Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Cancer Genomics & Proteomics
    • Anticancer Research
    • In Vivo

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Cancer Genomics & Proteomics
  • Other Publications
    • Cancer Genomics & Proteomics
    • Anticancer Research
    • In Vivo
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Cancer Genomics & Proteomics

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Research ArticleArticle
Open Access

An miRNA Signature Predicts Grading of Pancreatic Neuroendocrine Neoplasms

JAMES SALLER, DALEY WHITE, BROOKE HOUGH, SEAN YODER, JUNMIN WHITING, DUNG-TSA CHEN, ANTHONY MAGLIOCCO and DOMENICO COPPOLA
Cancer Genomics & Proteomics March 2023, 20 (2) 154-164; DOI: https://doi.org/10.21873/cgp.20370
JAMES SALLER
1Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DALEY WHITE
2Department of Biomedical Library, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
BROOKE HOUGH
1Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SEAN YODER
3Molecular Genomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JUNMIN WHITING
4Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DUNG-TSA CHEN
4Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ANTHONY MAGLIOCCO
5Protean Biodiagnostics, Orlando, FL, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOMENICO COPPOLA
1Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.;
6Department of Pathology Florida Digestive Health Specialists, Lakewood Ranch, FL, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: domenico.coppola@fdhs.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Grading pancreatic neuroendocrine neoplasms (PNENs) via mitotic rate and Ki-67 index score is complicated by interobserver variability. Differentially expressed miRNAs (DEMs) are useful for predicting tumour progression and may be useful for grading. Patients and Methods: Twelve PNENs were selected. Four patients had grade (G) 1 pancreatic neuroendocrine tumours (PNETs); 4 had G2 PNETs; and 4 had G3 PNENs (2 PNETs and 2 pancreatic neuroendocrine carcinomas). Samples were profiled using the miRNA NanoString Assay. Results: There were 6 statistically significant DEMs between different grades of PNENs. MiR1285-5p was the sole miRNA differentially expressed (p=0.03) between G1 and G2 PNETs. Six statistically significant DEMs (miR135a-5p, miR200a-3p, miR3151-5p, miR-345-5p, miR548d-5p and miR9-5p) (p<0.05) were identified between G1 PNETs and G3 PNENs. Finally, 5 DEMs (miR155-5p, miR15b-5p, miR222-3p, miR548d-5p and miR9-5p) (p<0.05) were identified between G2 PNETs and G3 PNENs. Conclusion: The identified miRNA candidates are concordant with their patterns of dysregulation in other tumour types. The reliability of these DEMs as discriminators of PNEN grades support further investigations using larger patient populations.

Key Words
  • microRNA
  • miRNA profiling
  • neuroendocrine
  • grading

The most recent grading system for pancreatic neuroendocrine neoplasms (PNENs) was informed by the 2017 WHO classification, which uses quantification of the Ki-67 proliferative index and mitotic rate as the 2 principal metrics for grading pancreatic neuroendocrine tumours (PNETs). This updated classification further refines grade (G) 3 as a category that is distinctly comprised of either well differentiated neuroendocrine tumours (NETs) or poorly differentiated neuroendocrine carcinomas (1-4). This clarification of G3 is corroborated by studies that used next-generation sequencing to demonstrate that there are distinct genomic alterations that distinguish well differentiated G3 NETs from poorly differentiated G3 neuroendocrine carcinomas (5-7).

Given the interobserver variability in determining Ki-67 proliferative index scores and mitotic rates, reliance upon these metrics could potentially complicate the assignment of a PNEN’s grade, especially for cases with a Ki-67 proliferative index score or mitotic rate that is at the borderline between grades. Moreover, mimickers, which can include apoptotic bodies, lymphocytes, and darkly stained nuclei, can confound determination of true mitotic figures and/or Ki-67–positive tumour cells and can further complicate cases that do not have a clear-cut grade (8).

Understandably, recent investigative efforts have evaluated ancillary methods to assist in improving the accuracy of grade assignment and interobserver reproducibility. For example, phosphorylated histone H3 (pHH3) has been used as a mitotic marker for mitotic rate assessment, and double-staining for Ki-67 with CD45 has been used to exclude lymphocytes as mimickers (9-15).

Studies have shown the utility of a profile or signature of microRNAs (miRNAs) and demonstrate that differentially expressed miRNAs (DEMs) can be used to distinguish between lower and higher grades of cancer (16-18). There have also been previous studies focused on using DEMs to characterize NETs (19-24). For instance, an miRNA expression profile identified significant DEMs that can differentiate types of neuroendocrine lung neoplasms (including typical carcinoids, atypical carcinoids, large-cell neuroendocrine lung cancer and small-cell lung cancer) (20). DEMs have been identified as prognostic predictors that effectively distinguish lower-grade (G1) and higher-grade (G2/3) NETs of the small intestine (19, 25). There have also been preliminary studies comparing DEMs from PNETs to DEMs from background pancreatic islets, as well as studies focused on determining candidate DEMs with prognostic value (26-28).

Given the precedent of previous studies that have used the NanoString platform to reliably identify DEMs for different tumour types (29, 30), we used the NanoString assay to identify DEMs that may be useful in differentiating PNETs by grade.

Patients and Methods

Patient selection. This retrospective study was approved by the Moffitt Cancer Center Institutional Review Board (IRB). Twelve patients with PNENs of different grades were selected from the pathology repository at Moffitt Cancer Center. A retrospective chart review was performed using PowerChart/PathNet.

Pathologic evaluation of samples. The relevant haematoxylin and eosin–(H&E-) stained slides for all selected cases underwent a comprehensive histopathologic review by 2 pathologists (DC and JS) to confirm their diagnoses. To achieve the highest possible tissue purity for each specimen, we excluded (via macrodissection) the other tissue components on the slide/block before the sample was submitted for NanoString analyses. The tissue collection and review processes were regulated by specific standard operating procedures and quality assurance/quality control (QA/QC) protocols. We juxtaposed the marked glass slides to the corresponding paraffin block, and using a scalpel, we macrodissected the marked PNEN area. The samples were submitted to the Moffitt molecular pathology laboratory, where RNA was extracted and subjected to NanoString analysis, as described below.

NanoString. The samples were profiled using the miRNA NanoString Assay (NanoString Technologies, Seattle, WA, USA). The NanoString procedures were performed as previously described (31). In brief, 100 ng of RNA was extracted from each formalin-fixed paraffin-embedded (FFPE) block and used as input. Mature miRNAs were ligated to a species-specific tag sequence (miRtag) using a thermally controlled splinted ligation. Unligated miRtags were removed with enzymatic purification, and miR-tagged mature miRNAs were then hybridized with an nCounter Human (V2) miRNA Expression Assay CodeSet (NanoString Technologies) overnight at 65° C. The unhybridized CodeSet was removed with automated purification, which was performed on an nCounter Prep Station (NanoString Technologies), and the remaining target:probe complexes were transferred and bound to an imaging surface, as previously described. Counts of the reporter probes were tabulated for each sample by the nCounter Digital Analyzer (NanoString Technologies), and raw data output was imported into nSolver (http://www.nanostring.com/products/nSolver) (32). The nSolver procedures have been previously described (31); in brief, positive control probes in the CodeSet were tested for their linearity with a correlation between the concentration of the added target and the resulting count; correlation ≥0.95 indicated high-quality data. The limit of detection for each assay was confirmed using positive and negative controls; the positive control (Pos-E, 0.5fM) was above the average of the negative control means (NanoString Technologies I. nCounter Expression Data Analysis Guide) (33). Data was normalized based on housekeeping genes (B2M, GAPDH, RPL19, ACTB and RPLP0) and transformed into log2 scale.

Statistical analysis. The normalized miRNA data were analysed using SAS and R software. Wilcoxon rank sum test or Kruskal-Wallis test were performed on 800 measured miRNAs to identify the most commonly differentially expressed miRNAs between different grades of PNENs. Further analyses were performed to identify miRNAs that had statistically significant differential expression between specific grades of PNENs. Due to the small sample size (n=12), the analysis was for exploratory purposes and not intended for correction of multiple testing.

Results

Of all 12 patients with PNENs, 4 had G1 PNETs, 4 had G2 PNETs, and 4 had G3 PNENs, of which 2 were PNETs and 2 were pancreatic neuroendocrine carcinomas. Seven patients were male (58%), and the average patient age was 62.6 years (range=40-85 years). Eight PNENs involved the pancreatic tail, 2 involved the pancreatic head, 1 involved the body of the pancreas, and 1 was non-specified. The average PNEN tumour size was 3.89 cm (range=1.7-7.5 cm).

