Next Article in Journal
Clinical Translatability of “Identified” Circulating miRNAs for Diagnosing Breast Cancer: Overview and Update
Next Article in Special Issue
Urinary Metabolic Signatures Detect Recurrences in Non-Muscle Invasive Bladder Cancer
Previous Article in Journal
Store-Operated Ca2+ Entry in Tumor Progression: From Molecular Mechanisms to Clinical Implications
Previous Article in Special Issue
Plasma microRNA Levels Combined with CEA and CA19-9 in the Follow-Up of Colorectal Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

miRNA Predictors of Pancreatic Cancer Chemotherapeutic Response: A Systematic Review and Meta-Analysis

by
Madhav Madurantakam Royam
1,
Rithika Ramesh
1,
Ritika Shanker
1,
Shanthi Sabarimurugan
1,
Chellan Kumarasamy
2,
Nachimuthu Ramesh
1,
Kodiveri Muthukalianan Gothandam
1,
Siddharta Baxi
3,
Ajay Gupta
4,
Sunil Krishnan
5 and
Rama Jayaraj
6,*
1
School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
2
University of Adelaide, Adelaide, SA 5005, Australia
3
Genesis Cancer Care Centre, Bunbury, WA 6014, Australia
4
National Heart Institute, New Delhi 110065, India
5
Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
6
College of Health and Human Sciences, Yellow 1.1.05, Ellengowan Drive, Charles Darwin University, Darwin, NT 0909, Australia
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(7), 900; https://doi.org/10.3390/cancers11070900
Submission received: 7 May 2019 / Revised: 13 June 2019 / Accepted: 21 June 2019 / Published: 27 June 2019
(This article belongs to the Special Issue New Biomarkers in Cancers)

Abstract

:
Background: pancreatic cancer (PC) has increasing incidence and mortality in developing countries, and drug resistance is a significant hindrance to the efficacy of successful treatment. The objective of this systematic review and meta-analysis was to evaluate the association between miRNAs and response to chemotherapy in pancreatic cancer patients. Methods: the systematic review and meta-analysis was based on articles collected from a thorough search of PubMed and Science Direct databases for publications spanning from January 2008 to December 2018. The articles were screened via a set of inclusion and exclusion criteria based on the preferred reporting items for systematic review and meta-analysis (PRISMA) guidelines. Data was extracted, collated and tabulated in MS Excel for further synthesis. Hazard ratio (HR) was selected as the effect size metric to be pooled across studies for the meta-analysis, with the random effects model being applied. Subgroup analysis was also conducted, and the presence of publication bias in the selected studies was assessed. Publication bias of the included studies was quantified. Findings: of the 169 articles screened, 43 studies were included in our systematic review and 13 articles were included in the meta-analysis. Gemcitabine was observed to be the principal drug used in a majority of the studies. A total of 48 miRNAs have been studied, and 18 were observed to have possible contributions to chemoresistance, while 15 were observed to have possible contributions to chemosensitivity. 41 drug-related genetic pathways have been identified, through which the highlighted miRNA may be affecting chemosensitivity/resistance. The pooled HR value for overall survival was 1.603; (95% Confidence Interval (CI) 1.2–2.143; p-value: 0.01), with the subgroup analysis for miR-21 showing HR for resistance of 2.061; 95% CI 1.195–3.556; p-value: 0.09. Interpretation: our results highlight multiple miRNAs that have possible associations with modulation of chemotherapy response in pancreatic cancer patients. Further studies are needed to discover the molecular mechanisms underlying these associations before they can be suggested for use as biomarkers of response to chemotherapeutic interventions in pancreatic cancer.

1. Introduction

The GLOBOCAN 2018 data estimates that 18.1 million new cancer cases and 9.6 million cancer deaths occur worldwide. Pancreatic cancer (PC) accounts for 2.5% of the new cases and 4.5% of the overall death [1]. PC is the 12th most prevalent malignancy throughout the globe with 338,000 new cases recognised in 2012, and the five-year average prevalence rate was found to be 4.1 in 100,000 people throughout the world [2]. It has one of the lowest survival rates among the predominant tumours with a single digit five-year survival rate (2–9%) [3]. According to GLOBOCAN 2015 data, 1000 cases of PC are diagnosed daily [4], and 985 deaths [5] occur worldwide daily. With regards to India, cancer is one of the leading causing of death behind cardiovascular disease [6]. According to a study, the estimated annual PC burden in India in 2001 was 14,230 cases.
The mainstay of non-metastatic PC treatment is surgery [7]. Surgery is the most useful local treatment, but despite this, survival rates are modest primarily due to problems with distant and local recurrence issues. Therapeutic agents that are utilised as adjuvant or neo-adjuvant therapies are radiotherapy and systemic therapies (chemotherapies, targeted therapies). These include 5-Fluorouracil (5-FU) [8], 5-FU-based combinations like doxorubicin and mitomycin (FAM) [9], docetaxel [10], irinotecan [11], Gemcitabine (GEM) [12], irinotecan and oxaliplatin (FOLFIRINOX), GEM-based combination therapies such as 5-FU [13], capecitabine [14], S-1 [15], cisplatin [16], irinotecan [17], cisplatin, epirubicin and 5-FU (PEFG) [18], oxaliplatin [19], erlotinib [20] and nab-paclitaxel [21]. Other management options include nanocarriers [22], palliative care [23], immunotherapy [24].
MicroRNAs (miRNAs) are short (20–25 nucleotide) non-coding sequences increasingly recognised as molecular markers for the early detection of cancer and prognostication of clinical outcomes [25,26,27,28,29]. miRNAs regulate cell proliferating genes such as c-Myc and E2F1 thereby playing a critical role in cancer progression [30,31]. Differential expression of apoptosis-regulating genes was found to be associated with the 5-FU and GEM resistance in PC cell lines [32]. A tumour suppressing the role of miRNA was illustrated by the miR-96 miRNA which acts by suppressing KRAS gene in PC both in vitro and in vivo [33]. miRNA profiling by real-time PCR in PC cell lines and patient PC tissue samples revealed a range of 95 miRNAs that were altered [34]. Li Y et al. (2009) demonstrated that miRNA 200 and let-7a lead to the reversal of the epithelial-mesenchymal transition (EMT) in GEM-resistant pancreatic cancer cells [35]. A study by Zhu et al. observed that miRNA 27a and 451 were upregulated in multidrug-resistant cell lines (A2780DX5 and KB-V1) along with the overexpression of P-glycoprotein (P-gp) [36]. In the case of resistance due to the upregulation of miRNA-451, the transfection of miRNA-451 in doxorubicin-resistant MCF-7 cell lines increased the sensitivity to doxorubicin significantly [37].
Drug resistance is classified as intrinsic if it is present before treatment and acquired resistance if it develops while on treatment. There are several reviews involving miRNA and chemotherapy resistance [38,39,40]. Ali S et al. (2010) induced GEM sensitivity in PC cells through modulation of miR-200 and 21 by curcumin [41]. Reversal of EMT was achieved through natural agents (3,3′-diindolylmethane (DIM) or isoflavone) by upregulating miR-200 and let-7a in GEM-resistant PC cell lines [35]. The Notch signalling pathway is linked with the acquisition of EMT in GEM-resistant PC cell lines [42]. CD44-positive cancer stem cells (CSCs) were recently noted to be responsible for GEM-resistance in PC [43].
An increasing number of publications in recent years correlate miRNA expression in PC with resistance or sensitivity towards chemotherapeutic targets. Currently, the data on the correlations between PC chemoresistance/sensitivity and miRNA expression has not yielded clinically relevant solutions, in the form of prognostic biomarkers, despite the ongoing research in this field. A systematic review and meta-analysis approach allows us to collate the data across all published studies in the field and possibly highlight the associated miRNA, which may have clinical relevance in directing decisions regarding chemotherapy in PC patients [44,45,46,47]. Our study would help prospective scholars and clinicians by cataloguing miRNA alterations associated with chemotherapeutic response in this deadly disease. Future studies can then define their utility as predictors of chemotherapy response.
Our study aims to elucidate the relationship between miRNA expression and chemotherapeutic resistance or susceptibility in PC through a systematic review and meta-analysis of extant literature.

2. Methods

2.1. Search Strategy and Study Selection

Current studies in PC involving miRNA related drug resistance were identified through PubMed and Science Direct using the search terms; “microRNAs or miRNAs” AND “drug resistance” AND “PC” in combination. Four authors of this study (RJ, MRM, RR, and RS) independently performed a literature search systematically using the databases mentioned above (Supplementary Table S1). The study period was January 2008 to December 2018, inclusive. The search was limited to 10 years to make the information obtained relevant and contemporary. No language restriction was applied. The cross-references from the selected studies were searched for additional articles. Corresponding authors were contacted when the relevant information was not available in the publication. Discrepancies were resolved through discussion and consensus with a third reviewer.

2.2. Selection Criteria

The current study follows the preferred reporting items for systematic review and meta-analysis (PRISMA) guidelines [48]. The selection of studies was made based on the following inclusion and exclusion criteria.

2.3. Inclusion Criteria

  • Studies involving miRNAs expression and PC.
  • Studies involving clinical patient data or preclinical data.
  • Studies focusing on the resistance to some form of chemotherapy.
  • Studies that reported the miRNA profiling platforms.
  • Articles using in vitro assays to analyse the expression of miRNAs or gene related studies.

2.4. Exclusion Criteria

  • Studies published in non-English language and that do not involve drug resistance in PC were removed.
  • Case reports, review articles, editorial, and studies with only in vitro or only PC patient samples data were excluded.

