declaration of conflicting interest

The Authors declare that there are no conflicts of interest.

Funding

This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

References

1. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006. CA Cancer J Clin 2006; 56: 106–130.
2. O’Reilly EM, Abou-Alfa GK. Cytotoxic therapy for advanced pancreatic adenocarcinoma. Semin Oncol 2007; 34: 347–353.
3. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2007; 25: 1960–1966.
4. Kohno M, Pouyssegur J. Targeting the ERK signaling pathway in cancer therapy. Ann Med 2006; 38: 200–211.
5. Chadha KS, Khoury T, Yu J, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol 2006; 13: 933–939.
6. Vicent S, López-Picazo JM, Toledo G, et al. ERK1/2 is activated in non-small-cell lung cancer and associated with advanced tumours. Br J Cancer 2004; 90: 1047–1052.
7. Yang JY, Zong CS, Xia W, et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat Cell Biol 2008; 10: 138–148.
8. Zhou L, Luan H, Liu Q, et al. Activation of PI3K/Akt and ERK signaling pathways antagonized sinomenine-induced lung cancer cell apoptosis. Mol Med Report 2012; 5: 1256–1260.
9. Nguyen TK, Jordan N, Friedberg J, et al. Inhibition of MEK/ERK1/2 sensitizes lymphoma cells to sorafenib-induced apoptosis. Leuk Res 2010; 34: 379–386.
10. Tsai MS, Kuo YH, Chiu YF, et al. Down-regulation of Rad51 expression overcomes drug resistance to gemcitabine in human non-small-cell lung cancer cells. J Pharmacol Exp Ther 2010; 335: 830–840.
11. Hawkins TA, Cavodeassi F, Erdélyi F, et al. The small molecule Mek1/2 inhibitor U0126 disrupts the chordamesoderm to notochord transition in zebrafish. BMC Dev Biol 2008; 8: 42–42.
12. Marampon F, Gravina GL, Di Rocco A, et al. MEK/ERK inhibitor U0126 increases the radiosensitivity of rhabdomyosarcoma cells in vitro and in vivo by downregulating growth and DNA repair signals. Mol Cancer Ther 2011; 10: 159–168.
13. Goan YG, Zhou B, Hu E, et al. Overexpression of ribonucleotide reductase as a mechanism of resistance to 2,2-difluorodeoxycytidine in the human KB cancer cell line. Cancer Res 1999; 59: 4204–4207.
14. Wang Z, Zhou J, Fan J, et al. Effect of rapamycin alone and in combination with sorafenib in an orthotopic model of human hepatocellular carcinoma. Clin Cancer Res 2008; 14: 5124–5130.
15. Duxbury MS, Ito H, Zinner MJ, et al. Inhibition of Src tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res 2004; 10: 2307–2318.
16. Jeong EK, Lee SY, Jeon HM, et al. Role of extracellular signal-regulated kinase (ERK)1/2 in multicellular resistance to docetaxel in MCF-7 cells. Int J Oncol 2010; 37: 655–661.
17. Mirmohammadsadegh A, Mota R, Gustrau A, et al. ERK1/2 is highly phosphorylated in melanoma metastases and protects melanoma cells from cisplatin-mediated apoptosis. J Invest Dermatol 2007; 127: 2207–2215.
18. Yoon H, Min JK, Lee JW, et al. Acquisition of chemoresistance in intrahepatic cholangiocarcinoma cells by activation of AKT and extracellular signal-regulated kinase (ERK)1/2. Biochem Biophys Res Commun 2011; 405: 333–337.
19. Wang X, Martindale JL, Holbrook NJ. Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem 2000; 275: 39435–39443.

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: February 6, 2013
Issue published: April 2013

Keywords

  1. Chemoresistance
  2. extracellular signal-regulated protein kinase
  3. ERK1/2
  4. gemcitabine
  5. pancreatic cancer

Rights and permissions

© The Author(s) 2013 Reprints and permissions: sagepub.co.uk/journalsPermissions.nav.
Request permissions for this article.
PubMed: 23569008

Authors

Affiliations

Chunning Zheng
Department of General Surgery, Jinan Central Hospital of Shandong University, Jinan, China
Xuelong Jiao
The Affiliated Hospital of the Medical College, Qingdao University, Qingdao, China
Yingsheng Jiang
Department of General Surgery, Jinan Central Hospital of Shandong University, Jinan, China
Shaochuan Sun
Department of General Surgery, Jinan Central Hospital of Shandong University, Jinan, China

Notes

Dr Shaochuan Sun, Department of General Surgery, Jinan Central Hospital of Shandong University, 105 Jiefang Road, Lixia District, Jinan 250013, China. Email: [email protected]

Metrics and citations

Metrics

Journals metrics

This article was published in Journal of International Medical Research.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 1686

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 51 view articles Opens in new tab

Crossref: 46

  1. Gemcitabine elaidate and ONC201 combination therapy for inhibiting pan...
    Go to citation Crossref Google Scholar
  2. Pancreatic stellate cell-induced gemcitabine resistance in pancreatic ...
    Go to citation Crossref Google Scholar
  3. Novel strategy for oncogenic alteration-induced lipid metabolism repro...
    Go to citation Crossref Google Scholar
  4. Chemomodulatory Effect of the Marine-Derived Metabolite “Terrein” on t...
    Go to citation Crossref Google Scholar
  5. Pan-KRAS inhibitors suppress proliferation through feedback regulation...
    Go to citation Crossref Google Scholar
  6. Exploration of the System-Level Mechanisms of the Herbal Drug FDY003 f...
    Go to citation Crossref Google Scholar
  7. Calcium channel blockers potentiate gemcitabine chemotherapy  in pancr...
    Go to citation Crossref Google Scholar
  8. An In Vitro Investigation into Cryoablation and Adjunctive Cryoablatio...
    Go to citation Crossref Google Scholar
  9. Pyruvate Dehydrogenase Contributes to Drug Resistance of Lung Cancer C...
    Go to citation Crossref Google Scholar
  10. A network pharmacology approach to reveal the pharmacological targets ...
    Go to citation Crossref Google Scholar
  11. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma an...
    Go to citation Crossref Google Scholar
  12. Targeting KRAS in Solid Tumors: Current Challenges and Future Opportun...
    Go to citation Crossref Google Scholar
  13. Inhibition of the Receptor for Advanced Glycation End Products Enhance...
    Go to citation Crossref Google Scholar
  14. pH-Sensitive Nanodrug Carriers for Codelivery of ERK Inhibitor and Gem...
    Go to citation Crossref Google Scholar
  15. The Ras-ERK1/2 signaling pathway regulates H3K9ac through PCAF to prom...
    Go to citation Crossref Google Scholar
  16. (3E,5E)-3,5-Bis(pyridin-3-methylene)-tetrahydrothiopyran-4-one enhance...
    Go to citation Crossref Google Scholar
  17. Nano-delivery of Gemcitabine Derivative as a Therapeutic Strategy in a...
    Go to citation Crossref Google Scholar
  18. Anti-Cancer Potential of Oxialis obtriangulata in Pancreatic Cancer Ce...
    Go to citation Crossref Google Scholar
  19. Can Hemp Help? Low-THC Cannabis and Non-THC Cannabinoids for the Treat...
    Go to citation Crossref Google Scholar
  20. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pa...
    Go to citation Crossref Google Scholar
  21. Secretion of fibronectin by human pancreatic stellate cells promotes c...
    Go to citation Crossref Google Scholar
  22. Targeting Autophagy by MPT0L145, a Highly Potent PIK3C3 Inhibitor, Pro...
    Go to citation Crossref Google Scholar
  23. EMT Contributes to Chemoresistance in Pancreatic Cancer
    Go to citation Crossref Google Scholar
  24. GPR55 signalling promotes proliferation of pancreatic cancer cells and...
    Go to citation Crossref Google Scholar
  25. Mitogen‑activated protein kinase inhibition enhances the antitumor eff...
    Go to citation Crossref Google Scholar
  26. Molecular and cellular mechanisms of chemoresistance in pancreatic can...
    Go to citation Crossref Google Scholar
  27. The Role of Platelet-Derived ADP and ATP in Promoting Pancreatic Cance...
    Go to citation Crossref Google Scholar
  28. Uncoupling the Oncogenic Engine
    Go to citation Crossref Google Scholar
  29. Inhibition of cancer migration and invasion by knocking down delta-5-d...
    Go to citation Crossref Google Scholar
  30. Engineered Resistant-Starch (ERS) Diet Shapes Colon Microbiota Profile...
    Go to citation Crossref Google Scholar
  31. The cornerstone K-RAS mutation in pancreatic adenocarcinoma: From cell...
    Go to citation Crossref Google Scholar
  32. Low-dose gemcitabine induces major histocompatibility complex class I-...
    Go to citation Crossref Google Scholar
  33. Forskolin sensitizes pancreatic cancer cells to gemcitabine via Stat3 ...
    Go to citation Crossref Google Scholar
  34. Epithelial-mesenchymal transition as a therapeutic target for overcomi...
    Go to citation Crossref Google Scholar
  35. Knockdown delta-5-desaturase promotes the formation of a novel free ra...
    Go to citation Crossref Google Scholar
  36. Paeoniflorin inhibits human pancreatic cancer cell apoptosis via suppr...
    Go to citation Crossref Google Scholar
  37. Hypoxia regulates ABCG 2 activity through ...
    Go to citation Crossref Google Scholar
  38. Blockade of dual-specificity phosphatase 28 decreases chemo-resistance...
    Go to citation Crossref Google Scholar
  39. Moxifloxacin and ciprofloxacin induces S-phase arrest and augments apo...
    Go to citation Crossref Google Scholar
  40. Downregulation of ASPP2 in pancreatic cancer cells contributes to incr...
    Go to citation Crossref Google Scholar
  41. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitab...
    Go to citation Crossref Google Scholar
  42. Salmonella -Based Therapy Targeting Indoleamine 2,3-Diox...
    Go to citation Crossref Google Scholar
  43. RLN2 Is a Positive Regulator of AKT-2-Induced Gene Expression Required...
    Go to citation Crossref Google Scholar
  44. Arginine deiminase augments the chemosensitivity of argininosuccinate ...
    Go to citation Crossref Google Scholar
  45. Curcumin as a cancer chemotherapy sensitizing agent
    Go to citation Crossref Google Scholar
  46. MicroRNA187 overexpression is related to tumor progression and determi...
    Go to citation Crossref Google Scholar

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:


Alternatively, view purchase options below:

Purchase 24 hour online access to view and download content.

Access journal content via a DeepDyve subscription or find out more about this option.