Of the 8 G1/2 PNETs, 1 G1 PNET was stage pT1, 3 were pT2, and 4 were pT3. All 4 G3 PNENs were stage pT3. Two patients with G2 PNETs had previously received neoadjuvant therapy, whereas the 10 remaining patients received no presurgical therapy. Two of the 8 G1/2 PNETs were metastatic, and 1 of the 4 G3 PNENs was metastatic. All 8 of the G1/2 PNETs had negative margins, and 2 of the 4 G3 PNENs had negative margins.

We identified miRNAs that had statistically significant differential expression between specific grades of PNENs; additionally, we identified miRNAs that had statistically significant differential expression consistently observed between all 3 grades of PNENs. Overall, there were 9 miRNAs (miR1285-5p, miR222-3p, miR200a-3p, miR3151-5p, miR15b-5p, miR155-5p, miR-345-5p, miR548d-5p and miR9-5p) that had a statistically significant differential expression (p<0.05) between different grades of PNENs. Of these, there were 6 statistically significant DEMs (miR1285-5p, miR15b-5p, miR155-5p, miR-345-5p, miR548d-5p, and miR9-5p) (p<0.05) between different grades of PNENs (G1/2/3) (Table I, Table II, Table III, and Table IV).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Comparison of miRNAs with statistically significant differential expression between grade 1, 2 and 3 pancreatic neuroendocrine neoplasms (NENs).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Comparison of miRNAs with statistically significant differential expression between grade 1 and 2 pancreatic neuroendocrine tumours (NETs).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table III.

Comparison of miRNAs with statistically significant differential expression between grade 1 and 3 pancreatic neuroendocrine neoplasms (NENs).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table IV.

Comparison of miRNAs with statistically significant differential expression between grade 2 and 3 pancreatic neuroendocrine neoplasms (NENs).

MiR1285-5p was the sole miRNA with a statistically significant differential expression (p=0.03) between G1 and G2 PNETs. Six statistically significant DEMs (miR135a-5p, miR200a-3p, miR3151-5p, miR-345-5p, miR548d-5p, and miR9-5p) (p<0.05) differentiated G1 PNETs from G3 PNENs (Figure 1 and Figure 2). Finally, 5 miRNAs (miR155-5p, miR15b-5p, miR222-3p, miR548d-5p, and miR9-5p) demonstrated a statistically significant differential expression (p<0.05) between G2 PNETs and G3 PNENs (Figure 3). MiR9-5p and miR548d-5p were the only 2 miRNAs that consistently maintained statistically significant differential expression (p<0.05) when compared across 3 separate groups (G1 and G3 PNENs, G2 and G3 PNENs, and G1, G2 and G3 PNENs). MiR9-5p and miR548d-5p also demonstrated p-values that were recurrently lower than the majority of the other miRNAs that they were compared with. MiR15b-5p and miR155-5p maintained statistically significant differential expression (p<0.05) when compared across G2 and G3 PNENs and G1, G2 and G3 PNENs. miR-345-5p maintained statistically significant differential expression (p<0.05) when compared across G1 and G3 PNENs and G1, G2 and G3 PNENs. 3 miRNAs (miR3151-5p, miR488-3p and miR33a-5p) had p>0.05 but were noteworthy because of their significant degree of consecutive down-regulation with increasing PNET grades.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Comparison of miRNAs with statistically significant differential expression between grade 1, 2 and 3 pancreatic neuroendocrine neoplasms (PNENs). miR9-5p and miR548d-5p were the most consistent in statistically significant differential expression (p<0.05) among comparisons across different grades of PNENs. Please note that miRNA data are normalized using log2-scale gene expression.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Comparison of miRNAs with statistically significant differential expression between grade 1 and 3 pancreatic neuroendocrine neoplasms (PNENs). miR9-5p and miR548d-5p were the most consistent in statistically significant differential expression (p<0.05) among comparisons across different grades of PNENs. Please note that miRNA data are normalized using log2-scale gene expression.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Comparison of miRNAs with statistically significant differential expression between grade 2 and 3 pancreatic neuroendocrine neoplasms (PNENs). miR9-5p and miR548d-5p were the most consistent in statistically significant differential expression (p<0.05) among comparisons across different grades of PNENs. Please note that miRNA data are normalized using log2-scale gene expression.

The 24 miRNAs in Table I, Table II, Table III, and Table IV were further evaluated for survival association in pancreatic cancer using Oncomir (http://www.oncomir.org/oncomir/survival_custom.html). Two miRs showed a p-value <0.05 by log-rank test: miR-33a-5p (p=0.037) and miR-488-3p (p=0.002) based on median cut-off of miRNA expression. Specifically, patients with higher miR-33a-5p or miR-488-3p expression had better overall survival compared to those with lower expression, suggesting potential tumour suppressive roles with higher expression for improved survival (Figure 4 and Figure 5).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Kaplan–Meier plot of overall survival for miR-33a-5p in pancreatic adenocarcinoma patients and the cohort with a score below median, labelled as low-expression. The figure was generated using the KM Plotter Online Tool.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Kaplan–Meier plot of overall survival for miR-488-3p in pancreatic adenocarcinoma patients and the cohort with a score below median was labelled as low-expression. The figure was generated using the KM Plotter Online Tool.

Discussion

There has been an evolving appreciation of the importance and diversity of the regulatory roles that non-coding RNAs (ncRNAs) play in normal cellular function and the consequences of ncRNA dysregulation in cancer development (34, 35). MiRNAs represent a significant portion of ncRNAs and have been shown to play a role in oncogenesis and metastasis in the context of transcriptomic dysregulation (36-40).

Previous studies have demonstrated that some miRNAs have behaviour that is oncogenic when up-regulated (i.e., oncomirs) or down-regulated (i.e., tumour-suppressive anti-oncomirs). Some miRNAs can function as either tumour-suppressive or oncogenic depending on which tumour type they are found to be dysregulated in. MiRNAs are generally non-stringent regarding the targets that they bind to; consequently, it is not uncommon for an miRNA that predominantly functions as an oncomir in most contexts to occasionally serve as an anti-oncomir. Conversely, an anti-oncomir in most contexts could occasionally serve as an oncomir (41).

Previous studies have characterized DEMs within tumour types as a basis for miRNA profiles that may have potential clinical utility (42). Likewise, the goal of our study was to aid in the development of an miRNA profile that has potential clinical utility for accurately grading PNENs.

Candidate miRNAs. Candidate miRNAs were identified from the miRNA expression profiles of 800 miRNAs. miR9-5p and miR548d-5p most consistently maintained significant differential expression across the 3 groups of comparisons. MiR-345-5p, miR15b-5p and miR155-5p were the next most consistently maintained, with statistically significant differential expression in 2 groups.

miR9-5p. MiR9-5p consistently functioned as an oncomir in our study, with consecutively up-regulated expression with each increase in PNEN grade. MiR-9-5p up-regulation in breast cancer has been shown to affect FOXO1, LIFR, and E-Cadherin expression, inducing resistance to therapy with alkylating agents (43, 44) and tamoxifen (43-45) and consequently increasing metastatic potential and negatively affecting patient outcomes. miR-9-5p up-regulation also correlates with increased metastatic potential and tumour recurrence of head and neck cancer (45, 46).

miR548d-5p. MiR548d-5p functioned as an anti-oncomir in our study, with consecutively down-regulated expression with each increase in PNEN grade. The results of previous studies corroborate that most members of the miR548 family typically behave in a tumour-suppressive manner (47-49). MiR548d-5p also has been proposed as a potential biomarker for responsiveness to neoadjuvant chemoradiotherapy (50). Preclinical data putatively support miR548d-5p having an inhibitory effect on PTPN12 (51). MiR548d-5p behaves as an anti-oncomir in pancreatic cancer, inhibiting growth and enhancing sensitization to gemcitabine (52). Interestingly, an miRNA that is homologous to miR548d-5p has been reported to be located within the fourth intron within the tumour suppressor gene FHIT (53).

miR-345-5p. MiR-345-5p likewise functioned as an anti-oncomir in our study, with consecutively down-regulated expression with each increase in PNEN grade. MiR-345-5p down-regulation in non–small cell lung cancer was associated with progression and poor prognosis (54). It has been shown that up-regulation of miR-345-5p in gastric cancer affects the migration and metastatic potential of gastric cancer cells by modulating FOXQ1 expression and epithelial-the mesenchymal transition (55). MiR-345-5p down-regulation in pancreatic cancer has shown to reduce apoptosis both through activation of caspase-dependent and caspase-independent pathways and by modulating BCL2 (56). In another study MiR345,5p was down-regulated and caused suppression of pancreatic cancer proliferation and metastasis by targeting CCL8 and NF-kB (57).