2.5. Data Extraction

MS Office Excel worksheet was used to collect information about studies that were included for extraction. Prior PRISMA guidelines were used to design the data sheet content. Full text and corresponding supplementary information of the following items were collected and recorded from the eligible studies; first author, year of publication, patient information, location of the study, ethnicity, gender, drug used, clinical stage, number of samples, lymph node metastasis, cell line(s) used, miRNA(s) involved, miRNA profiling platform, and drug pathways or genes associated.

2.6. Quality Assessment

The quality of eligible studies was assessed by two authors (RJ and MRM) critically according to the meta-analysis of observational studies in epidemiology (MOOSE) [49] for epidemiological studies by the predefined checklist. The qualifying studies had all the criteria either mentioned in the study or later denoted by the corresponding author (Supplementary Table S2).

2.7. Publication Bias

Two authors (R.J. and M.M.R.) assessed the risk of bias through a few distinct methods [47,50,51,52,53]. Egger’s and Begg’s bias indicator test was used to calculating publication bias along with an inverted funnel plot. Begg-Mazumdar bias indicator test was done to check the effect of publication and selection bias. Duval and Tweedie’s trim and fill calculation was calculated additionally to compute the effect size.

2.8. Meta-Analysis

A meta-analysis was performed for the obtained HR values and 95% confidence interval (CI) from the articles and Kaplan-Meier curves of the eligible studies using comprehensive meta-analysis (CMA) software version 3.0. Random effects models were used for meta-analysis. Cochran’s Q test and the I2 statistic [54] were performed to assess the statistical heterogeneity. If p-value > 0.05 heterogeneity was observed and the random effects model was performed. Forest plot was drawn to summarise the pooled HR estimate of the chemoresistance specific miRNAs.

3. Results

Figure 1 explains our search selection and strategy in the form of a flowchart. The initial search resulted in 2671 studies from PubMed (n = 251) and Science Direct (n = 2420). After implementing the exclusion criteria, 169 articles were deemed relevant. After full-text screening and applying inclusion criteria, a total of 43 studies with miRNA expression related chemosensitivity or chemoresistance totalling 1963 individuals with PC was obtained for this study. The eligible articles were further reviewed (R.J., M.M.R.) and examined for data extraction (R.R. and R.S.). All the papers studied in our systematic review and meta-analysis were published in English. Out of the 43 studies, 23 were from China, seven were from the USA, seven were from Japan, five were from Germany, and one was from the Netherlands. Almost all studies (39 studies) used GEM as the primary drug for the treatment of PC. Both frozen and formalin fixed paraffin embedded (FFPE) tissue samples were used in the studies. Table 1 represents the descriptive characteristics of the included studies.
The most common assays performed is represented in Figure 2. A total of 59 cell lines have been used in the 43 studies, and PANC-1,2, Capan-1,2, HPDE and BxPc3 were the most commonly used ones. miRNA-21 modulates biological functions of PC cells including their proliferation, invasion, and chemoresistance studies used the highest number of cell lines (n = 14) [94].
In total, 48 miRNA have been studied in our systematic review; 23 of them were downregulated, and 25 were upregulated. In particular, nine upregulated miRNAs (15b, 17-5p, 21, 155, 181c, 203, 221, 320c and 1246) exhibited chemotherapeutic resistance and six upregulated miRNAs (21, 33a, 138-5p, 509-5p, 1207 and 1243) exhibited chemotherapeutic sensitivity. In contrast, nine downregulated miRNAs (7, 100, 124, 210, 200c, 205, 220b, 374b-5p and 497) exhibited chemotherapeutic resistance and nine downregulated miRNAs (101, 101-3p, 153, 203, 205-5p, 494, 506, 3656, let-7a) exhibited chemotherapeutic sensitivity. Four miRNA were differentially expressed. Overall, chemotherapeutic resistance (n = 18) and chemotherapeutic sensitivity (n = 15) were influenced by the miRNAs studied. The studies used GEM, lapatinib, capecitabine, 5-FU, a gamma-secretase inhibitor, Tarceva, radiation therapy, and AZD8055.
Treatment with GEM led to the downregulation of miRNA 210 via the ABCC5 pathway, miRNA 124 via the polypyrimidine tract binding protein (PTBP1) and pyruvate kinase pathway, miRNA 103 via the ribonucleotide reductase M1 (RRM1) pathway, miRNA 100 via the FGFR3 pathway, miRNA 497 via the FGFR signalling pathway and miRNA 7 and 2015 via the class III b-tubulin (TUBB3) pathway; causing a chemoresistance phenotype.
Treatment with GEM also led to the upregulation of miRNA 17-5p via the PTEN pathway, miRNA 221 via the EGFR1 and HER2 pathway, miRNA 203 via the activation of salt-inducible kinase (SLK1), miRNA 181c via the Hippo signalling pathway, miRNA 15b via the SMAD specific proteins pathway, miRNA 21 via the PTEN/Akt pathway, and VEGF, MMP-2 and MMP-9 proteins. Some studies noted upregulated miRNAs such as miRNA 221, 10a-5p and 21 no mechanistic pathways were identified. The upregulation of these miRNAs due to GEM treatment resulted in chemoresistance.
GEM treatment also led to the downregulation of miRNAs, causing an increase in chemosensitivity, such as miRNA 3656 via EMT, miRNA let-7a via the HMGA2 pathway, miRNA 205-5p via the activation of K-Ras, Caveolin-1 and Ki-67, miRNA 153 via the SNAIL pathway, miRNA 101 via DNA-PKcs, miRNA 506 via the activation of NF-κB and SPHK1, miRNA 494 via SIRT1, c-myc pathway, miRNA 203 via the ZEB-1 pathway. GEM treatment upregulated some miRNAs causing an increase in chemosensitivities such as miRNA 509-5p and 1243 both via the E-cadherin pathway, miRNA 33a via the Akt/B-catenin pathway and miRNA 21 via the FasL/Fas pathway.
5-FU is another drug that affects the regulation of miRNAs. Treatment with 5-FU led to the downregulation of miRNAs, thereby increasing chemoresistance, such as miRNA 200c and 200b both via directly targeting SUZ12, ROCK2. Treatment with 5-FU led to the upregulation of miRNA 1246 via the CCNG2 pathway, increasing chemoresistance. 5-FU also led to the downregulation of miRNA 494 via the SIRT1/c-myc pathway. Also, the upregulation of miRNA 138-5p via the vimentin resulted in chemosensitisation (Figure 3).
The miRNA and their drug targets based on the chemotherapeutic resistance and sensitivity is separated and listed out in Table 2 and Table 3 respectively.

4. Meta-Analysis

Associations between miRNA expression and patient survival were analysed using meta-analysis. The thorough screening revealed that 30 out of 43 studies did not report the HR values and 95% CI. Consequently, 30 studies were omitted from our meta-analysis due to insufficient reportage of data on patient survival and miRNA expressions. Collectively, 1088 patients from 12 studies were included in our meta-analysis (Figure 4). A pooled HR value of 1.603; 95% CI 1.2–2.143; p-value = 0.001 was obtained from a meta-analysis. Subgroup analysis between miR-21 showed an HR value of 2.061; 95% CI 1.195–3.556 and p-value of 0.009. Heterogeneity (I2 value) was observed to be 83.833 (Figure 5).
PC patients with elevated expression of miR-10a-5p (HR = 2.878, 95% CI = 1.614–5.131), 17-5p (HR = 1.18, 95% CI = 0.979–1.414), 21 (HR = 1.71, 95% CI = 1.147–2.535), 21 (HR = 0.32, 95% CI = 0.166–0.6), 33a (HR = 1.08, 95% CI = 1.002–1.168), 153 (HR = 1.16, 95% CI = 0.43–3.16), 181c (HR = 2.03, 95% CI = 1.33–3.11), 497 (HR = 2.76, 95% CI = 1.159–6.579), 506 (HR = 1.88, 95% CI = 1.048–3.026), 744 (HR = 21.2, 95% CI = 3.17–436) and 3656 (HR = 2.32, 95% CI = 1.37–3.57) exhibited chemotherapeutic resistance and a poorer prognosis. The low expression of miRNA-374b-5p (HR = 0.3, 95% CI = 0.17–0.54) revealed chemotherapeutic sensitivity and a better prognosis.
Figure 4 represents the forest plot of the primary meta-analysis of the pooled HR values along with the 95% CI from a pancreatic cancer patient, which are calculated using comprehensive meta-analysis (CMA) software (version 3.3.070, Biostat, Englewood, NJ, USA). The graphical representation of the right side of the plot is the HR, and 95% of the included studies and the red squares with the line represents the effect size of miRNA expressions. If the HR value is more than 1, it indicates the increased risk of the patient’s survival and less than specifies the decreased risk of patient’s survival. The size of the box indicates the weight of the study.

5. Publication Bias Indicators

5.1. Classic Fail-Safe N

This meta-analysis incorporates data from 12 studies, which yield a z-value of 6.26484 and the corresponding two-tailed p-value of 0.00000. The fail-safe N is 111. This means that we would need to locate and include 111 ‘null’ studies in order for the combined two-tailed p-value to exceed 0.050. Stated in another way, there would need to be 5.7 missing studies for every observed study for the effect to be nullified.

5.2. Orwin Fail-Safe N

Here, the hazard ratio in observed studies is 1.150, which did not fall between the mean hazard ratio in the missing studies, so we could therefore not calculate the Orwin fail-safe N.

5.3. Begg and Mazumdar Rank Correlation Test

The Kendall′s tau b is 0.25758, with a one-tailed p-value of 1.16573 or a two-tailed p-value of 0.12186. This value compares the effect size and variance with the tau value and the value closes to 1 correlates to signify the publication bias.

5.4. Egger′s Test of the Intercept

In this case the intercept (B0) is 1.77800, 95% CI (−0.02031–3.57630), with t = 2.20298, df = 10. The 1-tailed p-value is 0.02609, and the 2-tailed p-value is 0.05218.

5.5. Duval and Tweedie′s Trim and Fill Test

Under the fixed effect model, the point estimate and 95% confidence interval for the combined studies are 1.15084 (1.07656, 1.23025). Using trim and fill, the imputed point estimate is 1.10505 (1.03516, 1.17967). Under the random effects model, the point estimate and 95% confidence interval for the combined studies are 1.60344 (1.19976, 2.14294). Using trim and fill, the imputed point estimate is 1.13206 (0.85429, 1.50016).
The hypothesis and heterogeneity testing are represented in Table 4.
Funnel plot is represented in Figure 6, and funnel plot with observed and imputed studies in Figure 7, which represents the possible bias between the included studies. If the studies have no publication bias, then the points will fall on the central line, indicating the symmetry. Every study or cohort included in the forest plot is represented as a point on the funnel plot.

6. Discussion

Emerging studies have revealed that specific miRNA expression in PC is related to chemotherapeutic sensitivity/resistance and regulation of molecular signalling pathways. Therefore, a systematic review and meta-analysis to catalogue the miRNA expression patterns and decipher the relationships and regulation of oncogenic signalling pathways to outcomes would help in the prediction of chemotherapy response and triaging of future clinical therapeutic strategies in PC.
We conducted this systematic review of published studies on miRNA expressions to inventory the full range of chemotherapeutic resistance and sensitivity in PC. To our knowledge, this is the first systematic review and meta-analysis describing the role of miRNA expression in chemotherapeutic resistance and sensitivity as well as molecular signalling pathways in PC. A total of 43 studies with 1963 patients were evaluated with 2207 PC tissue samples analysed. Also, 162 blood samples from the PC patients were examined in our study.
We found that distinct miRNAs were upregulated or downregulated in PC cell lines and tissues. In turn, they targeted specific molecular signalling pathways to mediate sensitivity or resistance to chemotherapeutic drugs in PC. Of the eight chemotherapeutic drugs studied individually and in combinations, GEM was the most studied followed by 5-FU.
In a study on lung cancer cells, miR-17-5p downregulation was associated with an increased expression of beclin 1 gene, which is an autophagy modulator in the survival pathway [96]. Furthermore, this miRNA belongs to the miR-17-92 cluster and is upregulated in PC, where it is linked to pancreatic carcinogenesis [97,98]. Downregulation of miRNA 7 and 205 resulted in chemoresistance in PC, and a possible target is TUBB3, as noted in another study [99,100].
Our meta-analysis showed a non-significant pooled effect size, suggesting that drug-resistance specific miRNAs may not necessarily be good predictors of patient survival. However, it is essential to note that we have used only nine clinical studies in our meta-analysis due to a general lack of studies reporting clinical outcomes via HR and 95% CI values. From the data on publication bias, we concluded that several factors might be responsible for high heterogeneity between the data, such as study strategy, inadequate information and sample size [101]. The publication bias results computed that the studies required for analysing the statistically non-significant overall effect, and there were no missing studies as per the report [44,46,102].
The fact that many of the included studies did not investigate the HR values, exhibits a significant limitation in our quantitative synthesis [46]. Several included studies have investigated differentially expressed miRNA as drivers of drug resistance in PC and as prognostic markers by comparing their expression level in a tumour and adjacent healthy tissue.

7. Limitations and Strengths of Our Study

High variability in the data was observed and could be the result of factors such as (1) different sample groups; (2) diverse validation methods; and (3) limited sampling. Overcoming tumour resistance remains a significant challenge in the treatment of PC. The significant factors in chemo-resistance are the presence of miRNA expressions and their genetic regulation. Our systematic review and analysis attempted to show the association between the expression levels of miRNAs and chemotherapeutic resistance in PC. This review would help achieve a better understanding of the overall network of the mechanisms contributing to drug resistance and the regulation of the correlated miRNAs.

8. Conclusions

This systematic review and meta-analysis of miRNA expressions identified critical determinants of chemotherapeutic sensitivity and resistance in PC and correlated these with oncogenic molecular signalling pathways. The knowledge based on the miRNA and drug targets could drive the choice of chemotherapeutic regimens used in patients, adoption of combination therapies that overcome therapeutic resistance, utilisation of miRNAs as biomarkers of chemotherapy response, and early adaptive modifications to treatment in response to alterations in miRNA profiles of tumours.

Supplementary Materials

The following are available online at https://www.mdpi.com/2072-6694/11/7/900/s1, Table S1: The search strategy, Table S2: Quality assessment of the included studies.

Author Contributions

Conceptualization, R.J., and M.M.R.; methodology R.J., M.M.R., R.R., R.S., S.S., and C.K.; software, R.J., C.K., and M.M.R..; validation R.J., M.M.R., R.R., R.S., S.S., S.B., A.G., C.K., S.K., N.R. and K.M.G..; formal analysis, R.J., S.S., C.K., and M.M.R.; investigation, M.M.R., R.R., R.S., S.S., and C.K.; resources, R.J.; data curation, R.J., M.M.R., R.R., R.S., S.S., and C.K.; writing—original draft preparation, R.J., and M.M.R.; writing—review and editing, R.R., R.S., S.S., S.B., A.G., C.K., S.K., N.R. and K.M.G.; supervision, R.J., S.B., A.G., C.K., S.K., N.R. and K.M.G.

Funding

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

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PCPancreatic cancer
HRHazard ratio
CIsConfidence Intervals
GEMGemcitabine
5-FU5-fluorouracil
CAPCapecitabine
LAPLapatinib
GSIγ-secretase inhibitor
OHPOxaliplatin
NMNot Mentioned
RHOFRas homolog family member F
EMTEpithelial–Mesenchymal Transition
CXCR4 C-X- C chemokine receptor type 4
HMGA2High-mobility group AT-hook 2
DNA-PKcsDNA- dependent protein kinase catalytic subunit
PTBP1Polypyrimidine tract binding protein 1
PKM2Pyruvate Kinase
SPHK1Sphingosine Kinase 1
NF-κBNuclear Factor kappa-light- chain-enhancer of activated B cells
PTENPhosphatase and Tensin Homolog
EGFREpidermal Growth Factor Receptor
HER2Human Epidermal growth factor Receptor 2
RMM1Ribonucleotide eductase M1
SIK1Salt-inducible Kinase 1
MST1/2Mammalian STE20-like protein kinase 1/2
LATS1/2Large Tumour Suppressor 1/2
SAV1Salvador homolog 1
MOB1MOB kinase activator 1
SMURF2SMAD specific E3 ubiquitin protein ligase 2
VIMVimentin
GSK-3 βGlycogen Synthase Kinase 3 beta
SIRT1Silent mating type Information Regulation 2 homolog 1
ZEB1Zinc finger e-box binding homeobox 1
FGFFibroblast Growth Factor
FGFR3Fibroblast Growth Factor Receptor 3
ATMAtaxia Telangiectasia Mutated protein
ROCK2Rho associated coiled-coil containing protein kinase 2
MTORMechanistic Target of Rapamycin
RRM2Ribonucleotide Reductase M2
TUBB3class III b-tubulin
VEGFVascular Endothelial Growth Factor
MMPMatrix Metalloproteinases