MiR-345-5p inhibits proliferation and metastatic potential of colon cancer cells by targeting BAG3 and is shown to be down-regulated in colorectal cancer (58). It is also down-regulated in hepatocellular carcinoma, where down-regulation correlates with a significantly decreased overall survival rate, and targets YAP1 (59). MiR-345-5p suppresses proliferation, migration, and invasion of prostate cancer cells by targeting SMAD1. Alternatively, miR-345-5p promotes oncogenic growth and migration of castration-resistant prostate cancer cells by inhibiting the tumour-suppressive activity of CDKN1A (60, 61).

miR15b-5p. MiR15b-5p functioned as an oncomir in our study, with up-regulated expression with the overall comparison of G1 PNETs to G3 PNENs, though down-regulation was observed when comparing G2 with G1. These findings are concordant with studies showing that up-regulation of miR15b-5p in gastric cancer promotes proliferation, migration, invasion, epithelial-mesenchymal transition, and metastasis by targeting the tumour suppressor gene PAQR3 (62, 63). Up-regulation of miR-15b in colorectal cancer down-regulates MTSS1 and Klotho proteins, correlating with metastasis, recurrence, and poor patient prognosis (64). Low miR-15b expression suppresses cell growth and induces apoptosis of hepatocellular carcinoma cells; improved overall survival rates were shown in patients with low miR-15b expression. Moreover, up-regulated miR-15b is a predictor of a worse prognoses for patients with hepatocellular carcinoma after curative hepatectomy (65). MiR-15b-5p was also found to be up-regulated in prostate cancer and exerts a tumorigenic effect by targeting the tumour suppressor gene RECK and facilitating tumour recurrence (66). MiR-15b-5p is also up-regulated in the transformation from classical to aggressive Mantle cell lymphoma (67) and in squamous cell carcinoma of the head and neck (68).

miR155-5p. MiR155-5p functioned as an oncomir in our study, with up-regulated expression with the overall comparison of G1 PNETs to G3 PNENs. Similar to miR15b-5p, miR155-5p was down-regulated when comparing G2 to G1.

MiR-155-5p has been described as a hypoxia-inducible oncomir that is mediated by HIF1α, wherein miR-155-5p down-regulates ELK3 (69). In colorectal cancer, miR-155-5p up-regulation is correlated with poor prognoses, and miR-155-5p posttranscriptional regulation of human antigen R promotes migration of colorectal cancer cells (70). Cancer cells also produce miR155 to convert normal fibroblasts to cancer associated fibroblasts (71) contributing to the EMT associated phenotype (72). Increased expression of miR-155 by pancreatic cancer was found to be an early event in the transition from non-neoplastic pancreatic tissue to intraductal pancreatic mucinous neoplasm (IPMN), a precursor of pancreatic adenocarcinoma. In their study, Hobbe et al. found such an increase in miR-155 to be present not only in pancreatic IPMN tissue but also in the fluid derived by pancreatic cysts harbouring IPMN. The authors suggested that miR-155 may be used as a biomarker to identify IPMN (73). MiR155-5p up-regulation in breast cancer is significantly decreased following surgical resection (74). A recent preclinical study demonstrated that inhibition of miR-155-5p subsequently prevented the formation of breast cancer stem cells (75). MiR-155-5p up-regulation in renal carcinoma promotes proliferation and invasion via inhibition of glycogen synthase kinase-3β (GSK-3β), which in turn up-regulates the Wnt/β-catenin signalling pathway (76). In diffuse large B-cell lymphoma, up-regulation inhibits SOCS3, which suppresses apoptosis and up-regulates the JAK-STAT3 signalling pathway (77). In a murine model, inhibiting miR-155-5p delayed diffuse large B-cell lymphoma progression (78).

MiR-155-5p can either up-regulate or down-regulate the mTOR pathway depending on the tumour type. In osteosarcoma, miR-155-5p inhibits PTEN, consequently up-regulating the mTOR pathway (79). Conversely, a recent study reported tumour-suppressive behaviour of miR-155-5p via inhibition of PDK1 and subsequent down-regulation the mTOR pathway in cervical cancer (80). MiR-155-5p down-regulation has also been shown to be tumour-suppressive by sensitizing gastric cancer cells to cisplatin (81). MiR-155-5p may have implications for immunotherapy, as it suppresses expression of programmed death ligand-1 (PD-L1) (82).

miR3151-5p, miR488-3p, and miR33a-5p. MiR3151-5p, miR488-3p, and miR33a-5p had p-values >0.05 and demonstrated consecutive down-regulation with increasing PNEN grade. A review of corroborating studies support potential tumour suppressive roles of all 3 of these miRNAs, including miR3151-5p down-regulation in chronic lymphocytic leukaemia; miR488-3p down-regulation in NETs of the small intestine and in gliomas; and miR33a-5p down-regulation in lung adenocarcinoma, osteosarcoma, melanoma, and prostate cancer (25, 83, 84).

It is also interesting that when all of the 24 miRNAs in Table I, Table II, Table III, and Table IV were evaluated for survival association in pancreatic adenocarcinoma using Oncomir (http://www.oncomir.org/oncomir/survival_custom.html), two miRs showed a p-value <0.05 by log-rank test: miR-33a-5p (p=0.037) and miR-488-3p (p=0.002), based on median cut-off of miRNA expression. Specifically, patients harbouring pancreatic ductal adenocarcinoma with higher miR-33a-5p or miR-488-3p expression had better overall survival compared to those with lower expression, suggesting potential tumour suppressive roles of these miRNA across different pancreatic tumour types.

Conclusion

The observed differential expression of the miRNAs in our study are concordant with patterns of dysregulation that have been established in prior studies of different tumour types. The aforementioned miRNA targets that have been reported in the literature may serve as a reference for investigating potential targets that may be responsible for the dysregulation observed in different grades of PNENs.

The reliability of these candidate miRNAs with statistically significant differential expression between PNEN grades supports further investigation in a larger population of patients, with the goal of identifying potential prognostic utility by providing clarity for PNENs with significant grading challenges. Furthermore, if future studies further corroborate the reliability of these candidate miRNAs for PNEN grading, a profile comprised of these miRNAs may have potential as an independent prognostic biomarker to accurately grade PNENs.

Acknowledgements

This work has been supported in part by the Departments of Pathology, Biostatistics & Bioinformatics, and the Tissue Core and the Molecular Genomics Core at the H. Lee Moffitt Cancer Center & Research Institute, a comprehensive cancer designated by the National Cancer Institute and funded in part by Moffitt’s Cancer Center Support Grant (P30-CA076292).

Footnotes

  • Conflicts of Interest

    The Authors have no conflicts of interest.

  • Authors’ Contributions

    Dr. James Saller collected the samples, He also collated the data and drafted the manuscript. Daley White contributed to the interpretation of data as well as drafting and critical revision of the manuscript; Brooke Hough helped with the literature search and review. Dr. Sean Yoder performed the NanoString analysis and evaluated the results, Drs. Junmin Whiting and Dung-Tsa Chen performed the statistical analysis. Dr. Anthony Magliocco supervised the analysis and critically reviewed the data. He also reviewed the final form of the manuscript. Dr. Domenico Coppola designed the study, critically evaluated the data, and reviewed the final form of the manuscript.