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. World Cancer Research Fund International. Pancreatic cancer statistics. Available online: http://www.wcrf.org/int/cancer-facts-figures/data-specific-cancers/pancreatic-cancer-statistics (accessed on 11 December 2017).
  3. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA A Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. GLOBCAN. Cancer Incidence, Mortality and Prevalence Worldwide, World Pancreas New Cancer Cases. Available online: http://globocan.iarc.fr/old/burden.asp?selection_pop=224900&Text-p=World&selection_cancer=23090&Text-c=Pancreas&pYear=3&type=0&window=1&submit=%C2%A0Execute (accessed on 11 December 2017).
  5. GLOBCAN. Cancer Incidence, Mortality and Prevalence Worldwide, World Pancreas Deaths. Available online: http://globocan.iarc.fr/old/burden.asp?selection_pop=224900&Text-p=World&selection_cancer=23090&Text-c=Pancreas&pYear=3&type=1&window=1&submit=%C2%A0Execute (accessed on 11 December 2017).
  6. INDIA AGAINST CANCER and NICPR. Cancer Factsheet. Available online: http://cancerindia.org.in/cancer-factsheet/ (accessed on 11 December 2017).
  7. Hartwig, W.; Hackert, T.; Hinz, U.; Gluth, A.; Bergmann, F.; Strobel, O.; Büchler, M.W.; Werner, J. Pancreatic cancer surgery in the new millennium: Better prediction of outcome. Ann. Surg. 2011, 254, 311–319. [Google Scholar] [CrossRef] [PubMed]
  8. Van Rijswijk, R.; Jeziorski, K.; Wagener, D.T.; Van Laethem, J.-L.; Reuse, S.; Baron, B.; Wils, J.; EORTC GastroIntestinal Tract Cancer Cooperative Group. Weekly high-dose 5-fluorouracil and folinic acid in metastatic pancreatic carcinoma: A phase II study of the EORTC GastroIntestinal Tract Cancer Cooperative Group. Eur. J. Cancer 2004, 40, 2077–2081. [Google Scholar] [CrossRef] [PubMed]
  9. Cullinan, S.A.; Moertel, C.G.; Fleming, T.R.; Rubin, J.R.; Krook, J.E.; Everson, L.K.; Windschitl, H.E.; Twito, D.I.; Marschke, R.F.; Foley, J.F. A comparison of three chemotherapeutic regimens in the treatment of advanced pancreatic and gastric carcinoma: Fluorouracil vs fluorouracil and doxorubicin vs fluorouracil, doxorubicin, and mitomycin. JAMA 1985, 253, 2061–2067. [Google Scholar] [CrossRef]
  10. Androulakis, N.; Kourousis, C.; Dimopoulos, M.A.; Samelis, G.; Kakolyris, S.; Tsavaris, N.; Genatas, K.; Aravantinos, G.; Papadimitriou, C.; Karabekios, S. Treatment of pancreatic cancer with docetaxel and granulocyte colony-stimulating factor: A multicenter phase II study. J. Clin. Oncol. 1999, 17, 1779. [Google Scholar] [CrossRef] [PubMed]
  11. Wagener, D.T.; Verdonk, H.; Dirix, L.; Catimel, G.; Siegenthaler, P.; Buitenhuis, M.; Mathieu-Boue, A.; Verweij, J. Phase II trial of CPT-11 in patients with advanced pancreatic cancer, an EORTC early clinical trials group study. Ann. Oncol. 1995, 6, 129–132. [Google Scholar] [CrossRef]
  12. Oettle, H.; Post, S.; Neuhaus, P.; Gellert, K.; Langrehr, J.; Ridwelski, K.; Schramm, H.; Fahlke, J.; Zuelke, C.; Burkart, C. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: A randomized controlled trial. Jama 2007, 297, 267–277. [Google Scholar] [CrossRef]
  13. Berlin, J.D.; Catalano, P.; Thomas, J.P.; Kugler, J.W.; Haller, D.G.; Benson III, A.B. Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J. Clin. Oncol. 2002, 20, 3270–3275. [Google Scholar] [CrossRef]
  14. Herrmann, R.; Bodoky, G.r.; Ruhstaller, T.; Glimelius, B.; Bajetta, E.; Schuller, J.; Saletti, P.; Bauer, J.; Figer, A.; Pestalozzi, B. Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: A randomized, multicenter, phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J. Clin. Oncol. 2007, 25, 2212–2217. [Google Scholar] [CrossRef]
  15. Ueno, H.; Ioka, T.; Ikeda, M.; Ohkawa, S.; Yanagimoto, H.; Boku, N.; Fukutomi, A.; Sugimori, K.; Baba, H.; Yamao, K. Randomized phase III study of gemcitabine plus S-1, S-1 alone, or gemcitabine alone in patients with locally advanced and metastatic pancreatic cancer in Japan and Taiwan: GEST study. J. Clin. Oncol. 2013, 31, 1640–1648. [Google Scholar] [CrossRef] [PubMed]
  16. Chao, Y.; Wu, C.-Y.; Wang, J.P.; Lee, R.-C.; Lee, W.-P.; Li, C.-P. A randomized controlled trial of gemcitabine plus cisplatin versus gemcitabine alone in the treatment of metastatic pancreatic cancer. Cancer Chemother. Pharmacol. 2013, 72, 637–642. [Google Scholar] [CrossRef] [PubMed]
  17. Stathopoulos, G.; Syrigos, K.; Aravantinos, G.; Polyzos, A.; Papakotoulas, P.; Fountzilas, G.; Potamianou, A.; Ziras, N.; Boukovinas, J.; Varthalitis, J. A multicenter phase III trial comparing irinotecan-gemcitabine (IG) with gemcitabine (G) monotherapy as first-line treatment in patients with locally advanced or metastatic pancreatic cancer. Br. J. Cancer 2006, 95, 587–592. [Google Scholar] [CrossRef] [PubMed]
  18. Reni, M.; Cordio, S.; Milandri, C.; Passoni, P.; Bonetto, E.; Oliani, C.; Luppi, G.; Nicoletti, R.; Galli, L.; Bordonaro, R. Gemcitabine versus cisplatin, epirubicin, fluorouracil, and gemcitabine in advanced pancreatic cancer: A randomised controlled multicentre phase III trial. Lancet Oncol. 2005, 6, 369–376. [Google Scholar] [CrossRef]
  19. Poplin, E.; Feng, Y.; Berlin, J.; Rothenberg, M.L.; Hochster, H.; Mitchell, E.; Alberts, S.; O’Dwyer, P.; Haller, D.; Catalano, P. Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: A trial of the Eastern Cooperative Oncology Group. J. Clin. Oncol. 2009, 27, 3778–3785. [Google Scholar] [CrossRef] [PubMed]
  20. Moore, M.J.; Goldstein, D.; Hamm, J.; Figer, A.; Hecht, J.R.; Gallinger, S.; Au, H.J.; Murawa, P.; Walde, D.; Wolff, R.A. 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. [Google Scholar] [CrossRef]
  21. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef]
  22. Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007, 2, 751–760. [Google Scholar] [CrossRef]
  23. Wiebe, L.A. A myriad of symptoms: New approaches to optimizing palliative care of patients with advanced pancreatic cancer. Am. Soc. Clin. Oncol. Educ. Book 2012, 32, 243–248. [Google Scholar]
  24. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef] [Green Version]
  25. Liu, R.; Chen, X.; Du, Y.; Yao, W.; Shen, L.; Wang, C.; Hu, Z.; Zhuang, R.; Ning, G.; Zhang, C. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin. Chem. 2012, 58, 610–618. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, E.J.; Gusev, Y.; Jiang, J.; Nuovo, G.J.; Lerner, M.R.; Frankel, W.L.; Morgan, D.L.; Postier, R.G.; Brackett, D.J.; Schmittgen, T.D. Expression profiling identifies microRNA signature in pancreatic cancer. Int. J. Cancer 2007, 120, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, J.; Chen, J.; Chang, P.; LeBlanc, A.; Li, D.; Abbruzzesse, J.L.; Frazier, M.L.; Killary, A.M.; Sen, S. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev. Res. 2009, 2, 807–813. [Google Scholar] [CrossRef] [PubMed]
  28. Dillhoff, M.; Liu, J.; Frankel, W.; Croce, C.; Bloomston, M. MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J. Gastrointest. Surg. 2008, 12, 2171. [Google Scholar] [CrossRef] [PubMed]
  29. Greither, T.; Grochola, L.F.; Udelnow, A.; Lautenschläger, C.; Würl, P.; Taubert, H. Elevated expression of microRNAs 155, 203, 210 and 222 in pancreatic tumors is associated with poorer survival. Int. J. Cancer 2010, 126, 73–80. [Google Scholar] [CrossRef]
  30. O’donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843. [Google Scholar] [CrossRef]
  31. Tagawa, H.; Karube, K.; Tsuzuki, S.; Ohshima, K.; Seto, M. Synergistic action of the microRNA-17 polycistron and Myc in aggressive cancer development. Cancer Sci. 2007, 98, 1482–1490. [Google Scholar] [CrossRef]
  32. Shi, X.; Liu, S.; Kleeff, J.; Friess, H.; Büchler, M.W. Acquired resistance of pancreatic cancer cells towards 5-Fluorouracil and gemcitabine is associated with altered expression of apoptosis-regulating genes. Oncology 2002, 62, 354–362. [Google Scholar] [CrossRef]
  33. Yu, S.; Lu, Z.; Liu, C.; Meng, Y.; Ma, Y.; Zhao, W.; Liu, J.; Yu, J.; Chen, J. miRNA-96 suppresses KRAS and functions as a tumor suppressor gene in pancreatic cancer. Cancer Res. 2010, 70, 6015–6025. [Google Scholar] [CrossRef]
  34. Zhang, Y.; Li, M.; Wang, H.; Fisher, W.E.; Lin, P.H.; Yao, Q.; Chen, C. Profiling of 95 microRNAs in pancreatic cancer cell lines and surgical specimens by real-time PCR analysis. World J. Surg. 2009, 33, 698. [Google Scholar] [CrossRef]
  35. Li, Y.; VandenBoom, T.G.; Kong, D.; Wang, Z.; Ali, S.; Philip, P.A.; Sarkar, F.H. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009, 69, 6704–6712. [Google Scholar] [CrossRef] [PubMed]
  36. Zhu, H.; Wu, H.; Liu, X.; Evans, B.R.; Medina, D.J.; Liu, C.-G.; Yang, J.-M. Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem. Pharmacol. 2008, 76, 582–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Kovalchuk, O.; Filkowski, J.; Meservy, J.; Ilnytskyy, Y.; Tryndyak, V.P.; Vasyl’F, C.; Pogribny, I.P. Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol. Cancer Ther. 2008, 7, 2152–2159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Ma, J.; Dong, C.; Ji, C. MicroRNA and drug resistance. Cancer Gene Ther. 2010, 17, 523–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Li, H.; Yang, B.B. MicroRNA-in drug resistance. Oncoscience 2014, 1, 3. [Google Scholar]
  40. Zheng, T.; Wang, J.; Chen, X.; Liu, L. Role of microRNA in anticancer drug resistance. Int. J. Cancer 2010, 126, 2–10. [Google Scholar] [CrossRef] [PubMed]
  41. Ali, S.; Ahmad, A.; Banerjee, S.; Padhye, S.; Dominiak, K.; Schaffert, J.M.; Wang, Z.; Philip, P.A.; Sarkar, F.H. Gemcitabine sensitivity can be induced in pancreatic cancer cells through modulation of miR-200 and miR-21 expression by curcumin or its analogue CDF. Cancer Res. 2010, 70, 3606–3617. [Google Scholar] [CrossRef]
  42. Wang, Z.; Li, Y.; Kong, D.; Banerjee, S.; Ahmad, A.; Azmi, A.S.; Ali, S.; Abbruzzese, J.L.; Gallick, G.E.; Sarkar, F.H. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009, 69, 2400–2407. [Google Scholar] [CrossRef]
  43. Hong, S.P.; Wen, J.; Bang, S.; Park, S.; Song, S.Y. CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int. J. Cancer 2009, 125, 2323–2331. [Google Scholar] [CrossRef]
  44. Jayaraj, R.; Kumarasamy, C. Systematic review and meta-analysis of cancer studies evaluating diagnostic test accuracy and prognostic values: Approaches to improve clinical interpretation of results. Cancer Manag. Res. 2018, 10, 4669–4670. [Google Scholar] [CrossRef]
  45. Jayaraj, R.; Kumarasamy, C.; Ramalingam, S.; Devi, A. Systematic review and meta-analysis of risk-reductive dental strategies for medication related osteonecrosis of the jaw among cancer patients: Approaches and strategies. Oral Oncol. 2018, 86, 312–313. [Google Scholar] [CrossRef] [PubMed]
  46. Sabarimurugan, S.; Royam, M.M.; Das, A.; Das, S.; Gothandam, K.M.; Jayaraj, R. Systematic Review and Meta-analysis of the Prognostic Significance of miRNAs in Melanoma Patients. Mol. Diagn. Ther. 2018, 1–17. [Google Scholar] [CrossRef] [PubMed]
  47. Jayaraj, R.; Kumarasamy, C.; Madhav, M.R.; Pandey, V.; Sabarimurugan, S.; Ramesh, N.; Gothandam, K.M.; Baxi, S. Comment on “Systematic Review and Meta-Analysis of Diagnostic Accuracy of miRNAs in Patients with Pancreatic Cancer”. Dis. Markers 2018, 2018, 2. [Google Scholar] [CrossRef] [PubMed]
  48. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Group, P. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed]
  49. Stroup, D.F.; Berlin, J.A.; Morton, S.C.; Olkin, I.; Williamson, G.D.; Rennie, D.; Moher, D.; Becker, B.J.; Sipe, T.A.; Thacker, S.B. Meta-analysis of observational studies in epidemiology: A proposal for reporting. JAMA 2000, 283, 2008–2012. [Google Scholar] [CrossRef] [PubMed]
  50. Jayaraj, R.; Kumarasamy, C. Comment on “Prognostic biomarkers for oral tongue squamous cell carcinoma: A systematic review and meta-analysis”. Br. J. Cancer 2018, 118, e11. [Google Scholar] [CrossRef] [PubMed]
  51. Jayaraj, R.; Kumarasamy, C. Comment on “Survival for HPV-positive oropharyngeal squamous cell carcinoma with surgical versus non-surgical treatment approach: A systematic review and meta-analysis”. Oral Oncol. 2019, 90, 137. [Google Scholar] [CrossRef]
  52. Jayaraj, R.; Kumarasamy, C.; Sabarimurugan, S.; Baxi, S. Commentary: Blood-Derived microRNAs for Pancreatic Cancer Diagnosis: A Narrative Review and Meta-Analysis. Front. Physiol. 2018, 9, 1896. [Google Scholar] [CrossRef]
  53. Jayaraj, R.; Kumarasamy, C. Conceptual interpretation of analysing and reporting of results on systematic review and meta-analysis of optimal extent of lateral neck dissection for well-differentiated thyroid carcinoma with metastatic lateral neck lymph nodes. Oral Oncol. 2019, 89, 153. [Google Scholar] [CrossRef]
  54. Higgins, J.; Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 2002, 21, 1539–1558. [Google Scholar] [CrossRef]
  55. Yang, R.-M.; Zhan, M.; Xu, S.-W.; Long, M.-M.; Yang, L.-H.; Chen, W.; Huang, S.; Liu, Q.; Zhou, J.; Zhu, J. miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death Dis. 2017, 8, e3129. [Google Scholar] [CrossRef] [PubMed]
  56. Xiao, G.; Wang, X.; Yu, Y. CXCR4/Let-7a Axis Regulates Metastasis and Chemoresistance of Pancreatic Cancer Cells Through Targeting HMGA2. Cell. Physiol. Biochem. 2017, 43, 840–851. [Google Scholar] [CrossRef] [PubMed]
  57. Hiramoto, H.; Muramatsu, T.; Ichikawa, D.; Tanimoto, K.; Yasukawa, S.; Otsuji, E.; Inazawa, J. miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Sci. Rep. 2017, 7, 4002. [Google Scholar] [CrossRef] [PubMed]
  58. Chaudhary, A.K.; Mondal, G.; Kumar, V.; Kattel, K.; Mahato, R.I. Chemosensitization and inhibition of pancreatic cancer stem cell proliferation by overexpression of microRNA-205. Cancer Lett. 2017, 402, 1–8. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, F.; Liu, B.; Qian, J.; Wu, G.; Li, J.; Ma, Z. miR-153 enhances the therapeutic effect of gemcitabine by targeting Snail in pancreatic cancer. Acta Biochim. Biophys. Sin. 2017, 49, 520–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K. MicroRNA-155 controls exosome synthesis and promotes gemcitabine resistance in pancreatic ductal adenocarcinoma. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef]
  61. Hu, H.; He, Y.; Wang, Y.; Chen, W.; Hu, B.; Gu, Y. micorRNA-101 silences DNA-PKcs and sensitizes pancreatic cancer cells to gemcitabine. Biochem. Biophys. Res. Commun. 2017, 483, 725–731. [Google Scholar] [CrossRef]
  62. Amponsah, P.S.; Fan, P.; Bauer, N.; Zhao, Z.; Gladkich, J.; Fellenberg, J.; Herr, I. microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer. Cancer Lett. 2017, 388, 107–117. [Google Scholar] [CrossRef]
  63. Li, C.; Zhao, Z.; Zhou, Z.; Liu, R. Linc-ROR confers gemcitabine resistance to pancreatic cancer cells via inducing autophagy and modulating the miR-124/PTBP1/PKM2 axis. Cancer Chemother. Pharmacol. 2016, 78, 1199–1207. [Google Scholar] [CrossRef]
  64. Li, J.; Wu, H.; Li, W.; Yin, L.; Guo, S.; Xu, X.; Ouyang, Y.; Zhao, Z.; Liu, S.; Tian, Y. Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-κB signaling. Oncogene 2016, 35, 5501. [Google Scholar] [CrossRef]
  65. Gu, J.; Wang, D.; Zhang, J.; Zhu, Y.; Li, Y.; Chen, H.; Shi, M.; Wang, X.; Shen, B.; Deng, X. GFRα2 prompts cell growth and chemoresistance through down-regulating tumor suppressor gene PTEN via Mir-17-5p in pancreatic cancer. Cancer Lett. 2016, 380, 434–441. [Google Scholar] [CrossRef] [PubMed]
  66. Tian, X.; Shivapurkar, N.; Wu, Z.; Hwang, J.J.; Pishvaian, M.J.; Weiner, L.M.; Ley, L.; Zhou, D.; Zhi, X.; Wellstein, A. Circulating microRNA profile predicts disease progression in patients receiving second-line treatment of lapatinib and capecitabine for metastatic pancreatic cancer. Oncology Lett. 2016, 11, 1645–1650. [Google Scholar] [CrossRef] [PubMed]
  67. Fan, P.; Liu, L.; Yin, Y.; Zhao, Z.; Zhang, Y.; Amponsah, P.S.; Xiao, X.; Bauer, N.; Abukiwan, A.; Nwaeburu, C.C. MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett. 2016, 373, 130–137. [Google Scholar] [CrossRef] [PubMed]
  68. Yao, J.; Li, Z.; Wang, X.; Xu, P.; Zhao, L.; Qian, J. MiR-125a regulates chemo-sensitivity to gemcitabine in human pancreatic cancer cells through targeting A20. Acta Biochim. Biophys. Sin. 2016, 48, 202–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ren, Z.-G.; Dong, S.-X.; Han, P.; Qi, J. miR-203 promotes proliferation, migration and invasion by degrading SIK1 in pancreatic cancer. Oncol. Rep. 2016, 35, 1365–1374. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, M.; Wang, M.; Xu, S.; Guo, X.; Jiang, J. Upregulation of miR-181c contributes to chemoresistance in pancreatic cancer by inactivating the Hippo signaling pathway. Oncotarget 2015, 6, 44466. [Google Scholar] [CrossRef] [PubMed]
  71. Miyamae, M.; Komatsu, S.; Ichikawa, D.; Kawaguchi, T.; Hirajima, S.; Okajima, W.; Ohashi, T.; Imamura, T.; Konishi, H.; Shiozaki, A. Plasma microRNA profiles: Identification of miR-744 as a novel diagnostic and prognostic biomarker in pancreatic cancer. Br. J. Cancer 2015, 113, 1467. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, W.-L.; Zhang, J.-H.; Wu, X.-Z.; Yan, T.; Lv, W. miR-15b promotes epithelial-mesenchymal transition by inhibiting SMURF2 in pancreatic cancer. Int. J. Oncol. 2015, 47, 1043–1053. [Google Scholar] [CrossRef] [Green Version]
  73. Yu, C.; Wang, M.; Chen, M.; Huang, Y.; Jiang, J. Upregulation of microRNA-138-5p inhibits pancreatic cancer cell migration and increases chemotherapy sensitivity. Mol. Med. Rep. 2015, 12, 5135–5140. [Google Scholar] [CrossRef]
  74. Liang, C.; Yu, X.-J.; Guo, X.-Z.; Sun, M.-H.; Wang, Z.; Song, Y.; Ni, Q.-X.; Li, H.-Y.; Mukaida, N.; Li, Y.-Y. MicroRNA-33a-mediated downregulation of Pim-3 kinase expression renders human pancreatic cancer cells sensitivity to gemcitabine. Oncotarget 2015, 6, 14440. [Google Scholar] [CrossRef]
  75. Liu, Y.; Li, X.; Zhu, S.; Zhang, J.; Yang, M.; Qin, Q.; Deng, S.; Wang, B.; Tian, K.; Liu, L. Ectopic expression of miR-494 inhibited the proliferation, invasion and chemoresistance of pancreatic cancer by regulating SIRT1 and c-Myc. Gene Ther. 2015, 22, 729. [Google Scholar] [CrossRef] [PubMed]
  76. Meidhof, S.; Brabletz, S.; Lehmann, W.; Preca, B.T.; Mock, K.; Ruh, M.; Schüler, J.; Berthold, M.; Weber, A.; Burk, U. ZEB1-associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat. EMBO Mol. Med. 2015, 7, 831–847. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, Y.; Zhao, L.; Ischenko, I.; Bao, Q.; Schwarz, B.; Nieß, H.; Wang, Y.; Renner, A.; Mysliwietz, J.; Jauch, K.-W. Antisense inhibition of microRNA-21 and microRNA-221 in tumor-initiating stem-like cells modulates tumorigenesis, metastasis, and chemotherapy resistance in pancreatic cancer. Target. Oncol. 2015, 10, 535–548. [Google Scholar] [CrossRef] [PubMed]
  78. Li, Z.; Li, X.; Yu, C.; Wang, M.; Peng, F.; Xiao, J.; Tian, R.; Jiang, J.; Sun, C. MicroRNA-100 regulates pancreatic cancer cells growth and sensitivity to chemotherapy through targeting FGFR3. Tumor Bio. 2014, 35, 11751–11759. [Google Scholar] [CrossRef] [PubMed]
  79. Xu, J.; Wang, T.; Cao, Z.; Huang, H.; Li, J.; Liu, W.; Liu, S.; You, L.; Zhou, L.; Zhang, T. MiR-497 downregulation contributes to the malignancy of pancreatic cancer and associates with a poor prognosis. Oncotarget 2014, 5, 6983. [Google Scholar] [CrossRef] [PubMed]