  • Received October 24, 2022.
  • Revision received January 11, 2023.
  • Accepted January 13, 2023.
  • Copyright © 2023, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Inzani F,
    2. Petrone G and
    3. Rindi G
    : The New World Health Organization Classification for pancreatic neuroendocrine neoplasia. Endocrinol Metab Clin North Am 47(3): 463-470, 2018. PMID: 30098710. DOI: 10.1016/j.ecl.2018.04.008
    OpenUrlCrossRefPubMed
    1. McCall CM,
    2. Shi C,
    3. Cornish TC,
    4. Klimstra DS,
    5. Tang LH,
    6. Basturk O,
    7. Mun LJ,
    8. Ellison TA,
    9. Wolfgang CL,
    10. Choti MA,
    11. Schulick RD,
    12. Edil BH and
    13. Hruban RH
    : Grading of well-differentiated pancreatic neuroendocrine tumors is improved by the inclusion of both Ki67 proliferative index and mitotic rate. Am J Surg Pathol 37(11): 1671-1677, 2013. PMID: 24121170. DOI: 10.1097/PAS.0000000000000089
    OpenUrlCrossRefPubMed
    1. Hasegawa T,
    2. Yamao K,
    3. Hijioka S,
    4. Bhatia V,
    5. Mizuno N,
    6. Hara K,
    7. Imaoka H,
    8. Niwa Y,
    9. Tajika M,
    10. Kondo S,
    11. Tanaka T,
    12. Shimizu Y,
    13. Kinoshita T,
    14. Kohsaki T,
    15. Nishimori I,
    16. Iwasaki S,
    17. Saibara T,
    18. Hosoda W and
    19. Yatabe Y
    : Evaluation of Ki-67 index in EUS-FNA specimens for the assessment of malignancy risk in pancreatic neuroendocrine tumors. Endoscopy 46(1): 32-38, 2014. PMID: 24218309. DOI: 10.1055/s-0033-1344958
    OpenUrlCrossRefPubMed
  2. ↵
    1. Fazio N and
    2. Milione M
    : Heterogeneity of grade 3 gastroenteropancreatic neuroendocrine carcinomas: New insights and treatment implications. Cancer Treat Rev 50: 61-67, 2016. PMID: 27636009. DOI: 10.1016/j.ctrv.2016.08.006
    OpenUrlCrossRefPubMed
  3. ↵
    1. Jiao Y,
    2. Shi C,
    3. Edil BH,
    4. de Wilde RF,
    5. Klimstra DS,
    6. Maitra A,
    7. Schulick RD,
    8. Tang LH,
    9. Wolfgang CL,
    10. Choti MA,
    11. Velculescu VE,
    12. Diaz LA Jr.,
    13. Vogelstein B,
    14. Kinzler KW,
    15. Hruban RH and
    16. Papadopoulos N
    : DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331(6021): 1199-1203, 2011. PMID: 21252315. DOI: 10.1126/science.1200609
    OpenUrlAbstract/FREE Full Text
    1. Glenn ST,
    2. Jones CA,
    3. Sexton S,
    4. LeVea CM,
    5. Caraker SM,
    6. Hajduczok G and
    7. Gross KW
    : Conditional deletion of p53 and Rb in the renin-expressing compartment of the pancreas leads to a highly penetrant metastatic pancreatic neuroendocrine carcinoma. Oncogene 33(50): 5706-5715, 2014. PMID: 24292676. DOI: 10.1038/onc.2013.514
    OpenUrlCrossRefPubMed
  4. ↵
    1. Yachida S,
    2. Vakiani E,
    3. White CM,
    4. Zhong Y,
    5. Saunders T,
    6. Morgan R,
    7. de Wilde RF,
    8. Maitra A,
    9. Hicks J,
    10. Demarzo AM,
    11. Shi C,
    12. Sharma R,
    13. Laheru D,
    14. Edil BH,
    15. Wolfgang CL,
    16. Schulick RD,
    17. Hruban RH,
    18. Tang LH,
    19. Klimstra DS and
    20. Iacobuzio-Donahue CA
    : Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol 36(2): 173-184, 2012. PMID: 22251937. DOI: 10.1097/PAS.0b013e3182417d36
    OpenUrlCrossRefPubMed
  5. ↵
    1. Boiesen P,
    2. Bendahl PO,
    3. Anagnostaki L,
    4. Domanski H,
    5. Holm E,
    6. Idvall I,
    7. Johansson S,
    8. Ljungberg O,
    9. Ringberg A,
    10. Ostberg G and
    11. Fernö M
    : Histologic grading in breast cancer – reproducibility between seven pathologic departments. South Sweden Breast Cancer Group. Acta Oncol 39(1): 41-45, 2000. PMID: 10752652. DOI: 10.1080/028418600430950
    OpenUrlCrossRefPubMed
  6. ↵
    1. Voss SM,
    2. Riley MP,
    3. Lokhandwala PM,
    4. Wang M and
    5. Yang Z
    : Mitotic count by phosphohistone H3 immunohistochemical staining predicts survival and improves interobserver reproducibility in well-differentiated neuroendocrine tumors of the pancreas. Am J Surg Pathol 39(1): 13-24, 2015. PMID: 25353284. DOI: 10.1097/PAS.0000000000000341
    OpenUrlCrossRefPubMed
    1. Villani V,
    2. Mahadevan KK,
    3. Ligorio M,
    4. Fernández-Del Castillo C,
    5. Ting DT,
    6. Sabbatino F,
    7. Zhang I,
    8. Vangel M,
    9. Ferrone S,
    10. Warshaw AL,
    11. Lillemoe KD,
    12. Wargo J,
    13. Deshpande V and
    14. Ferrone CR
    : Phosphorylated histone H3 (PHH3) is a superior proliferation marker for prognosis of pancreatic neuroendocrine tumors. Ann Surg Oncol 23(Suppl 5): 609-617, 2016. PMID: 27020585. DOI: 10.1245/s10434-016-5171-x
    OpenUrlCrossRefPubMed
    1. Kim MJ,
    2. Kwon MJ,
    3. Kang HS,
    4. Choi KC,
    5. Nam ES,
    6. Cho SJ,
    7. Park HR,
    8. Min SK,
    9. Seo J,
    10. Choe JY and
    11. Park HC
    : Identification of phosphohistone H3 cutoff values corresponding to original WHO grades but distinguishable in well-differentiated gastrointestinal neuroendocrine tumors. Biomed Res Int 2018: 1013640, 2018. PMID: 29780816. DOI: 10.1155/2018/1013640
    OpenUrlCrossRefPubMed
    1. van der Loos CM,
    2. de Boer OJ,
    3. Mackaaij C,
    4. Hoekstra LT,
    5. van Gulik TM and
    6. Verheij J
    : Accurate quantitation of Ki67-positive proliferating hepatocytes in rabbit liver by a multicolor immunohistochemical (IHC) approach analyzed with automated tissue and cell segmentation software. J Histochem Cytochem 61(1): 11-18, 2013. PMID: 22941418. DOI: 10.1369/0022155412461154
    OpenUrlCrossRefPubMed
    1. Kim JY,
    2. Jeong HS,
    3. Chung T,
    4. Kim M,
    5. Lee JH,
    6. Jung WH and
    7. Koo JS
    : The value of phosphohistone H3 as a proliferation marker for evaluating invasive breast cancers: A comparative study with Ki67. Oncotarget 8(39): 65064-65076, 2017. PMID: 29029412. DOI: 10.18632/oncotarget.17775
    OpenUrlCrossRefPubMed
    1. Murphy CE,
    2. McCormick KA,
    3. Shankaran V,
    4. Reddi DM,
    5. Swanson PE,
    6. Upton MP,
    7. Papanicolau-Sengos A,
    8. Khor S and
    9. Westerhoff M
    : Grade assignment by Ki-67 proliferative index, mitotic count, and phosphohistone H3 count in surgically resected gastrointestinal and pancreatic neuroendocrine tumors. Pancreas 46(10): 1359-1365, 2017. PMID: 29040195. DOI: 10.1097/MPA.0000000000000923
    OpenUrlCrossRefPubMed
  7. ↵
    1. Tracht J,
    2. Zhang K and
    3. Peker D
    : Grading and prognostication of neuroendocrine tumors of the pancreas: a comparison study of Ki67 and PHH3. J Histochem Cytochem 65(7): 399-405, 2017. PMID: 28651471. DOI: 10.1369/0022155417708186