  80. Hasegawa, S.; Eguchi, H.; Nagano, H.; Konno, M.; Tomimaru, Y.; Wada, H.; Hama, N.; Kawamoto, K.; Kobayashi, S.; Nishida, N. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br. J. Cancer 2014, 111, 1572. [Google Scholar] [CrossRef] [PubMed]
  81. Lai, I.-L.; Chou, C.-C.; Lai, P.-T.; Fang, C.-S.; Shirley, L.A.; Yan, R.; Mo, X.; Bloomston, M.; Kulp, S.K.; Bekaii-Saab, T. Targeting the Warburg effect with a novel glucose transporter inhibitor to overcome gemcitabine resistance in pancreatic cancer cells. Carcinogenesis 2014, 35, 2203–2213. [Google Scholar] [CrossRef] [PubMed]
  82. Song, W.-F.; Wang, L.; Huang, W.-Y.; Cai, X.; Cui, J.-J.; Wang, L.-W. MiR-21 upregulation induced by promoter zone histone acetylation is associated with chemoresistance to gemcitabine and enhanced malignancy of pancreatic cancer cells. Asian Pac. J. Cancer Prev. 2013, 14, 7529–7536. [Google Scholar] [CrossRef]
  83. Peng, F.; Jiang, J.; Yu, Y.; Tian, R.; Guo, X.; Li, X.; Shen, M.; Xu, M.; Zhu, F.; Shi, C. Direct targeting of SUZ12/ROCK2 by miR-200b/c inhibits cholangiocarcinoma tumourigenesis and metastasis. Br. J. Cancer 2013, 109, 3092. [Google Scholar] [CrossRef]
  84. Nagano, H.; Tomimaru, Y.; Eguchi, H.; Hama, N.; Wada, H.; Kawamoto, K.; Kobayashi, S.; Mori, M.; Doki, Y. MicroRNA-29a induces resistance to gemcitabine through the Wnt/β-catenin signaling pathway in pancreatic cancer cells. Int. J. Oncol. 2013, 43, 1066–1072. [Google Scholar] [CrossRef]
  85. Wei, F.; Liu, Y.; Guo, Y.; Xiang, A.; Wang, G.; Xue, X.; Lu, Z. miR-99b-targeted mTOR induction contributes to irradiation resistance in pancreatic cancer. Mol. Cancer 2013, 12, 81. [Google Scholar] [CrossRef] [PubMed]
  86. Iwagami, Y.; Eguchi, H.; Nagano, H.; Akita, H.; Hama, N.; Wada, H.; Kawamoto, K.; Kobayashi, S.; Tomokuni, A.; Tomimaru, Y. miR-320c regulates gemcitabine-resistance in pancreatic cancer via SMARCC1. Br. J. Cancer 2013, 109, 502. [Google Scholar] [CrossRef] [PubMed]
  87. Bhutia, Y.D.; Hung, S.W.; Krentz, M.; Patel, D.; Lovin, D.; Manoharan, R.; Thomson, J.M.; Govindarajan, R. Differential processing of let-7a precursors influences RRM2 expression and chemosensitivity in pancreatic cancer: Role of LIN-28 and SET oncoprotein. PLoS ONE 2013, 8, e53436. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, P.; Zhuang, L.; Zhang, J.; Fan, J.; Luo, J.; Chen, H.; Wang, K.; Liu, L.; Chen, Z.; Meng, Z. The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FasL. Mol. Oncol. 2013, 7, 334–345. [Google Scholar] [CrossRef] [PubMed]
  89. Singh, S.; Chitkara, D.; Kumar, V.; Behrman, S.W.; Mahato, R.I. miRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett. 2013, 334, 211–220. [Google Scholar] [CrossRef] [PubMed]
  90. Brabletz, S.; Bajdak, K.; Meidhof, S.; Burk, U.; Niedermann, G.; Firat, E.; Wellner, U.; Dimmler, A.; Faller, G.; Schubert, J. The ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells. EMBO J. 2011, 30, 770–782. [Google Scholar] [CrossRef] [PubMed]
  91. Ali, S.; Almhanna, K.; Chen, W.; Philip, P.A.; Sarkar, F.H. Differentially expressed miRNAs in the plasma may provide a molecular signature for aggressive pancreatic cancer. Am. J. Transl. Res. 2011, 3, 28. [Google Scholar]
  92. Hwang, J.-H.; Voortman, J.; Giovannetti, E.; Steinberg, S.M.; Leon, L.G.; Kim, Y.-T.; Funel, N.; Park, J.K.; Kim, M.A.; Kang, G.H. Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PloS ONE 2010, 5, e10630. [Google Scholar] [CrossRef]
  93. Giovannetti, E.; Funel, N.; Peters, G.J.; Del Chiaro, M.; Erozenci, L.A.; Vasile, E.; Leon, L.G.; Pollina, L.E.; Groen, A.; Falcone, A. MicroRNA-21 in pancreatic cancer: Correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010, 70, 4528–4538. [Google Scholar] [CrossRef]
  94. Moriyama, T.; Ohuchida, K.; Mizumoto, K.; Yu, J.; Sato, N.; Nabae, T.; Takahata, S.; Toma, H.; Nagai, E.; Tanaka, M. MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol. Cancer Ther. 2009, 8, 1067–1074. [Google Scholar] [CrossRef] [Green Version]
  95. Xiong, G.; Huang, H.; Feng, M.; Yang, G.; Zheng, S.; You, L.; Zheng, L.; Hu, Y.; Zhang, T.; Zhao, Y. MiR-10a-5p targets TFAP2C to promote gemcitabine resistance in pancreatic ductal adenocarcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 76. [Google Scholar] [CrossRef] [PubMed]
  96. Chatterjee, A.; Chattopadhyay, D.; Chakrabarti, G. miR-17-5p downregulation contributes to paclitaxel resistance of lung cancer cells through altering beclin1 expression. PLoS ONE 2014, 9, e95716. [Google Scholar] [CrossRef] [PubMed]
  97. Yu, J.; Ohuchida, K.; Mizumoto, K.; Fujita, H.; Nakata, K.; Tanaka, M. MicroRNA miR-17-5p is overexpressed in pancreatic cancer, associated with a poor prognosis, and involved in cancer cell proliferation and invasion. Cancer Biol. Ther. 2010, 10, 748–757. [Google Scholar] [CrossRef] [PubMed]
  98. Garajová, I.; Le Large, T.Y.; Frampton, A.E.; Rolfo, C.; Voortman, J.; Giovannetti, E. Molecular mechanisms underlying the role of microRNAs in the chemoresistance of pancreatic cancer. BioMed. Res. Int. 2014. [Google Scholar] [CrossRef] [PubMed]
  99. Prislei, S.; Martinelli, E.; Mariani, M.; Raspaglio, G.; Sieber, S.; Ferrandina, G.; Shahabi, S.; Scambia, G.; Ferlini, C. MiR-200c and HuR in ovarian cancer. BMC Cancer 2013, 13, 72. [Google Scholar] [CrossRef] [PubMed]
  100. Cochrane, D.R.; Spoelstra, N.S.; Howe, E.N.; Nordeen, S.K.; Richer, J.K. MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol. Cancer Ther. 2009, 8, 1055–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Jayaraj, R.; Kumarasamy, C.; Piedrafita, D. Systematic review and meta-analysis protocol for Fasciola DNA vaccines. Online J. Vet. Res. 2018, 22, 517. [Google Scholar]
  102. Kumarasamy, C.; Devi, A.; Jayaraj, R. Prognostic value of microRNAs in head and neck cancers: A systematic review and meta-analysis protocol. Syst. Rev. 2018, 7, 150. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the literature study process and selection.
Figure 1. Flowchart of the literature study process and selection.
Cancers 11 00900 g001
Figure 2. Commonly performed in vitro assays in the included articles. ISH: in-situ Hybridization; IHC: immuno histo-chemistry; TUNEL: terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling.
Figure 2. Commonly performed in vitro assays in the included articles. ISH: in-situ Hybridization; IHC: immuno histo-chemistry; TUNEL: terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling.
Cancers 11 00900 g002
Figure 3. Hallmarks of PC. The shells highlighted in red are miRNAs that are downregulated while green denotes upregulated in pancreatic cancers in comparison to normal tissue.
Figure 3. Hallmarks of PC. The shells highlighted in red are miRNAs that are downregulated while green denotes upregulated in pancreatic cancers in comparison to normal tissue.
Cancers 11 00900 g003
Figure 4. Forest plot for the HR and 95% CI of PC studies using meta-analysis. Random effect model; I squared = 83.833%; tau = 0.403; Q value = 68.042; df = 11.
Figure 4. Forest plot for the HR and 95% CI of PC studies using meta-analysis. Random effect model; I squared = 83.833%; tau = 0.403; Q value = 68.042; df = 11.
Cancers 11 00900 g004
Figure 5. Forest plot of sub-group analysis for miRNA-21 of the included studies.
Figure 5. Forest plot of sub-group analysis for miRNA-21 of the included studies.
Cancers 11 00900 g005
Figure 6. Funnel plot of studies included in the meta-analysis.
Figure 6. Funnel plot of studies included in the meta-analysis.
Cancers 11 00900 g006
Figure 7. Funnel plot of studies including the imputed studies in our meta-analysis.
Figure 7. Funnel plot of studies including the imputed studies in our meta-analysis.
Cancers 11 00900 g007
Table 1. Characteristics of 43 included studies.
Table 1. Characteristics of 43 included studies.
S.NoAuthorEthnicityPeriod of StudyDrugNo. of Samples (Cancer/Normal)Cell Culture TypeResistant CellsmiRNAmiRNA Profiling PlatformPathways Associated with/Gene
1Yang, R. et al. (2017) [55]Chinese2013–2016GEM157/157Human PC cell lines Capan-2, HPAC, SW1990, PANC-1, CFPAC-1, BXPC-3, ASPC-1, PATU-8988, HPDE6-C7 and HPNEPANC-1-GR and BXPC-3 GR3656Taqman microRNA Reverse Transcription kits (Thermo Fisher Scientific, Dreieich, Germany)Ras Homolog Family Member F (RHOF)/Epithelial–mesenchymal transition (EMT)
2Xiao, G. et al. (2017) [56]Chinese2015–2016GEM15/15HPDE6-C7, PC cell lines Panc-1 and BxPc3NMLet-7aTAKARA PrimeScript KitC-X-C chemokine receptor type 4 (CXCR4)/let-7a/High-mobility group AT-hook 2 (HMGA2)
3Hiramoto, H. et al. (2017) [57]Japanese2000–2011GEM50Panc1, KP4-4, SU.86.86, BxPC3 and MDA-MB-231NM509-5p, 1243 Custom Taqman miRNA Assays kit (Applied Biosystems, San Diego, CA, USA)E-cadherin
4Chaudhary, A.K. et al. (2017) [58]AmericanNMGEMNMHPDEGEM-resistant MIA PaCa-2R cells205-5pSYBR Green-based pathway-focused miScript miRNA PCR Array (catalog number 102ZF, Qiagen, MD, USA) using Roche Light Cycler 480® (Roche, Indianapolis, IN, USA)K-ras, Caveolin-1, and Ki-67
5Liu. F. et al. (2017) [59]ChineseJanuary 2010–December 2014GEM87BxPC-3, Panc-1, Capan-2, SW1990, Paca-2, AsPc-1, and CFPAC-1, HEK293T and HPDE Capan-2, Panc-1, and AsPc-1153SYBR Premix Ex Taq (TaKaRa, Dalian, China) and run with an Applied Biosystems ViiATM 7 Real-Time PCR System (Applied Biosystems)Snail
6Mikamori, M. et al. (2017) [60]JapaneseMarch 2007–August 2015GEM45Panc1, MiaPaCa2, and PSN1 cell linesPanc1-GR1, -GR3, and -GR4 cells155TaqMan MicroRNA Assays (Applied Biosystems) and the ABI7900HT system (Applied Biosystems)Anti-apoptotic (RAB27B)
7Hu, H. et al. (2016) [61]ChineseNMGEM15/15PANC-1NM101TaqMan microRNA assay using ABI-7300 Real-Time machine (Shanghai, China)DNA-dependent protein kinase catalytic subunit (DNA-PKcs)
8Amponsah, P. et al. (2016) [62]DeutschNMGEM92/5ASAN-PaCa, BxPC-3, AsPC-1 and MIA-PaCa2Bx-GEM210Human HT-12 v4 Expression Bead Chip Kit or the Human miR Microarray (Release 19.0)ABCC5
9Li, C. et al. (2016) [63]Chinese2013-2015GEM31/31HPDE6, PANC-1, MIAPaCa-2 and SW1990 cellsNM124TaqMan microRNA assays (Applied Biosystems)miR-124/polypyrimidine tract binding protein 1 (PTBP1)/Pyruvate kinase (PKM2)
10Li, J. et al. (2016) [64]ChineseNMGEM84/20HPC-Y5, AsPC-1, PANC-1, BxPC-3, Hs766t and CFPAC-1NM506Agilent ArraySphingosine kinase 1 (SPHK1)/Protein kinase B(Akt)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)
11Gu, J. et al. (2016) [65]Chinese2008–2010GEM58PanC-1, Mia Paca-2 and HEK-293T NM17-5p SYBR Green Mix (Roche) using All-in-One miRNA qPCR Detection Kit (GeneCopoeia, Rock, MD, USA)Phosphatase and tensin homolog (PTEN)
12Tian, X. et al. (2016) [66]AmericanMarch 2009–September 2013GEM, Lapatinib, and Capecitabine17PANC-1, MIA PaCa-2 and BXCP-3 cell linesNM7, 21, 210, 221RT2 miRNA first strand kit (Qiagen, Inc.) and Applied Biosystems 7900HT Fast Real-Time PCR system (Thermo Fisher Scientifc, Inc., Waltham, MA, USA)Epidermal growth factor receptor (EGFR)1 and human epidermal growth factor receptor (HER)2 pathways
13Fan, P. et al. (2016) [67]DeutschNMGEM21ASAN-PaCa, AsPC-1, PANC-1, MIA-PaCa2 and BxPC-3Bx-GEM101-3p Human HT-12 v4 Expression Bead Chip Kit or the Human miR Microarray (Release 19.0).Ribonucleotide reductase M1 (RRM1)
14Yao, J. et al. (2016) [68]ChineseNMGEM26SW1990 and HEK293T cellsSW1990GZ125aTaqMan MicroRNA Reverse Transcription Kit (Takara), miRscript SYBR Green PCR Kit and SYBR Green PCR Kit (Takara, Dalian, China) TNF Alpha-Induced Protein 3 (A20)
15Ren, Z. et al. (2016) [69]ChineseNMGEM10/10L3.6pl, BxPC-3, CFPAC, MiaPaCa-2, ASPC-1, PANC-1, MPanc96, HPAC, SU86.86 and HS766TNM203mirVana RT-qPCR miRNA Detection kit (cat no. AM7659; Ambion, Austin, TX, USA)Salt-inducible kinase 1 (SIK1)
16Chen, M. et al. (2015) [70]Chinese2008–2011GEM124/10PANC-1 and BXPC3NM181c miRNA-specific TaqMan MiRNA Assay Kit (Applied Biosystems).Mammalian STE20-like protein kinase 1/2 (MST1/2), and large tumour suppressor 1/2 (LATS1/2), together with the adaptor proteins Salvador homolog 1 (SAV1) and MOB kinase activator 1 (MOB1) (Hippo signalling pathway)
17Miyamae, M. et al. (2015) [71]JapaneseJanuary 2010–April 2013GEM94/68PK-45H, PANC-1, PK-59, KP4-1, and PK-1NM550a, 557, 575, 615-5p, 675, 7443D-Gene miRNA microarray platform (Toray Industries, Kamakura, Japan and human TaqMan MicroRNA Assay Kit (Applied Biosystems, Foster City, CA, USA)NM
18Zhang, W. et al. (2015) [72]ChineseNMGEM19HPAC, BxPC-3, Colo357, and L3.6plASPC-1, Panc-1 and MiaPaCa-215b, 155, 212mirVana qRT-PCR miRNA detection kit (Ambion)SMAD specific E3 ubiquitin protein ligase 2 (SMURF2)
19Yu, C. et al. (2015) [73]Chinese2013–20145-FU18 AsPC-1, BxPc-3, Capan-1, Capan-2, CFPAC-1, PANC-1, MIA PaCa-2 & SW1990NM138-5pFluorescence-activated cell sorting (FACSnCanto II flow cytometer; BD Biosciences, San Jose, CA, USA)Vimentin (VIM)
20Liang, C. et al. (2015) [74]Chinese2010–2012GEM 106PCI35 & PCI55, SW1990, MiaPaca-2, PANC-1, BxPC-3, Capan-1 NM33aNMAKT/Gsk-3β/β-catenin pathway
21Liu, Y. et al. (2015) [75]Chinese2007–20105-FU, GEM86/41AsPC-1, BXPC-3, SW1990, MIAPaCa-2, PANC-1 & HPDENM494NMmiR-494/c-Myc/SIRT1 pathway
22Meidhof, S. et al. (2015) [76]DeutschNMGEM 27/27Panc-1, MDA-MB-231, BxPC3, H358, DU-145, hPaca-1 and hPaca-2BxPC3 GEM-resistant cells, Tarceva-resistant H358 cells203Roche LightCycler 480ZEB-1
23Zhao, Y. et al. (2015) [77]DeutschNMGEM28/28L3.6plL3.6pl - GemR21,221miScript SYBR® Green PCR Kit (Qiagen, USA)NM
24Li, Z. et al. (2014) [78]Chinese2013-2014GEM23/23AsPC1, BxPc-3, Capan-1, Capan-2, CFPAC-1, PANC-1, MIA PaCa-2, SW1990NM100TaqMan miRNA Assay (Applied Biosystems)FGFR3
25Xu, J. et al. (2014) [79]ChineseNMGEM 87SW1990, MiaPaCa-2 SW1990/GEM497NMFGF/FGFR signalling pathway
26Hasegawa, S. et al. (2014) [80]Japanese2007-20105-FU, GEM24Panc1-P, Panc1-GR Panc-1GemR1246Comparative CT methodCCNG2
27Lai, I.-L. et al. (2014) [81]AmericansNMGEM NMPanc-1, AsPC-1 and BxPC-3 Panc-1GemR, BxPC3GemR and AsPC-1GemR 520fBio-Rad CFX Manager 2.1 detection system and miScript PCR starter kit (Qiagen)ATM/ATR checkpoint pathway
28Song, W.-F. et al. (2013) [82]Chinese2010–2012GEM 41BxPc3, HPAF, HPAC, Capan, PANC-1 and PL-45 cell linesHPAC and PANC-1/GEM21Specific Taqman MicroRNA assays (Applied Biosystems)PTEN/Akt pathway
29Peng, F. et al. (2013) [83]Chinese2010–20115-FU14TFK-1, QBC939 cell lineNM220b, 200c and 429mirVana miRNA Isolation Kit (Ambion, Austin, TX, USA), Agilent Human miRNA Microarray Kit (V2) (Agilent Inc, Santa Clara, CA, USA) for analysis. SUZ12, ROCK2 direct targets
30Nagano, H. et al. (2013) [84]JapaneseSeptember 1999–February 2004GEM 18MIAPaCa-2, PSN-1, BxPC-3, Panc-1NM29aTRIzol agent (Invitrogen, Carlsbad, CA, USA) Wnt/β-catenin signaling pathway
31Wei, F. et al. (2013) [85]ChineseNMRadiation and AZD8055NMPANC-1, Capan-2, BxPC-3NM99bNMmTOR
32Iwagami, Y. et al. (2013) [86]Japanese1992–2008GEM 66MiaPaCa2 and PSN1 MiaPaCa2-RGs, PSN1-RGs320cNanoDrop ND-1000 spectrophotometer (NanoDrop Technologies, Wilmington, DE, USASMARCC1 mediated anti-cancer effect of GEM
33Bhutia, Y.D. et al. (2013) [87]AmericansNMGEM 10/2MIA PaCa-2L3.6pl and Capan-1/GEMlet-7amiRNA Isolation Kit and the TaqManH MicroRNA Reverse Transcription Kit (Applied Biosystems)RRM2
34Wang, P. et al. (2012) [88]ChineseCohort1: 2003–2005 Cohort2: 2009–2010GEMNMPanc-1, BxPC3NM21RecoverAll Total Nucleic Acid Isolation Kit (Ambion)FasL/Fas pathway
35Singh, S. et al. (2013) [89]NMNMGEMNMMIA PaCa-2MI PaCa-2/GEM7, 146, 205SYBR Green dye universal master mix on a Light Cycler 480 (Roche, Indianapolis)Class III b-tubulin (TUBB3)
36Brabletz, S. et al. (2011) [90]NMNMGSINMPanc1, HPAF2, MCF7, MiaPaCa2 NM200Pfu Ultra Hotstart 2 Master Mix (Stratagene, Santa Clara)Notch signaling
37Ali, S. et al. (2010) [91]NMNMGEM, OHP, tarceva50/10MIAPaCa-2, AsPc-1 MIAPaCa-GR (GEM resistant), AsPc-1OR (oxaliplatin resistant), MIAPaCa-GTR, AsPc-1GTR (GEM and tarceva resistant) 21, 146a, 200b, 200c, 221, let-7b and let-7dTaqMan MicroRNA Assay kit (Applied Biosystems)NM
38Hwang, J.-H. et al. (2010) [92]Korean, Italian1999–2007 and 2001–2004GEM, 5-FU127 BxPc3, HPAF-II, HPAC, PANC1, PL45NM21TaqMan-microRNA assays and the 7900 HT-Fast RealTime PCR (Applied Biosystems, Foster City, CA, USA)NM
39Giovannetti, E. et al. (2010) [93]Deutsch2001–2004GEM 81hTERT-HPNE, Hhs27, LPc006, LPc033, LPc067, LPc111, LPc167, PP437NM217500HT sequence detection system (Applied Biosystems)PTEN and PI3K-Akt pathway
40Moriyama, T. et al. (2009) [94]Japanese2000–2008GEM 25/25AsPC-1, KP-1N, KP-2, KP-3, PANC-1, SUIT-2 MIA PaCa-2, CAPAN-1, CAPAN-2, CFPAC-1, H48N, HS766T, SW1990, NOR-P1 NM21mirVana qRT-PCR miRNA Detection Kit, and mirVana Primer Sets (all from Ambion)VEGF and MMP-2 and MMP-9
41Xiong G et al. (2018) [95]ChineseNMGEM90/90AsPC-1, BxPC-3, MiaPaCa-2, PANC-1, Su86. 86, T3M4AsPC-1-Gem10a-5pGenepharma (Shanghai, China)TFPA2C
42Sun, D. et al. (2018)ChineseJanuary 2007–December 2015GEM87/8BxPC-3, PANC-1, AsPC-1, SW1990, Capan-1, Capan-2, CFPAC-1 and MIA PaCa-2NM374b-5pLightCycler® 480
SYBR-Green I Master (Roche Diagnostics, Basel, Switzerland).
BIAPRC-3 and XIAP
43You, L. et al. (2018)ChineseNMGEM10293T, MIA
PaCa-2, Su.86.86, Capan-1, PANC-1, SW1990, BxPC-3 and AsPC-1
GEM-R cells BxPC-3 and PANC-11207(Bio-Rad, Hercules, CA, USA)PVT1
NM-Not Mentioned.
Table 2. miRNAs that are involved in chemoresistance and pathways that are modulated.
Table 2. miRNAs that are involved in chemoresistance and pathways that are modulated.
Chemoresistance
DownregulatedUpregulated
miRNAPathwaymiRNAPathway
GEM GEM
210ABCC517-5pPTEN
124PTBP1/PKM2221EGFR and HER pathways
101-3pRRM1203SIK1
100FGFR3181cMST1/2, and LATS1/2, together with the adaptor proteins SAV1 and MOB1 (Hippo signalling pathway)
497FGF/FGFR signalling pathway15bSMURF2
7TUBB321NM
205TUBB3221NM
374b-5pBIAPRC-3 and XIAP1246CCNG2
5-FU 21PTEN/Akt pathway
200cSUZ12, ROCK2 direct targets320cSMARCC1 mediated the anti-cancer effect of GEM
220bSUZ12, ROCK2 direct targets155Anti-apoptotic (RAB27B)
21NM
221NM
21VEGF and MMP-2 and MMP-9
10a-5pTFAP2C
5-FU
1246CCNG2
Table 3. miRNAs that are involved in chemosensitivity and pathways that are modulated.
Table 3. miRNAs that are involved in chemosensitivity and pathways that are modulated.
Chemosensitivity
DownregulatedUpregulated
miRNAPathwaymiRNAPathway
GEM GEM
3656RHOF/EMT509-5pE-cadherin
Let-7aCXCR4/HMGA21243E-cadherin
205-5pK-ras, Caveolin-1 and Ki-6733aAKT/Gsk-3β/β-catenin pathway
153SNAIL21FasL/Fas pathway
101DNA-PKcs1207PVT1
506SPHK1/Akt/NF-κB5-FU
494c-Myc/SIRT1 pathway138-5pvimentin
203ZEB-1
5-FU
494c-Myc/SIRT1 pathway
Table 4. Publication bias of the included studies.
Table 4. Publication bias of the included studies.
GroupsClinical OutcomesClassic Fail-Safe NOrwin Fail-Safe NBegg and MazumdarEgger′s RegressionDual and Tweedie (Random Effects)
z-Valuep-ValueHR in ObservedTauz-Valuep-ValueInterceptp-ValuedfObservedq-ValueAdjustedq-Value
MainMain Meta-analysis6.26401.1500.2571.1650.2431.7780.052101.60368.0411.132107.980
FixedMixed/RandomHypothesis Test
SubgroupsHeterogeneityHR95% CIHR95% CIFixed effects modelRandom effects model
QPI2LowHighLowHighZPStudiesZPStudies
miR-211.6830.19540.5671.9131.3402.7322.0611.1953.5563.569022.5990.0092