    OpenUrlCrossRefPubMed
  8. ↵
    1. Catto JW,
    2. Miah S,
    3. Owen HC,
    4. Bryant H,
    5. Myers K,
    6. Dudziec E,
    7. Larré S,
    8. Milo M,
    9. Rehman I,
    10. Rosario DJ,
    11. Di Martino E,
    12. Knowles MA,
    13. Meuth M,
    14. Harris AL and
    15. Hamdy FC
    : Distinct microRNA alterations characterize high- and low-grade bladder cancer. Cancer Res 69(21): 8472-8481, 2009. PMID: 19843843. DOI: 10.1158/0008-5472.CAN-09-0744
    OpenUrlAbstract/FREE Full Text
    1. Yang Z,
    2. Wu L,
    3. Wang A,
    4. Tang W,
    5. Zhao Y,
    6. Zhao H and
    7. Teschendorff AE
    : dbDEMC 2.0: updated database of differentially expressed miRNAs in human cancers. Nucleic Acids Res 45(D1): D812-D818, 2017. PMID: 27899556. DOI: 10.1093/nar/gkw1079
    OpenUrlCrossRefPubMed
  9. ↵
    1. Calin GA,
    2. Ferracin M,
    3. Cimmino A,
    4. Di Leva G,
    5. Shimizu M,
    6. Wojcik SE,
    7. Iorio MV,
    8. Visone R,
    9. Sever NI,
    10. Fabbri M,
    11. Iuliano R,
    12. Palumbo T,
    13. Pichiorri F,
    14. Roldo C,
    15. Garzon R,
    16. Sevignani C,
    17. Rassenti L,
    18. Alder H,
    19. Volinia S,
    20. Liu CG,
    21. Kipps TJ,
    22. Negrini M and
    23. Croce CM
    : A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med 353(17): 1793-1801, 2005. PMID: 16251535. DOI: 10.1056/NEJMoa050995
    OpenUrlCrossRefPubMed
  10. ↵
    1. Demes M,
    2. Aszyk C,
    3. Bartsch H,
    4. Schirren J and
    5. Fisseler-Eckhoff A
    : Differential miRNA-expression as an adjunctive diagnostic tool in neuroendocrine tumors of the lung. Cancers (Basel) 8(4): 38, 2016. PMID: 27023611. DOI: 10.3390/cancers8040038
    OpenUrlCrossRefPubMed
  11. ↵
    1. Mairinger FD,
    2. Ting S,
    3. Werner R,
    4. Walter RF,
    5. Hager T,
    6. Vollbrecht C,
    7. Christoph D,
    8. Worm K,
    9. Mairinger T,
    10. Sheu-Grabellus SY,
    11. Theegarten D,
    12. Schmid KW and
    13. Wohlschlaeger J
    : Different micro-RNA expression profiles distinguish subtypes of neuroendocrine tumors of the lung: results of a profiling study. Mod Pathol 27(12): 1632-1640, 2014. PMID: 24875640. DOI: 10.1038/modpathol.2014.74
    OpenUrlCrossRefPubMed
    1. Rapa I,
    2. Votta A,
    3. Gatti G,
    4. Izzo S,
    5. Buono NL,
    6. Giorgio E,
    7. Vatrano S,
    8. Napoli F,
    9. Scarpa A,
    10. Scagliotti G,
    11. Papotti M and
    12. Volante M
    : High miR-100 expression is associated with aggressive features and modulates TORC1 complex activation in lung carcinoids. Oncotarget 9(44): 27535-27546, 2018. PMID: 29938004. DOI: 10.18632/oncotarget.25541
    OpenUrlCrossRefPubMed
    1. Malczewska A,
    2. Kidd M,
    3. Matar S,
    4. Kos-Kudla B and
    5. Modlin IM
    : A comprehensive assessment of the role of miRNAs as biomarkers in gastroenteropancreatic neuroendocrine tumors. Neuroendocrinology 107(1): 73-90, 2018. PMID: 29566385. DOI: 10.1159/000487326
    OpenUrlCrossRefPubMed
    1. Lee HW,
    2. Lee EH,
    3. Ha SY,
    4. Lee CH,
    5. Chang HK,
    6. Chang S,
    7. Kwon KY,
    8. Hwang IS,
    9. Roh MS and
    10. Seo JW
    : Altered expression of microRNA miR-21, miR-155, and let-7a and their roles in pulmonary neuroendocrine tumors. Pathol Int 62(9): 583-591, 2012. PMID: 22924844. DOI: 10.1111/j.1440-1827.2012.02845.x
    OpenUrlCrossRefPubMed
  12. ↵
    1. Vicentini C,
    2. Fassan M,
    3. D’Angelo E,
    4. Corbo V,
    5. Silvestris N,
    6. Nuovo GJ and
    7. Scarpa A
    : Clinical application of microRNA testing in neuroendocrine tumors of the gastrointestinal tract. Molecules 19(2): 2458-2468, 2014. PMID: 24566314. DOI: 10.3390/molecules19022458
    OpenUrlCrossRefPubMed
  13. ↵
    1. Arvidsson Y,
    2. Rehammar A,
    3. Bergström A,
    4. Andersson E,
    5. Altiparmak G,
    6. Swärd C,
    7. Wängberg B,
    8. Kristiansson E and
    9. Nilsson O
    : miRNA profiling of small intestinal neuroendocrine tumors defines novel molecular subtypes and identifies miR-375 as a biomarker of patient survival. Mod Pathol 31(8): 1302-1317, 2018. PMID: 29487354. DOI: 10.1038/s41379-018-0010-1
    OpenUrlCrossRefPubMed
  14. ↵
    1. Thorns C,
    2. Schurmann C,
    3. Gebauer N,
    4. Wallaschofski H,
    5. Kümpers C,
    6. Bernard V,
    7. Feller AC,
    8. Keck T,
    9. Habermann JK,
    10. Begum N,
    11. Lehnert H and
    12. Brabant G
    : Global microRNA profiling of pancreatic neuroendocrine neoplasias. Anticancer Res 34(5): 2249-2254, 2014. PMID: 24778027.
    OpenUrlAbstract/FREE Full Text
    1. Roldo C,
    2. Missiaglia E,
    3. Hagan JP,
    4. Falconi M,
    5. Capelli P,
    6. Bersani S,
    7. Calin GA,
    8. Volinia S,
    9. Liu CG,
    10. Scarpa A and
    11. Croce CM
    : MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol 24(29): 4677-4684, 2006. PMID: 16966691. DOI: 10.1200/JCO.2005.05.5194
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Lee YS,
    2. Kim H,
    3. Kim HW,
    4. Lee JC,
    5. Paik KH,
    6. Kang J,
    7. Kim J,
    8. Yoon YS,
    9. Han HS,
    10. Sohn I,
    11. Cho J and
    12. Hwang JH
    : High expression of microRNA-196a indicates poor prognosis in resected pancreatic neuroendocrine tumor. Medicine (Baltimore) 94(50): e2224, 2015. PMID: 26683934. DOI: 10.1097/MD.0000000000002224
    OpenUrlCrossRefPubMed
  16. ↵
    1. Koduru SV,
    2. Leberfinger AN,
    3. Kawasawa YI,
    4. Mahajan M,
    5. Gusani NJ,
    6. Sanyal AJ and
    7. Ravnic DJ
    : Non-coding RNAs in various stages of liver disease leading to hepatocellular carcinoma: differential expression of miRNAs, piRNAs, lncRNAs, circRNAs, and sno/mt-RNAs. Sci Rep 8(1): 7967, 2018. PMID: 29789629. DOI: 10.1038/s41598-018-26360-1
    OpenUrlCrossRefPubMed
  17. ↵
    1. Bladen JC,
    2. Wang J,
    3. Sangaralingam A,
    4. Moosajee M,
    5. Fitchett C,
    6. Chelala C,
    7. Beaconsfield M,
    8. O’Toole EA,
    9. Philpott MP and
    10. Ezra DG
    : MicroRNA and transcriptome analysis in periocular Sebaceous Gland Carcinoma. Sci Rep 8(1): 7531, 2018. PMID: 29760516. DOI: 10.1038/s41598-018-25900-z
    OpenUrlCrossRefPubMed
  18. ↵
    1. Saller J,
    2. Jiang K,
    3. Xiong Y,
    4. Yoder SJ,
    5. Neill K,
    6. Pimiento JM,
    7. Pena L,
    8. Corbett FS,
    9. Magliocco A and
    10. Coppola D
    : A microRNA signature identifies patients at risk of barrett esophagus progression to dysplasia and cancer. Dig Dis Sci 67(2): 516-523, 2022. PMID: 33713247. DOI: 10.1007/s10620-021-06863-0
    OpenUrlCrossRefPubMed
  19. ↵
    1. Chatterjee A,
    2. Leichter AL,
    3. Fan V,
    4. Tsai P,
    5. Purcell RV,
    6. Sullivan MJ and
    7. Eccles MR
    : A cross comparison of technologies for the detection of microRNAs in clinical FFPE samples of hepatoblastoma patients. Sci Rep 5: 10438, 2015. PMID: 26039282. DOI: 10.1038/srep10438
    OpenUrlCrossRefPubMed
  20. ↵
    1. Gene Expression Data Analysis Guidelines | NanoString
    . Available at: https://nanostring.com/wp-content/uploads/Gene_Expression_Data_Analysis_Guidelines.pdf [Last accessed on January 11, 2023]
  21. ↵
    1. Ludwig N,
    2. Leidinger P,
    3. Becker K,
    4. Backes C,
    5. Fehlmann T,
    6. Pallasch C,
    7. Rheinheimer S,
    8. Meder B,
    9. Stähler C,
    10. Meese E and
    11. Keller A
    : Distribution of miRNA expression across human tissues. Nucleic Acids Res 44(8): 3865-3877, 2016. PMID: 26921406. DOI: 10.1093/nar/gkw116
    OpenUrlCrossRefPubMed
  22. ↵
    1. Dhawan A,
    2. Scott JG,
    3. Harris AL and
    4. Buffa FM
    : Pan-cancer characterisation of microRNA across cancer hallmarks reveals microRNA-mediated downregulation of tumour suppressors. Nat Commun 9(1): 5228, 2018. PMID: 30531873. DOI: 10.1038/s41467-018-07657-1
    OpenUrlCrossRefPubMed
  23. ↵
    1. Griffiths-Jones S,
    2. Grocock RJ,
    3. van Dongen S,
    4. Bateman A and
    5. Enright AJ
    : miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res 34(Database issue): D140-D144, 2006. PMID: 16381832. DOI: 10.1093/nar/gkj112
    OpenUrlCrossRefPubMed
    1. Coebergh van den Braak RRJ,
    2. Sieuwerts AM,
    3. Lalmahomed ZS,
    4. Smid M,
    5. Wilting SM,
    6. Bril SI,
    7. Xiang S,
    8. van der Vlugt-Daane M,
    9. de Weerd V,
    10. van Galen A,
    11. Biermann K,
    12. van Krieken JHJM,
    13. Kloosterman WP,
    14. Foekens JA, MATCH study group*,
    15. Martens JWM and
    16. IJzermans JNM
    : Confirmation of a metastasis-specific microRNA signature in primary colon cancer. Sci Rep 8(1): 5242, 2018. PMID: 29588449. DOI: 10.1038/s41598-018-22532-1
    OpenUrlCrossRefPubMed
    1. Kim J,
    2. Yao F,
    3. Xiao Z,
    4. Sun Y and
    5. Ma L
    : MicroRNAs and metastasis: small RNAs play big roles. Cancer Metastasis Rev 37(1): 5-15, 2018. PMID: 29234933. DOI: 10.1007/s10555-017-9712-y
    OpenUrlCrossRefPubMed
    1. Chan SH and
    2. Wang LH
    : Regulation of cancer metastasis by microRNAs. J Biomed Sci 22(1): 9, 2015. PMID: 25614041. DOI: 10.1186/s12929-015-0113-7
    OpenUrlCrossRefPubMed
  24. ↵
    1. Svoronos AA,
    2. Engelman DM and
    3. Slack FJ
    : OncomiR or tumor suppressor? The duplicity of microRNAs in cancer. Cancer Res 76(13): 3666-3670, 2016. PMID: 27325641. DOI: 10.1158/0008-5472.CAN-16-0359
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Leong I
    : A microRNA panel for thyroid nodules. Nat Rev Endocrinol 14(10): 565, 2018. PMID: 30076348. DOI: 10.1038/s41574-018-0079-0
    OpenUrlCrossRefPubMed
  26. ↵
    1. Nowek K,
    2. Wiemer EAC and
    3. Jongen-Lavrencic M
    : The versatile nature of miR-9/9(*) in human cancer. Oncotarget 9(29): 20838-20854, 2018. PMID: 29755694. DOI: 10.18632/oncotarget.24889
    OpenUrlCrossRefPubMed
  27. ↵
    1. Citron F,
    2. Armenia J,
    3. Franchin G,
    4. Polesel J,
    5. Talamini R,
    6. D’Andrea S,
    7. Sulfaro S,
    8. Croce CM,
    9. Klement W,
    10. Otasek D,
    11. Pastrello C,
    12. Tokar T,
    13. Jurisica I,
    14. French D,
    15. Bomben R,
    16. Vaccher E,
    17. Serraino D,
    18. Belletti B,
    19. Vecchione A,
    20. Barzan L and
    21. Baldassarre G
    : An integrated approach identifies mediators of local recurrence in head and neck squamous carcinoma. Clin Cancer Res 23(14): 3769-3780, 2017. PMID: 28174235. DOI: 10.1158/1078-0432.CCR-16-2814
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Shi Y,
    2. Qiu M,
    3. Wu Y and
    4. Hai L
    : MiR-548-3p functions as an anti-oncogenic regulator in breast cancer. Biomed Pharmacother 75: 111-116, 2015. PMID: 26297544. DOI: 10.1016/j.biopha.2015.07.027
    OpenUrlCrossRefPubMed
  29. ↵
    1. Lu J,
    2. Zhang M,
    3. Yang X,
    4. Cui T and
    5. Dai J
    : MicroRNA-548c-3p inhibits T98G glioma cell proliferation and migration by downregulating c-Myb. Oncol Lett 13(5): 3866-3872, 2017. PMID: 28536644. DOI: 10.3892/ol.2017.5870
    OpenUrlCrossRefPubMed
  30. ↵
    1. Dilsiz N,
    2. Balik A,
    3. Mutaf F,
    4. Yesil C and
    5. Borazan E
    : Differential expression of miRNAs in colorectal cancer. J Can Sci Res 3: S2, 2016. DOI: 10.4172/2576-1447.1000S2-001
    OpenUrlCrossRef
  31. ↵
    1. Fang L,
    2. Zhang HB,
    3. Li H,
    4. Fu Y and
    5. Yang GS
    : miR-548c-5p inhibits proliferation and migration and promotes apoptosis in CD90(+) HepG2 cells. Radiol Oncol 46(3): 233-241, 2012. PMID: 23077462. DOI: 10.2478/v10019-012-0025-z
    OpenUrlCrossRefPubMed
    1. Ke H,
    2. Zhao L,
    3. Feng X,
    4. Xu H,
    5. Zou L,
    6. Yang Q,
    7. Su X,
    8. Peng L and
    9. Jiao B
    : NEAT1 is required for survival of breast cancer cells through FUS and miR-548. Gene Regul Syst Bio 10(Suppl 1): 11-17, 2016. PMID: 27147820. DOI: 10.4137/GRSB.S29414
    OpenUrlCrossRefPubMed
  32. ↵
    1. Du B,
    2. Wang X,
    3. Wu D,
    4. Wang T,
    5. Yang X,
    6. Wang J,
    7. Shi X,
    8. Chen L and
    9. Zhang W
    : MicroRNA expression profiles identify biomarkers for predicting the response to chemoradiotherapy in rectal cancer. Mol Med Rep 18(2): 1909-1916, 2018. PMID: 29956755. DOI: 10.3892/mmr.2018.9215
    OpenUrlCrossRefPubMed
  33. ↵
    1. Ivashchenko AT,
    2. Issabekova AS and
    3. Berillo OA
    : miR-1279, miR-548j, miR-548m, and miR-548d-5p binding sites in CDSs of paralogous and orthologous PTPN12, MSH6, and ZEB1 Genes. Biomed Res Int 2013: 902467, 2013. PMID: 23957009. DOI: 10.1155/2013/902467
    OpenUrlCrossRefPubMed
  34. ↵
    1. Heyn H,
    2. Schreek S,
    3. Buurman R,
    4. Focken T,
    5. Schlegelberger B and
    6. Beger C
    : MicroRNA miR-548d is a superior regulator in pancreatic cancer. Pancreas 41(2): 218-221, 2012. PMID: 21946813. DOI: 10.1097/MPA.0b013e318224b701
    OpenUrlCrossRefPubMed
  35. ↵
    1. Hu B,
    2. Ying X,
    3. Wang J,
    4. Piriyapongsa J,
    5. Jordan IK,
    6. Sheng J,
    7. Yu F,
    8. Zhao P,
    9. Li Y,
    10. Wang H,
    11. Ng WL,
    12. Hu S,
    13. Wang X,
    14. Wang C,
    15. Zheng X,
    16. Li W,
    17. Curran WJ and
    18. Wang Y
    : Identification of a tumor-suppressive human-specific microRNA within the FHIT tumor-suppressor gene. Cancer Res 74(8): 2283-2294, 2014. PMID: 24556720. DOI: 10.1158/0008-5472.CAN-13-3279
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Chen L,
    2. Li X and
    3. Chen X
    : Prognostic significance of tissue miR-345 downregulation in non-small cell lung cancer. Int J Clin Exp Med 8(11): 20971-20976, 2015. PMID: 26885027.
    OpenUrlPubMed
  37. ↵
    1. Feng A,
    2. Yuan X and
    3. Li X
    : MicroRNA-345 inhibits metastasis and epithelial-mesenchymal transition of gastric cancer by targeting FOXQ1. Oncol Rep 38(5): 2752-2760, 2017. PMID: 29048674. DOI: 10.3892/or.2017.6001
    OpenUrlCrossRefPubMed
  38. ↵
    1. Srivastava SK,
    2. Bhardwaj A,
    3. Arora S,
    4. Tyagi N,
    5. Singh S,
    6. Andrews J,
    7. McClellan S,
    8. Wang B and
    9. Singh AP
    : MicroRNA-345 induces apoptosis in pancreatic cancer cells through potentiation of caspase-dependent and -independent pathways. Br J Cancer 113(4): 660-668, 2015. PMID: 26247574. DOI: 10.1038/bjc.2015.252
    OpenUrlCrossRefPubMed
  39. ↵
    1. Tang JT,
    2. Wang JL,
    3. Du W,
    4. Hong J,
    5. Zhao SL,
    6. Wang YC,
    7. Xiong H,
    8. Chen HM and
    9. Fang JY
    : MicroRNA 345, a methylation-sensitive microRNA is involved in cell proliferation and invasion in human colorectal cancer. Carcinogenesis 32(8): 1207-1215, 2011. PMID: 21665895. DOI: 10.1093/carcin/bgr114
    OpenUrlCrossRefPubMed
  40. ↵
    1. Zhang H,
    2. Liu H and
    3. Bi H
    : MicroRNA-345 inhibits hepatocellular carcinoma metastasis by inhibiting YAP1. Oncol Rep 38(2): 843-849, 2017. PMID: 28677785. DOI: 10.3892/or.2017.5772
    OpenUrlCrossRefPubMed
  41. ↵
    1. Tinay I,
    2. Tan M,
    3. Gui B,
    4. Werner L,
    5. Kibel AS and
    6. Jia L
    : Functional roles and potential clinical application of miRNA-345-5p in prostate cancer. Prostate 78(12): 927-937, 2018. PMID: 29748958. DOI: 10.1002/pros.23650
    OpenUrlCrossRefPubMed
  42. ↵
    1. Chen QG,
    2. Zhou W,
    3. Han T,
    4. Du SQ,
    5. Li ZH,
    6. Zhang Z,
    7. Shan GY and
    8. Kong CZ
    : MiR-345 suppresses proliferation, migration and invasion by targeting Smad1 in human prostate cancer. J Cancer Res Clin Oncol 142(1): 213-224, 2016. PMID: 26227059. DOI: 10.1007/s00432-015-2016-0
    OpenUrlCrossRefPubMed
  43. ↵
    1. Zhao C,
    2. Li Y,
    3. Chen G,
    4. Wang F,
    5. Shen Z and
    6. Zhou R
    : Overexpression of miR-15b-5p promotes gastric cancer metastasis by regulating PAQR3. Oncol Rep 38(1): 352-358, 2017. PMID: 28560431. DOI: 10.3892/or.2017.5673
    OpenUrlCrossRefPubMed
  44. ↵
    1. Link A and
    2. Kupcinskas J
    : MicroRNAs as non-invasive diagnostic biomarkers for gastric cancer: Current insights and future perspectives. World J Gastroenterol 24(30): 3313-3329, 2018. PMID: 30122873. DOI: 10.3748/wjg.v24.i30.3313
    OpenUrlCrossRefPubMed
  45. ↵
    1. Li J,
    2. Chen Y,
    3. Guo X,
    4. Zhou L,
    5. Jia Z,
    6. Tang Y,
    7. Lin L,
    8. Liu W and
    9. Ren C
    : Inhibition of miR-15b decreases cell migration and metastasis in colorectal cancer. Tumour Biol 37(7): 8765-8773, 2016. PMID: 26743779. DOI: 10.1007/s13277-015-4396-9
    OpenUrlCrossRefPubMed
  46. ↵
    1. Ji WB,
    2. Liu X,
    3. Luo Y and
    4. Zhang WZ
    : High expression of miR-15b predicts poor prognosis for hepatocellular carcinoma after curative hepatectomy. Oncol Rep 36(4): 1901-1908, 2016. PMID: 27499071. DOI: 10.3892/or.2016.4982
    OpenUrlCrossRefPubMed
  47. ↵
    1. Chen R,
    2. Sheng L,
    3. Zhang HJ,
    4. Ji M and
    5. Qian WQ
    : miR-15b-5p facilitates the tumorigenicity by targeting RECK and predicts tumour recurrence in prostate cancer. J Cell Mol Med 22(3): 1855-1863, 2018. PMID: 29363862. DOI: 10.1111/jcmm.13469
    OpenUrlCrossRefPubMed
  48. ↵
    1. Arakawa F,
    2. Kimura Y,
    3. Yoshida N,
    4. Miyoshi H,
    5. Doi A,
    6. Yasuda K,
    7. Nakajima K,
    8. Kiyasu J,
    9. Niino D,
    10. Sugita Y,
    11. Tashiro K,
    12. Kuhara S,
    13. Seto M and
    14. Ohshima K
    : Identification of miR-15b as a transformation-related factor in mantle cell lymphoma. Int J Oncol 48(2): 485-492, 2016. PMID: 26676972. DOI: 10.3892/ijo.2015.3295
    OpenUrlCrossRefPubMed
  49. ↵
    1. Lu YC,
    2. Chen YJ,
    3. Wang HM,
    4. Tsai CY,
    5. Chen WH,
    6. Huang YC,
    7. Fan KH,
    8. Tsai CN,
    9. Huang SF,
    10. Kang CJ,
    11. Chang JT and
    12. Cheng AJ
    : Oncogenic function and early detection potential of miRNA-10b in oral cancer as identified by microRNA profiling. Cancer Prev Res (Phila) 5(4): 665-674, 2012. PMID: 22318752. DOI: 10.1158/1940-6207.CAPR-11-0358
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Robertson ED,
    2. Wasylyk C,
    3. Ye T,
    4. Jung AC and
    5. Wasylyk B
    : The oncogenic MicroRNA Hsa-miR-155-5p targets the transcription factor ELK3 and links it to the hypoxia response. PLoS One 9(11): e113050, 2014. PMID: 25401928. DOI: 10.1371/journal.pone.0113050
    OpenUrlCrossRefPubMed
  51. ↵
    1. Al-Haidari A,
    2. Algaber A,
    3. Madhi R,
    4. Syk I and
    5. Thorlacius H
    : MiR-155-5p controls colon cancer cell migration via post-transcriptional regulation of Human Antigen R (HuR). Cancer Lett 421: 145-151, 2018. PMID: 29471005. DOI: 10.1016/.canlet.2018.02.026
    OpenUrlCrossRefPubMed
  52. ↵
    1. Han JG,
    2. Jiang YD,
    3. Zhang CH,
    4. Yang YM,
    5. Pang D,
    6. Song YN and
    7. Zhang GQ
    : A novel panel of serum miR-21/miR-155/miR-365 as a potential diagnostic biomarker for breast cancer. Ann Surg Treat Res 92(2): 55-66, 2017. PMID: 28203552. DOI: 10.4174/astr.2017.92.2.55
    OpenUrlCrossRefPubMed
  53. ↵
    1. Zuo J,
    2. Yu Y,
    3. Zhu M,
    4. Jing W,
    5. Yu M,
    6. Chai H,
    7. Liang C and
    8. Tu J
    : Inhibition of miR-155, a therapeutic target for breast cancer, prevented in cancer stem cell formation. Cancer Biomark 21(2): 383-392, 2018. PMID: 29103027. DOI: 10.3233/CBM-170642
    OpenUrlCrossRefPubMed
  54. ↵
    1. Wei RJ,
    2. Zhang CH and
    3. Yang WZ
    : MiR-155 affects renal carcinoma cell proliferation, invasion and apoptosis through regulating GSK-3β/β-catenin signaling pathway. Eur Rev Med Pharmacol Sci 24(16): 8239, 2020. PMID: 32894521. DOI: 10.26355/eurrev_202008_22579
    OpenUrlCrossRefPubMed
  55. ↵
    1. Li XD,
    2. Li XM,
    3. Gu JW and
    4. Sun XC
    : MiR-155 regulates lymphoma cell proliferation and apoptosis through targeting SOCS3/JAK-STAT3 signaling pathway. Eur Rev Med Pharmacol Sci 24(14): 7577, 2020. PMID: 32744682. DOI: 10.26355/eurrev_202007_22251
    OpenUrlCrossRefPubMed
  56. ↵
    1. Zhu FQ,
    2. Zeng L,
    3. Tang N,
    4. Tang YP,
    5. Zhou BP,
    6. Li FF,
    7. Wu WG,
    8. Zeng XB and
    9. Peng SS
    : MicroRNA-155 downregulation promotes cell cycle arrest and apoptosis in diffuse large B-cell lymphoma. Oncol Res 24(6): 415-427, 2016. PMID: 28281962. DOI: 10.3727/096504016X14685034103473
    OpenUrlCrossRefPubMed
  57. ↵
    1. Wang L,
    2. Tang B,
    3. Han H,
    4. Mao D,
    5. Chen J,
    6. Zeng Y and
    7. Xiong M
    : miR-155 affects osteosarcoma MG-63 cell autophagy induced by adriamycin through regulating PTEN-PI3K/AKT/mTOR signaling pathway. Cancer Biother Radiopharm 33(1): 32-38, 2018. PMID: 29412697. DOI: 10.1089/cbr.2017.2306
    OpenUrlCrossRefPubMed
  58. ↵
    1. Wang F,
    2. Shan S,
    3. Huo Y,
    4. Xie Z,
    5. Fang Y,
    6. Qi Z,
    7. Chen F,
    8. Li Y and
    9. Sun B
    : MiR-155-5p inhibits PDK1 and promotes autophagy via the mTOR pathway in cervical cancer. Int J Biochem Cell Biol 99: 91-99, 2018. PMID: 29627439. DOI: 10.1016/j.biocel.2018.04.005
    OpenUrlCrossRefPubMed
  59. ↵
    1. Li S,
    2. Zhang T,
    3. Zhou X,
    4. Du Z,
    5. Chen F,
    6. Luo J and
    7. Liu Q
    : The tumor suppressor role of miR-155-5p in gastric cancer. Oncol Lett 16(2): 2709-2714, 2018. PMID: 30008945. DOI: 10.3892/ol.2018.8932
    OpenUrlCrossRefPubMed
  60. ↵
    1. Yee D,
    2. Shah KM,
    3. Coles MC,
    4. Sharp TV and
    5. Lagos D
    : MicroRNA-155 induction via TNF-α and IFN-γ suppresses expression of programmed death ligand-1 (PD-L1) in human primary cells. J Biol Chem 292(50): 20683-20693, 2017. PMID: 29066622. DOI: 10.1074/jbc.M117.809053
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Zhang J,
    2. Wang D,
    3. Xiong J,
    4. Chen L and
    5. Huang J
    : MicroRNA-33a-5p suppresses growth of osteosarcoma cells and is downregulated in human osteosarcoma. Oncol Lett 10(4): 2135-2141, 2015. PMID: 26622808. DOI: 10.3892/ol.2015.3503
    OpenUrlCrossRefPubMed
  62. ↵
    1. Wang LQ,
    2. Wong KY,
    3. Rosèn A and
    4. Chim CS
    : Epigenetic silencing of tumor suppressor miR-3151 contributes to Chinese chronic lymphocytic leukemia by constitutive activation of MADD/ERK and PIK3R2/AKT signaling pathways. Oncotarget 6(42): 44422-44436, 2015. PMID: 26517243. DOI: 10.18632/oncotarget.6251
    OpenUrlCrossRefPubMed
  63. ↵
    1. Xue W,
    2. Chen J,
    3. Liu X,
    4. Gong W,
    5. Zheng J,
    6. Guo X,
    7. Liu Y,
    8. Liu L,
    9. Ma J,
    10. Wang P,
    11. Li Z and
    12. Xue Y
    : PVT1 regulates the malignant behaviors of human glioma cells by targeting miR-190a-5p and miR-488-3p. Biochim Biophys Acta Mol Basis Dis 1864(5 Pt A): 1783-1794, 2018. PMID: 29501773. DOI: 10.1016/j.bbadis.2018.02.022
    OpenUrlCrossRefPubMed
  64. ↵
    1. Karatas OF,
    2. Wang J,
    3. Shao L,
    4. Ozen M,
    5. Zhang Y,
    6. Creighton CJ and
    7. Ittmann M
    : miR-33a is a tumor suppressor microRNA that is decreased in prostate cancer. Oncotarget 8(36): 60243-60256, 2017. PMID: 28947967. DOI: 10.18632/oncotarget.19521
    OpenUrlCrossRefPubMed
  65. ↵
    1. Zhou J,
    2. Xu D,
    3. Xie H,
    4. Tang J,
    5. Liu R,
    6. Li J,
    7. Wang S,
    8. Chen X,
    9. Su J,
    10. Zhou X,
    11. Xia K,
    12. He Q,
    13. Chen J,
    14. Xiong W,
    15. Cao P and
    16. Cao K
    : miR-33a functions as a tumor suppressor in melanoma by targeting HIF-1α. Cancer Biol Ther 16(6): 846-855, 2015. PMID: 25891797. DOI: 10.1080/15384047.2015.1030545
    OpenUrlCrossRefPubMed
  66. ↵
    1. Boldrini L,
    2. Giordano M,
    3. Niccoli C,
    4. Melfi F,
    5. Lucchi M,
    6. Mussi A and
    7. Fontanini G
    : Role of microRNA-33a in regulating the expression of PD-1 in lung adenocarcinoma. Cancer Cell Int 17: 105, 2017. PMID: 29176936. DOI: 10.1186/s12935-017-0474-y
    OpenUrlCrossRefPubMed
  67. ↵
    1. Kuo PL,
    2. Liao SH,
    3. Hung JY,
    4. Huang MS and
    5. Hsu YL
    : MicroRNA-33a functions as a bone metastasis suppressor in lung cancer by targeting parathyroid hormone related protein. Biochim Biophys Acta 1830(6): 3756-3766, 2013. PMID: 23458685. DOI: 10.1016/j.bbagen.2013.02.022
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Cancer Genomics - Proteomics: 20 (2)
Cancer Genomics & Proteomics
Vol. 20, Issue 2
March-April 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Cancer Genomics & Proteomics.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
An miRNA Signature Predicts Grading of Pancreatic Neuroendocrine Neoplasms
(Your Name) has sent you a message from Cancer Genomics & Proteomics
(Your Name) thought you would like to see the Cancer Genomics & Proteomics web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
4 + 15 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
An miRNA Signature Predicts Grading of Pancreatic Neuroendocrine Neoplasms
JAMES SALLER, DALEY WHITE, BROOKE HOUGH, SEAN YODER, JUNMIN WHITING, DUNG-TSA CHEN, ANTHONY MAGLIOCCO, DOMENICO COPPOLA
Cancer Genomics & Proteomics Mar 2023, 20 (2) 154-164; DOI: 10.21873/cgp.20370

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
An miRNA Signature Predicts Grading of Pancreatic Neuroendocrine Neoplasms
JAMES SALLER, DALEY WHITE, BROOKE HOUGH, SEAN YODER, JUNMIN WHITING, DUNG-TSA CHEN, ANTHONY MAGLIOCCO, DOMENICO COPPOLA
Cancer Genomics & Proteomics Mar 2023, 20 (2) 154-164; DOI: 10.21873/cgp.20370
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Conclusion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Recurrent 8q11-13 Aberrations Leading to PLAG1 Rearrangements, Including Novel Chimeras HNRNPA2B1::PLAG1 and SDCBP::PLAG1, in Lipomatous Tumors
  • Receptor for Hyaluronic Acid-mediated Motility (RHAMM) Is Associated With Prostate Cancer Migration and Poor Prognosis
  • Oncogenes and Methionine Addiction of Cancer: Role of c-MYC
Show more Article

Similar Articles

Keywords

  • microRNA
  • miRNA profiling
  • neuroendocrine
  • grading
Cancer & Genome Proteomics

© 2023 Cancer Genomics & Proteomics

Powered by HighWire