Share and Cite

MDPI and ACS Style

Madurantakam Royam, M.; Ramesh, R.; Shanker, R.; Sabarimurugan, S.; Kumarasamy, C.; Ramesh, N.; Gothandam, K.M.; Baxi, S.; Gupta, A.; Krishnan, S.; et al. miRNA Predictors of Pancreatic Cancer Chemotherapeutic Response: A Systematic Review and Meta-Analysis. Cancers 2019, 11, 900. https://doi.org/10.3390/cancers11070900

AMA Style

Madurantakam Royam M, Ramesh R, Shanker R, Sabarimurugan S, Kumarasamy C, Ramesh N, Gothandam KM, Baxi S, Gupta A, Krishnan S, et al. miRNA Predictors of Pancreatic Cancer Chemotherapeutic Response: A Systematic Review and Meta-Analysis. Cancers. 2019; 11(7):900. https://doi.org/10.3390/cancers11070900

Chicago/Turabian Style

Madurantakam Royam, Madhav, Rithika Ramesh, Ritika Shanker, Shanthi Sabarimurugan, Chellan Kumarasamy, Nachimuthu Ramesh, Kodiveri Muthukalianan Gothandam, Siddharta Baxi, Ajay Gupta, Sunil Krishnan, and et al. 2019. "miRNA Predictors of Pancreatic Cancer Chemotherapeutic Response: A Systematic Review and Meta-Analysis" Cancers 11, no. 7: 900. https://doi.org/10.3390/cancers11070900

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop