Next Article in Journal
Single-Agent and Associated Therapies with Monoclonal Antibodies: What About Follicular Lymphoma?
Next Article in Special Issue
From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models
Previous Article in Journal
Real-World Management of High-Risk Prostate Cancer Post-Radical Prostatectomy: Insights from a Regional Quality Collaborative
Previous Article in Special Issue
Stromal Cells in Early Inflammation-Related Pancreatic Carcinogenesis—Biology and Its Potential Role in Therapeutic Targeting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression

1
Section for RNA Biology and Pathogenesis, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
2
Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
3
Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
4
Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(10), 1601; https://doi.org/10.3390/cancers17101601
Submission received: 27 March 2025 / Revised: 4 May 2025 / Accepted: 6 May 2025 / Published: 8 May 2025
(This article belongs to the Special Issue Management of Pancreatic Cancer)

Simple Summary

Pancreatic cancer is a deadly disease which is often detected at advanced stages. Novel and innovative approaches are urgently needed to improve diagnosis and personalized treatment strategies to achieve significant clinical benefits. Here, we introduce two interesting yet largely understudied classes of disease-associated molecules, i.e., long non-coding RNAs and RNA-binding proteins. We highlight their potential as biomarkers as well as therapeutic targets, and we provide examples of their functional diversity contributing to pancreatic cancer development and progression.

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is responsible for about 467,000 cancer deaths annually. An oftentimes asymptomatic early phase of this disease results in a delayed diagnosis, and patients often present with advanced disease. Current treatment options have limited survival benefits, and only a minor patient population carries actionable genomic alterations. Hence, innovative personalized treatment strategies that consider molecular, cellular and functional analyses are urgently needed for pancreatic cancer patients. However, the majority of the genetic alterations found in PDAC are currently undruggable, or patients’ response is not as expected. Therefore, non-genomic biomarkers and alternative molecular targets should be considered in order to advance the clinical management of PDAC patients. In line with this, recent gene expression and single-cell transcriptome analyses have identified molecular subtypes and transcriptional cell states that affect disease progression and drug efficiency. In this review, we will introduce long non-coding RNAs (lncRNAs) as well as RNA-binding proteins (RBPs) that are able to modulate the transcriptome of a cell through diverse mechanisms, thereby contributing to disease progression. We will provide a brief overview about the general functions of lncRNAs and RBPs, respectively. Subsequently, we will highlight selected lncRNAs and RBPs that have been shown to play a role in PDAC development, progression and drug response. Finally, we will present strategies aiming to interfere with the expression and function of lncRNAs and RBPs.

1. Introduction

The most recent global cancer statistics for the year 2022 estimate a total of almost 20 million new cancer cases and about 9.7 million cancer deaths [1]. With nearly 510,716 new cases and 467,000 deaths, pancreatic cancer ranks at position 12 and 6 in terms of incidence and mortality, respectively. Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is a devastating disease with an overall 5-year survival rate of approximately 13% [2]. An oftentimes asymptomatic early phase leading to a delayed diagnosis of PDAC in patients presenting with advanced and metastatic disease, in conjunction with a high resistance to currently available therapies, is a critical factor responsible for this high mortality rate. Of note, surgery as well as chemotherapy and radiotherapy is the major treatment option, and despite advances in surgical and perioperative approaches, high recurrence rates (up to 79% after three years) after curative intended surgery are observed [3]. Importantly, somatic mutations in key cancer genes such as KRAS, TP53, CDKN2A, and SMAD4 as well as a large number of genomic and epigenetic passenger alterations generate a remarkable inter- and intratumoural heterogeneity, which induces a considerable variation in responses to anticancer therapies [4]. Moreover, the performance status and comorbidities of the patient often dictate the choice of regimen and limit the application of non-specific and toxic chemotherapy, thereby affecting therapy resistance and tumour progression. Hence, for pancreatic cancer patients, personalized treatment strategies that consider molecular, cellular and functional analyses are urgently needed to overcome these limitations. However, only 12–25% of pancreatic cancer cases harbour actionable genomic alterations [5]. For example, about 7% of patients carry BRCA1/2 germline mutations that sensitize their tumours to the PARP inhibitor olaparib, which was shown to extend progression-free survival in metastatic pancreatic adenocarcinoma [6]. Additional targeted therapies for rare molecular pancreatic cancer subgroups like NTRK-fusion-positive, the KRASG12C mutant, DNA mismatch repair-deficient, or microsatellite-instable (MSIhigh) tumours might be viable options as well [7,8,9,10]. However, the majority of the genetic alterations found in PDAC are currently undruggable, or patients’ response is not as expected despite targeting actionable mutations with tailored drugs. Therefore, non-genomic biomarkers and alternative molecular targets should be considered in order to advance the clinical management of PDAC patients. In line with this, recent gene expression and single-cell transcriptome analyses have identified molecular subtypes and transcriptional cell states that affect disease progression and drug efficiency [11,12,13,14].
In this review, we will introduce long non-coding RNAs (lncRNAs) as well as RNA-binding proteins (RBPs), which have been shown to be associated with certain PDAC subtypes and are able to modulate the transcriptome of a cell through diverse mechanisms [15,16,17,18]. We will provide a brief overview about the general functions of lncRNAs and RBPs, respectively. Subsequently, we will highlight selected lncRNAs and RBPs that have been shown to play a role in PDAC development, progression, and drug response. Last but not least, we will present strategies aiming to interfere with the expression and function of lncRNAs and RBPs.

2. Role of lncRNAs in PDAC Development and Progression

Even though less than 2% of the human genome consists of protein-coding genes, transcription is pervasive, and modern RNA sequencing technologies have shown that transcripts are generated from 75 to 85% of the genome [19,20]. Some of these non-protein-coding RNAs (ncRNAs), such as ribosomal RNAs, transfer RNAs and microRNAs (miRNAs), can be categorized into classes based on their specific functions. What remains is a vast number of mostly RNA polymerase II (Pol II)-derived transcripts that vary in size, function and distribution. LncRNAs are somewhat arbitrarily, as they are defined as transcripts that are longer than 500 nucleotides (nts) [21]. More than 100,000 human lncRNAs have been recorded, exceeding the ~20,000 protein-coding genes in number. Their lengths range from one to tens of thousands of nucleotides, and they can occupy various genomic locations [22,23,24]. Intergenic lncRNAs are derived from regions devoid of coding potential, while intronic and sense/antisense lncRNAs are co-localized with protein-coding genes. Bidirectional lncRNAs share promoters with protein-coding genes and are transcribed in the antisense direction. Many lncRNAs share features with messenger RNAs (mRNAs) in that they are often polyadenylated and 5′capped, tend to comprise multiple exons and undergo alternative splicing [25,26]. Major differences lie in their overall lower degree of sequence conservation between species and their more restricted expression patterns, often being limited to specific cell types or developmental stages [27,28].
On a molecular level, lncRNAs can form complex secondary and tertiary structures that can interface with DNA, proteins and other RNAs and may be found in both nucleus and cytoplasm, enabling them to regulate gene expression on every level [29]. Through RNA-DNA interactions, they can alter the chromatin architecture and control transcription [30,31]. Via RNA-RNA actions, they affect the processing, splicing, editing, stability and localization of RNAs, in general, as well as influence the translation rates of mRNAs [32,33,34,35]. Finally, they can regulate protein localization, turnover and modification through RNA–protein interactions and serve as scaffolds for large protein complexes and biomolecular condensates [36,37,38,39,40]. When it comes to classifying lncRNAs based on their functions, the broadest approach is to differentiate between cis-acting and trans-acting lncRNAs. Cis-acting lncRNAs accumulate at and interact with the locus from which they are transcribed [41]. Many lncRNAs are found associated with the chromatin, tethered to their transcriptional origins by Pol II, and these proximity-based mechanisms may offset their often low expression levels [42,43]. Other lncRNAs vacate their sites of transcription to operate in trans and exert their functions in other nuclear regions or the cytoplasm. Table 1 lists a selection of common modes of action that have been found for lncRNAs acting in both cis and trans. Notably, a given lncRNA can have multiple interaction partners and functional modes depending on the biological context.
Importantly, lncRNAs have been associated with a broad spectrum of human diseases, and it was discovered that the vast majority (~99%) of somatic single-nucleotide variations (SNVs) occur in non-coding regions including lncRNA loci [62]. Importantly, gene expression analyses uncovered a frequent dysregulation of lncRNAs in cancer, and functional studies identified individual non-coding transcripts controlling cancer hallmarks [63,64,65]. Accordingly, lncRNAs have gained relevance as biomarkers for the early detection of cancer, prognostic markers and therapeutic targets [66]. Their expression patterns also have the tendency to be more cancer-type specific compared to pseudogenes and protein-coding genes, further underscoring their diagnostic value [67]. Given the dire trajectory of pancreatic cancer incidence and mortality, the potential for new biomarkers and therapeutic avenues offered by lncRNAs holds great potential. Of note, a large selection of lncRNAs has been found to be dysregulated in pancreatic tumours and PDAC cell lines and was shown to be associated with clinical disease parameters [68,69,70,71,72]. Many of these transcripts are upregulated in PDAC acting as oncogenes, promoting proliferation, survival and invasiveness. Several lncRNAs that were previously known to promote tumours in other cancer types have also been found to be increased in PDAC, e.g., H19, MALAT1 and HOTAIR [73,74,75]. Tumour-suppressive lncRNAs have also been identified, with examples including DGCR5, MEG3 and lnc-PCTST [76,77,78]. In the following paragraphs, we will highlight individual lncRNAs that have been linked with PDAC development and oncogenic signalling pathways underlying pancreatic cancer progression.

2.1. LncRNAs Associated with PDAC Initiation and Early Development

Frequently, dysregulated lncRNAs in PDAC are identified by comparing the gene expression signatures of late-stage tumour samples and normal pancreatic tissues. In order to gain insights into the early stages of carcinogenesis, precursor lesions or preceding benign pancreatic diseases should be included in such analyses, and liquid biopsy samples might be considered for early biomarker discovery. In fact, the lncRNAs ABHD11-AS1 and HULC were both detected in the serum of pancreatic cancer patients with increasing levels following the disease’s progression from healthy patients to those with benign diseases like chronic pancreatitis, as well as to those with fully developed pancreatic cancer [79,80]. Another study focused on intraductal papillary mucinous neoplasms (IPMNs), a common type of cystic PDAC precursor lesion. LncRNA signatures in the plasma from a cohort of IPMN cases could differentiate both IPMNs from non-disease controls as well as aggressive IPMNs from benign precursors [81]. However, patient biopsies from early-stage PDAC cases are often difficult to obtain due to the late diagnosis of the disease. Here, genetically engineered mouse models (GEMMs) present a powerful tool to follow the entire progression, from precursor to metastatic cancer, and study the role of lncRNAs in this cascade. For example, Mello et al. used a mouse model with pancreas-specific expression of oncogenic KrasG12D to study the role of the lncRNA Neat1 in tumour initiation. In this model, Neat1 proved critical in suppressing the development of premalignant pancreatic intraepithelial neoplasia (PanIN) and cystic lesions [82]. Of note, this conserved lncRNA has been implicated in a variety of cellular functions and is best known for its architectural role in nuclear paraspeckles, a type of nuclear body which is itself multifunctional [83]. It is therefore perhaps not surprising that its role in cancer is also multifaceted, promoting or suppressing tumour development depending on the cancer type and context. Indeed, in developed pancreatic cancer and fully transformed PDAC cell lines, human NEAT1 has mostly been assigned oncogenic roles [84,85,86]. The example of NEAT1 highlights the importance of resolving the functions of lncRNAs in early cancer stages to gain a complete mechanistic understanding of the disease and the stage-specific contributions of lncRNAs.

2.2. LncRNAs Associated with Oncogenic KRAS Signalling

In the majority of PDAC cases, mutations cause a molecular switch in KRAS to assume a constitutive ‘ON’ state, resulting in a constant stream of proliferation and survival signals. The G12D and G12V mutations in the KRAS gene are sufficient to initiate the development of precursor lesions, and the gene dosage of mutant KRAS remains relevant throughout the full progression of the disease up to the growth and maintenance of metastases [87,88,89]. Interestingly, some lncRNAs have been found to regulate the expression and activity of KRAS. For example, MALAT1 and NUTF2P3-01 were both found to be overexpressed in pancreatic cancer, and their knockdown led to a reduction in KRAS protein level. Both lncRNAs share miRNA binding sites with the KRAS mRNA, suggesting a ceRNA network that is involved in tuning intracellular KRAS expression [90,91]. Additionally, several studies have reported the upregulation of the lncRNA UCA1 in PDAC tissues and cell lines [92]. One of these studies observed that UCA1 promoted the interaction between the KRAS and hnRNPAB1 proteins [93]. This interaction had previously been shown to positively regulate the activation of PI3K/AKT/mTOR signalling through oncogenic KRAS [94]. The authors also observed that UCA1 knockdown decreased KRAS expression, and KRAS depletion reduced UCA1 levels, indicating a positive feedback loop [93]. Furthermore, UCA1 was found to modulate the downstream signalling pathways of KRAS. In particular, MAPK/ERK signalling was shown to be positively regulated by UCA1, which increased mitochondrial dynamics and thereby affected the migratory ability of pancreatic cancer cells [95]. Another study corroborated the enhancement of ERK signalling by UCA1 and revealed that CUDR, an alternative transcript variant of UCA1, could promote the migration and invasion of pancreatic cancer cells by activating AKT/FAK and ERK signalling to induce an epithelial-to-mesenchymal transition (EMT) [96,97].
In addition to UCA1, several other lncRNAs have been shown to modulate KRAS downstream signalling in pancreatic cancer. For example, ABHD11-AS1 and SNHG1 are both overexpressed in PDAC and promote the proliferation and survival of cancer cells by up-regulating PI3K/AKT signalling [98,99]. In addition, the lncRNA LUCAT1 was found to increase both AKT- and p38-mediated MAPK signalling, thereby supporting proliferation and invasion [100,101]. One lncRNA that has been studied in more detail on the mechanistic level is LINC01232. Like UCA1, it has also been shown to interact with hnRNPAB1 [102]. In addition to the KRAS mRNA, hnRNPAB1 also binds to the pre-mRNA of the downstream effector ARAF and regulates its splicing. This interaction favours the generation of the full-length, kinase-proficient ARAF protein [103]. LINC01232 stabilizes hnRNPAB1 by reducing its ubiquitination and proteasomal degradation, thereby promoting the splicing of the active ARAF isoform and subsequent downstream MAPK signalling [102].
In addition to PDAC-associated lncRNAs that regulate KRAS and its downstream pathways, several studies have also identified non-coding transcripts whose expression is regulated by these signalling cues. For instance, a recent study systematically deactivated MAPK/ERK signalling in a panel of PDAC cell lines and performed comparative transcriptome analyses to identify MAPK-associated lncRNAs [104]. Out of a list of 45 candidates, LINC00941 showed the most consistent downregulation upon MAPK abrogation, and the authors identified a binding site for the MAPK-activated transcription factor ETS-1 in its promoter region [104]. LINC00941 has been linked to PDAC progression in several studies and has been implicated in enhancing aerobic glycolysis by modulating the Hippo pathway and inducing AKT/FAK signalling by binding and stabilizing the ANXA2 protein [105,106]. Overall, these findings highlight the intimate link between lncRNAs and KRAS/MAPK/ERK and their associated downstream signalling in PDAC.

2.3. LncRNAs Associated with Oncogenic and Tumour-Suppressive TGF-β Signalling

TGF-β signalling has an ambivalent role throughout PDAC carcinogenesis. In the early stages of the disease, it presents a serious impediment to cancer progression by blocking the cell cycle and inducing apoptosis. In contrast, advanced tumours benefit from TGF-β-mediated immune evasion and the increased cell motility and metastatic potential stemming from a TGF-β-mediated epigenetic and transcriptional reprogramming that leads to EMT [107]. The loss of the TGF-β downstream signalling protein and tumour suppressor SMAD4, which occurs in up to 60% of PDAC, is often instrumental in this switch [108,109,110,111]. Many of the tumour suppressive effects of TGF-β are mediated by SMAD4, while the oncogenic pathways are mostly SMAD4-independent [112]. Intriguingly, recent studies identified regulatory lncRNAs that modulate the expression of SMAD4 in pancreatic cancer cells. One prominent lncRNA that has been extensively studied in cancer, in part due to the close proximity of its genomic locus to the well-known MYC oncogene, is PVT1 [113,114]. Increased levels of PVT1 were detected in PDAC tissues and cell lines, and its depletion inhibited TGF-β/SMAD signalling, including the phosphorylation of SMAD2 and SMAD3, as well as the expression of TGF-β1, but at the same time, it increased SMAD4 levels. Conversely, transient overexpression of PVT1 decreased SMAD4 levels, enhanced cellular proliferation and activated an EMT programme, thereby promoting cell migration and invasion [115]. In addition, LINC00909 was recently identified as an upregulated lncRNA in PDAC tissues compared to a normal pancreas, and its expression was associated with poor clinicopathological features and patient outcomes. Importantly, experimental studies revealed a cytoplasmic localization of this lncRNA in PDAC cell lines, and the modulation of its expression inversely affected the stability of the SMAD4 mRNA. Hence, this study established LINC00909 as an important regulator of pluripotency factors, cancer stemness and metastasis by inhibiting SMAD4 expression [116].
While the downregulation or inactivation of SMAD4 is important for PDAC progression, the TGF-β pathway has several other members whose expression or activity could be modulated by lncRNAs. In fact, several studies have identified lncRNAs that apply diverse modes of action in order to promote or inhibit TGF-β signalling in PDAC. Two lncRNAs, namely BC037916 and LINC00462, were found to be upregulated in PDAC, and they positively regulate the core TGF-β pathway, as evaluated by the increased phosphorylation of SMAD2/3 [117,118]. BC037916 was shown to activate JAK/STAT in addition to SMAD signalling, thereby promoting an EMT programme that increased cellular invasion and metastasis in xenograft models [117]. In contrast, LINC00462 was found to sequester miR-665, thereby upregulating TGFBR1 and TGFBR2 protein levels and activating migration and invasion through EMT induction [118].
In addition to these oncogenic lncRNAs, previous studies also highlighted the role of lncRNAs acting as tumour suppressors by modulating TGF-β signalling. For example, LINC-PINT was found to be downregulated in PDAC, and low plasma LINC-PINT expression might serve as a biomarker for early pancreatic cancer detection, whereas low levels of LINC-PINT in tumour tissues correlated with a poor prognosis after pancreatectomy [119,120]. Of note, the plasma levels of LINC-PINT and TGF-β1 were positively correlated in early-stage PDAC patients but not in healthy controls. Furthermore, the overexpression of LINC-PINT upregulated TGF-β1 expression in a pancreatic cancer cell line but not in telomerase-immortalized pancreatic ductal cells [120]. Importantly, LINC-PINT overexpression as well as exogenous TGF-β1 stimulation specifically reduced the proliferation of the pancreatic cancer cell line without affecting the growth of the non-malignant immortalized cells. The inhibitory effects of both LINC-PINT and TGF-β1 could be blocked by the simultaneous application of SD-208, a TGFBR1 inhibitor [120]. In conclusion, LINC-PINT may inhibit early-stage PDAC growth through TGF-β pathway activation. Last but not least, our own studies have identified LINC00261 as a TGF-β-regulated tumour-suppressive lncRNA that is involved in maintaining a pro-epithelial cell state, which is associated with a favourable disease outcome. We showed that the depletion of LINC00261 induced the expression of EMT-related genes, decreased E-cadherin levels and increased cell migration and invasion [121].
These examples underscore the diagnostic potential and functional relevance of lncRNAs in modulating the oncogenic as well as tumour-suppressive properties of the TGF-β signalling pathway in pancreatic cancer cells. Table 2 lists and Figure 1 schematically summarizes the findings on lncRNAs associated with KRAS and TGF-β signalling that have been described in this section. These principal cellular pathways controlling early PDAC progression evidently undergo regulation by lncRNAs on many levels.
The large number and functional versatility of lncRNAs, in general, suggest that this list is by no means completed yet. Systematic approaches towards mapping lncRNA dependencies could reveal many more potential biomarkers and therapeutic targets. Future work should incorporate samples from patients with benign precursor lesions and early-stage cancer models whenever possible. As illustrated by the examples of NEAT1 and the TGF-β pathway, the roles of lncRNAs and the processes involving them may change dynamically as pancreatic cancer progresses.

3. Role of RNA-Binding Proteins in PDAC

RNA-binding proteins are essential regulators of RNA biology, overseeing key stages of RNA life, including RNA splicing, processing, transport, stability, turnover and translation [123,124,125]. These multifunctional proteins are integral for maintaining cellular homeostasis and, consequently, human health. However, RBPs can become dysregulated, contributing to various diseases including cancer [126]. In PDAC, several RBPs are considered as disease drivers with prognostic relevance [127]. Some RBPs like LIN28A/B, ELAVL1, IGF2BP or MUSASHI proteins also exhibit long-standing and well-characterized roles in carcinogenesis or tumour suppression in other cancer types. In these cases, small-molecule inhibitors have been developed and employed to target malignant phenotypes. Despite these advancements, novel cancer-related functions for RBPs have recently been identified, continuously increasing the portfolio of RBP-based therapeutic strategies. This is particularly relevant in the context of PDAC where emerging evidence links specific RBPs to disease progression. In this section, we explore recent pancreatic cancer-related findings of selected RBPs, delving into the molecular mechanisms that underpin their influence on tumour growth, survival and metastasis (Figure 2). We will provide an overview of the regulatory pathways affected by the selected RBPs and summarize the potential of targeting these proteins.

3.1. ELAV-Like RNA Binding Protein 1 (ELAVL1)

ELAVL1, also known as human antigen R (HuR), plays a crucial role in the post-transcriptional regulation of numerous mRNAs involved in proliferation, cell survival, the stress response and chemoresistance. The protein is primarily located in the nucleus but shuttles to the cytoplasm to control the expression of its target transcripts. HuR binds to AU- or U-rich elements in the 3′ untranslated regions (3′UTRs) of target mRNAs to stabilize and/or enhance the translation of the respective transcripts [128]. HuR was initially discovered to be overexpressed in various cancer entities associated with a poor prognosis [129,130]. In PDAC, the HuR protein was detected in the nucleus and cytoplasm, and its cytoplasmic localization was associated with the tumour (T) stage. Interestingly, high cytoplasmic expression of HuR was associated with a favourable outcome in patients who received adjuvant therapy with gemcitabine [131]. Mechanistically, it was shown that HuR promotes the expression of DCK, an enzyme that activates gemcitabine, by stabilizing the DCK mRNA and enhancing its translation [132]. However, a retrospective study conducted in a cohort of 175 patients with resected periampullary adenocarcinomas, including pancreatic cancer, could not confirm the association between HuR and DCK expression and found that high HuR levels or a high HuR cytoplasmic-to-nuclear ratio is associated with poor survival in patients that received gemcitabine [133]. This finding is well in line with other reports that have assigned tumour-promoting functions to HuR. For instance, HuR was shown to stabilize the GPRC5A mRNA, encoding an orphan G-protein-coupled receptor, which is upregulated by gemcitabine and contributes to resistance mechanisms in pancreatic cancer cells [134].
Furthermore, HuR was identified as a regulator of proliferation, the cell cycle and apoptotic pathways in PDAC-derived cells under normal and stress conditions. In detail, the treatment of pancreatic cancer cells with DNA-damaging agents led to an accumulation of HuR in the cytoplasm and silenced expression-sensitized cells to these agents. Mechanistically, HuR was identified as a post-transcriptional regulator of the mitotic inhibitor kinase WEE1 by stabilizing the corresponding mRNA of this kinase, especially after DNA damage. Importantly, the upregulation of WEE1 induces a G2/M cell cycle arrest that enables cancer cells to repair their DNA [135]. Another study confirmed this DNA repair-promoting function of HuR by regulating the expression of BARD1 mRNA, thereby ensuring effective DNA repair via the homologous recombination repair pathway [136].
In addition to these DNA damage response-related functions, HuR was also shown to enhance the resilience of PDAC cells to cope with metabolic stress. For example, the depletion of HuR-sensitized pancreatic cancer cells to glucose deprivation led to increased apoptosis and reduced anchorage-independent colony formation. Interestingly, glucose deprivation induced the cytoplasmic translocation of HuR and allowed for the binding of this RBP to mRNAs encoding for several metabolic enzymes, including GPI, PRPS2 and IDH1. Of note, the depletion of HuR reduced the transcript and protein abundance of these metabolic enzymes in three pancreatic cancer cell lines, suggesting a critical role of HuR in modulating pancreatic cancer cell metabolism and survival under acute glucose deprivation [137]. This pro-survival effect of HuR in pancreatic cancer cells was observed in several other studies, and additional HuR target genes like YAP1 and DR5/TNFRSF10B as well as their associated pathways have been shown to be activated or inhibited by HuR [138,139].
Besides proliferation and apoptosis, HuR has been shown to promote cancer cell migration, invasion and metastasis as well as stem cell properties through its regulation of key genes, such as SNAI1, that play a role in EMT [140]. Intriguingly, the interruption of the HuR-RNA interaction by a novel compound called KH-3 reduced PDAC cell viability, EMT and metastatic potential both in vitro and in vivo [140].
In addition to the aforementioned tumour cell intrinsic functions, HuR was also shown to contribute to the remodelling of the tumour microenvironment (TME) by regulating the secretion of PDGFA, which led to increased collagen deposition and stromal activation in PDAC [141].
In summary, given its multifaceted role in promoting PDAC cell growth and survival, drug resistance and DNA repair, metabolic adaptation, EMT as well as metastasis, HuR represents an interesting as well as challenging therapeutic target. Inhibiting HuR alone or in combination with, e.g., DNA-damaging agents or WEE1 inhibitors, may alter therapy responses and patient outcomes in pancreatic cancer.

3.2. Insulin-Like Growth Factor II mRNA-Binding Proteins (IGF2BPs)

The family of oncofetal IGF2BPs, commonly upregulated or de novo expressed in various solid cancers, is notably active in several types of cancer, including PDAC, where these RBPs regulate a range of cellular processes such as proliferation, migration, invasion, metabolic pathways, chemoresistance and interactions within the TME [142,143]. As highly conserved regulators of mRNA turnover and translation, IGF2BPs can act as N6-methyladenosine (m6A) readers, binding predominantly to the 3′UTR of target mRNAs [144,145]. Through this binding, IGF2BPs protect mRNAs from miRNA-mediated decay, thereby influencing the stability and translation efficiency of various oncogenic transcripts [146]. In detail, IGF2BP1 has been identified as a key player in pancreatic cancer and was found to be significantly upregulated in this malignancy [147,148]. The overexpression of IGF2BP1 was correlated with tumour size and shorter overall survival (OS) in pancreatic cancer patients, highlighting its clinical relevance as a prognostic marker [147,148]. Mechanistically, IGF2BP1 was shown to promote cell cycle progression and proliferation via CDC25A and E2F1 and by activating AKT signalling [147,148,149,150]. Moreover, IGF2BP1 was shown to contribute to gemcitabine resistance of pancreatic cancer cells via multiple mechanisms. One study unravelled a complex RNA regulatory network that is controlled by the interaction of IGF2BP1 with m6A-modified SH3BP5-AS1, which increased the expression and stability of this lncRNA. In turn, SH3BP5-AS1 activated the WNT signalling pathway by sponging miR-139-5p, upregulated CTBP1 expression and thereby contributed to increased pancreatic cancer cell invasion, migration and stemness, as well as enhanced resistance to gemcitabine [151]. Another study could show that gemcitabine treatment inhibited pancreatic cancer cell proliferation and migration and decreased the overall m6A level potentially via the treatment-induced downregulation of the m6A writer WTAP [152]. The reduction in WTAP led to a downregulation of the m6A-modified MYC mRNA as well as the MYC protein. Since the MYC mRNA is known to be bound and stabilized by IGF2BP1 in a m6A-dependent manner, the authors speculated that gemcitabine might interfere with the WTAP/MYC/IGF2BP1 axis to inhibit pancreatic cancer progression [152,153].
The second member of the IGF2BP family, namely IGF2BP2, was found to be overexpressed in pancreatic cancer tissues as well, where it has been associated with significantly shorter OS [154,155]. The overexpression of IGF2BP2 might be driven by multiple mechanisms, including the loss of tumour-suppressive miRNAs, gene amplification and upregulation mediated by the lncRNA LINC00901 [154,156]. Interestingly, high IGF2BP2 expression is often found in high-risk tumours that exhibit lower immune response activity [157]. Moreover, a negative correlation with immune signature markers and a direct association with the expression of PD-L1, a key immune checkpoint protein, have been described for IGF2BP2. In fact, by regulating PD-L1 expression, IGF2BP2 may contribute to immune evasion, further promoting tumour survival in the immune-suppressive microenvironment of pancreatic cancer [158]. In addition, IGF2BP2 was shown to promote cell proliferation and metabolic reprogramming to support rapid tumour growth through the activation of the PI3K/AKT signalling pathway as well as through the stimulation of the glycolytic activity in PDAC cells by stabilizing the mRNA of GLUT1/SLC2A1, thus enhancing its expression level and facilitating increased glucose uptake [154,159].
Last but not least, the third family member, IGF2BP3, is most frequently used as a biomarker for solid tumours [160]. In 1997, IGF2BP3 was discovered in a large-scale screen for differentially expressed genes in pancreatic cancer, and it was cloned and initially named as a KH domain-containing protein overexpressed in cancer (KOC) [161]. Later studies confirmed its overexpression and revealed the prognostic relevance of IGF2BP3 in PDAC [161,162,163,164,165]. Importantly, based on biopsies or microdissections, IGF2BP3 is considered as a biomarker that detects early pancreatic lesions with high sensitivity and specificity [162,163,164,165,166,167,168]. Similar to IGF2BP2, IGF2BP3 is part of a prognostic five-gene signature that allows for the stratification of patients into low-risk and high-risk groups. The latter shows a high tumour mutational burden with lower response rates to immune checkpoint therapies [157]. Furthermore, elevated IGF2BP3 levels are associated with increased cell proliferation, migration and invasion, although its exact mechanisms remain incompletely understood. A recent study suggested that IGF2BP3 might act by blocking or mediating the interaction of specific transcripts with the RNA-induced silencing complex (RISC), thereby modulating their susceptibility to miRNA-mediated post-transcriptional gene silencing [169]. Further data suggest that IGF2BP3 might facilitate pancreatic cancer cell invasion and metastasis through additional mechanisms, including the regulation of localized translation at cell protrusions, altered adhesion by controlling CD44 expression or by stabilizing the SMS mRNA [170,171,172,173]. Furthermore, IGF2BP3 was shown to upregulate UBE2K, which reinforces the proliferation and stem cell-like phenotype in PDAC [174].
Collectively, these studies highlight the functional relevance of the individual IGF2BP family members in pancreatic cancer by employing overlapping as well as specific regulatory mechanisms to activate oncogenic pathways while at the same time inhibiting tumour suppressive genes and immune cell functions. Hence, these proteins are prime candidates for drug development efforts, and their oncofetal expression patterns should allow for cancer-specific targeting with minimal side effects.

3.3. BicC Family RNA-Binding Protein 1 (BICC1)

BICC1 plays a crucial role in regulating cell fate through post-transcriptional mechanisms [175]. Originally identified in Drosophila melanogaster, BICC1 was recognized as a gene associated with maternal-effect mutations that cause a double-abdomen phenotype [176,177]. Later it was shown that the RBP BICC1 regulates anterior–posterior patterning in early development by controlling the localized translation of oskar, a key mRNA in Drosophila axis formation [178]. Recent studies highlight the upregulation of BICC1 in PDAC, particularly in lymph node metastases. Elevated BICC1 expression might affect immune cell infiltration and was shown to correlate with oncogenic pathways like EMT, TNFα/NF-kB and TGF-β signalling, which might underlie the poor prognosis of pancreatic cancer patients that express high levels of BICC1 in their tumours [179]. Mechanistically, BICC1 is able to influences tumour growth and progression through its post-transcriptional control of several molecular targets and pathways. In pancreatic cancer, BICC1 was shown to facilitate angiogenesis in a VEGF-independent manner [180]. In detail, an AU-rich sequence within the 3′UTR of the LCN2 mRNA was recognized by BICC1, and this stabilizing interaction increased the expression of the LCN2 protein. Elevated levels of LCN2 subsequently activated the JAK2/STAT3 signalling pathway and resulted in the enhanced expression and secretion of the pro-angiogenic factor CXCL1. Intriguingly, the anti-tumour effect of gemcitabine could be improved by treating patient-derived xenograft models with elevated BICC1 expression with an anti-LCN2 antibody but not with the anti-VEGF antibody Bevacizumab [180]. Another possibility to improve gemcitabine efficacy in PDAC by targeting BICC1 and its downstream signalling was recently described. In particular, BICC1 has been identified as an activator of tryptophan catabolism by increasing the expression of IDO1 [181]. The resulting increase in tryptophan metabolites contributes to the synthesis of nicotinamide adenine dinucleotide (NAD+) and promotes oxidative phosphorylation, creating a metabolic environment conducive to a stem cell-like phenotype in cancer cells. Notably, the inhibition of the BICC1/IDO1/tryptophan metabolic axis has shown potential in improving the efficacy of gemcitabine by reducing drug resistance and limiting the stem cell-like qualities of tumour cells [181]. Hence, BICC1 emerges as a multifaceted regulator in pancreatic cancer, influencing processes such as EMT, immune infiltration, angiogenesis, chemoresistance and stemness. Its regulatory impact on LCN2 and IDO1 expression highlights its potential as a therapeutic target to overcome VEGF inhibitor resistance and improve responses to chemotherapy in PDAC.
Taken together, the RBPs introduced in this section highlight the important contribution of post-transcriptional regulators in modulating gene expression programmes in pancreatic cancers. Table 3 provides a comprehensive overview about additional pancreatic cancer-associated RBPs. While these RBPs represent interesting targets to treat PDAC, only a limited number of inhibitors targeting a specific RBP have been developed so far. In the final section, we will provide a general overview about the diverse opportunities that enable interference with RBPs as well as lncRNAs.

4. Strategies to Target lncRNAs and RBPs

In general, targeting lncRNAs or RBPs in human PDAC patients is not a trivial task, and commonly applied strategies try to modulate the expression or interfere with molecular interactions. More recently, small molecules that mediate the selective degradation of RNAs and proteins have been developed and represent an innovative and promising opportunity (Figure 3). In this section, we will briefly introduce these strategies in more detail, and we discuss their advantages as well as their limitations.

4.1. RNA Interference-Based Targeting Approaches

RNA interference (RNAi) is a conserved cellular mechanism that utilizes small, double-stranded RNA molecules, such as small interfering RNA (siRNA), short hairpin RNA (shRNA) or miRNAs, to silence gene expression by targeting complementary RNA sequences [206]. This mechanism provides a versatile and fairly specific method for reducing lncRNA and RBP expression by effectively eliminating the respective transcripts that are mainly found in the cytoplasm. The specificity of the method minimizes the risk of off-target effects, which can be a concern with other therapeutic approaches. Furthermore, RNAi-based therapies offer the flexibility to inhibit multiple targets simultaneously, enabling a more comprehensive treatment design tailored to the complexities of the disease. Hence, this multifaceted approach can potentially address various pathways that contribute to tumour growth and resistance, enhancing overall treatment efficacy. However, despite its promising capabilities, RNA-based therapy faces significant challenges, particularly concerning delivery mechanisms. RNAi agents, such as siRNA and shRNA, are highly susceptible to rapid degradation in the bloodstream, which can significantly diminish their therapeutic potential. To achieve the effective silencing of target genes within PDAC cells, RNAi agents often require the use of specialized delivery vectors, such as lipid nanoparticles or viral vectors. These vectors play a crucial role in enhancing the stability of RNAi agents and facilitating their successful delivery to target cells. Consequently, ongoing research aims at improving the delivery and stability of RNAi agents, which is essential for advancing the clinical applicability of RNAi in cancer therapy [207]. For example, novel exosome-based delivery systems have been engineered to carry siRNAs or shRNAs that targeted mutant KRASG12D in multiple mouse models of pancreatic cancer [208].
In summary, RNAi offers a precise and flexible approach for gene silencing, providing a strategy for overcoming treatment resistance and enhancing chemotherapy efficacy by targeting lncRNAs and RBPs highlighted herein. While challenges related to delivery and stability persist, advancements in RNAi technologies hold promise for translating this innovative approach into effective clinical therapies for PDAC patients.

4.2. Antisense Oligonucleotides to Target lncRNAs and RBPs

Antisense oligonucleotides (ASOs) represent a powerful therapeutic strategy for targeting lncRNAs and RBPs. These short, single-stranded nucleic acids are designed to bind to complementary RNA sequences, providing a means to modulate gene expression at the transcript level. Importantly, their precise mechanisms of action depend on the specific design and chemical modification of ASOs. A major mechanism by which ASOs exert their effects is through their interaction with ribonuclease H (RNase H). This endogenous enzyme cleaves the RNA strand in RNA-DNA hybrids, and ASOs designed to leverage this interaction therefore contain a central DNA region flanked by chemically modified nucleotides [207,209]. Because RNase H cleavage occurs in the nucleus and cytoplasm, ASOs might be the preferred agents to manipulate the expression of lncRNAs that reside in the nucleus [209,210]. Another clinically relevant mechanism of action involves the regulation of alternative splicing by ASOs [211]. By binding to splice sites or regulatory sequences, ASOs can modulate the splicing patterns of target genes. Similarly, ASOs can function as steric blockers to prevent intra- and intermolecular RNA-RNA as well as RNA-DNA and RNA–protein interactions. This mode of action could be harnessed to inhibit mRNA translation, regulate polyadenylation or interfere with miRNA-mediated RNAi. Hence, ASOs similar to RNAi agents offer high specificity and versatility, and their design can be tailored for particular applications, enhancing their therapeutic potential. Furthermore, ASOs generally have manageable toxicity profiles, as they can be engineered to avoid interactions with non-targeted RNAs and proteins. Nevertheless, ASO-based therapies also face similar limitations like other RNA-based agents, particularly regarding their specific delivery. ASOs are often susceptible to rapid degradation in the bloodstream, which can hinder their effectiveness. To reach target cells, ASOs may require specialized delivery systems, such as nanoparticle-based carriers or conjugation to cell-penetrating peptides [207,212]. Thus, advancements in delivery strategies are crucial for improving the therapeutic efficacy of ASOs and ensuring that they effectively reach their intended targets within cancer cells.
In conclusion, ASOs are powerful agents that enable the targeting of nuclear and cytoplasmic protein-coding and non-coding transcripts. By inhibiting the expression of critical PDAC-associated lncRNAs and RBPs, ASOs can potentially disrupt the pathways that support tumour growth and survival. However, innovative ASO delivery systems need to be developed to enhance the effectiveness of these therapies in the fight against pancreatic cancer.

4.3. Small-Molecule Inhibitors

In addition to the aforementioned oligonucleotide-based targeting strategies, small molecules comprise another interesting approach to target lncRNAs and RBPs. These molecules are either rationally designed to bind to specific regions within their targets or they can be discovered using unbiased screening approaches. The major mechanism of action of such small-molecule inhibitors is to interfere with intra- and/or intermolecular interactions, thereby, for example, causing RNA misfolding or blocking the interaction between RBPs and their target transcripts. Indeed, several small-molecule inhibitors have been developed in recent years, and interested readers are referred to excellent reviews on this exciting topic [17,213,214,215,216,217,218]. Importantly, small-molecule inhibitors offer several advantages in targeting lncRNAs and RBPs. For example, by designing small molecules that bind to specific domains or structural elements only present in the target lncRNA or RBP, these molecules can be engineered for specificity, allowing for targeted inhibition with potentially minimal impact on non-target RNAs or proteins. Furthermore, many small-molecule inhibitors are designed for oral bioavailability and are optimized for pharmacokinetics and pharmacodynamics, which can simplify their administration and improve patient compliance. Compared to oligonucleotides and viral delivery systems, small molecules are typically easier and cheaper to synthesize and modify. Nevertheless, the identification of specific and effective small-molecule inhibitors to target RNAs and RBPs is not a trivial task given the lack of enzymatic activities that would provide an easy read-out for measuring on-target activity. Moreover, the lack of suitable binding pockets within most RBPs as well as the conserved structure of commonly found RNA-binding domains and therefore the lack of RBP-specific structural motifs as a suitable target region to disrupt intermolecular interactions complicates the rational design of small molecules. Moreover, combinatorial recognition of target RNA sequences or structures may be a common phenomenon, which makes it difficult to design small molecules that are able to block multiple binding sites. In the case of lncRNAs, structural information is often not available, and functionally relevant regions within the transcript are often unknown. Last but not least, a small molecule binding to its lncRNA or RBP target might not be sufficient to interfere with the activity of the respective target. However, once identified, specific small molecule binders can be converted to degraders as will be discussed in the next paragraph.

4.4. Targeting of lncRNAs and RBPs via Proximity-Inducing Bifunctional Molecules

The targeted degradation of disease-relevant proteins is a therapeutic concept that has gained lots of attention in recent years and has been transferred recently to the RNA world as well. The general idea is to induce proximity between a target molecule, e.g., RBP or lncRNA, and a machinery that is able to degrade the target. In order to achieve targeted protein degradation (TPD), so-called proteolysis-targeting chimeras (PROTACs) have been developed. These are heterobifunctional small molecules consisting of two ligands: one that binds a specific target protein and another one that binds an E3 ubiquitin ligase. Both ligands are joined by a linker, and the simultaneous binding of both ligands to their intended proteins induces the ubiquitination and subsequent degradation of the target protein [219]. This concept was recently applied to degrade the splicing factor SF3B1 as well as the eukaryotic translation initiation factor EIF4E [220,221]. An interesting variation of this concept, called RNA-PROTAC, has been developed and applied to degrade two RBPs, namely LIN28A and RBFOX1 [222]. The RNA-PROTAC approach requires a chimeric molecule which consists of a short oligo-ribonucleotide corresponding to the consensus binding sequence of the RBP that is conjugated to a peptide derived from HIF1A, thereby enabling the recruitment of VHL, an E3 ubiquitin ligase. The adaptation of the concept of proximity-induced degradation to eliminate specific RNAs led to the development of ribonuclease-targeting chimeras (RIBOTACs). Here, small molecules that bind to the RNA of interest are modified to carry a short 2′-5′oligoadenylate or another small molecule that binds to and locally activates RNase L [223,224,225]. This approach was recently leveraged to convert strong yet inactive binding interactions into potent and specific modulators of RNA function [226]. Of note, additional ASO and aptamer-based strategies have been invented to engage RNase L in targeted RNA degradation [227,228,229,230]. Thus, the field is rapidly evolving, and the development and application of PROTACs and RIBOTACs to target RBPs and lncRNAs in pancreatic cancer might change the trajectories of this devastating disease.
In summary, several strategies enable the targeting of lncRNAs and RBPs, thereby opening new avenues for biomedical research and development. However, targeting these new classes of molecules comes with additional clinical challenges and limitations. For example, targeting RBPs with housekeeping functions, e.g., regulators of splicing or translation, might cause systemic on-target toxicities that need to be carefully assessed using appropriate preclinical models. In addition, off-target toxicities of the therapeutic molecules could also occur and should be taken into account. Furthermore, genes with redundant functions, e.g., members of a specific RBP family, might require concurrent targeting in order to achieve therapeutic effects and avoid treatment resistance. Hence, potential resistance mechanisms, which are hard to predict, should be investigated early on using in vitro and in vivo approaches including unbiased functional genetic screens and molecular studies in order to understand the mode of action and drug resistance. This could also enable the development of companion diagnostics to enhance safety and efficacy. Last but not least, it is unlikely that single-target/single-agent strategies will achieve an enduring and deep clinical response in cancer therapy. Therefore, targeting a specific RBP or lncRNA in combination with already approved treatment regimen, e.g., gemcitabine/nab-paclitaxel in the case of PDAC, might be a promising strategy that should be explored systematically.

5. Conclusions and Future Perspectives

LncRNAs and RBPs are critical regulators of chemoresistance, cancer stem cell properties and oncogenic signalling in PDAC, making them compelling candidates for therapeutic interventions and disease biomarkers. In particular, the oncofetal expression pattern of individual RBPs and the tissue- and cell-type specific expression of certain lncRNAs represent unique opportunities for making future therapies more specific and early detection more sensitive. Therefore, future research should focus, on the one hand, on elucidating the molecular functions of individual, disease-associated lncRNAs and RBPs as well as their interconnections to better understand the complex regulatory networks that drive PDAC progression. Importantly, interfering with these interactions could be a valid therapeutic strategy. For example, targeting LINC01232 or UCA1 might affect hnRNPA2/B1 expression and its oncogenic functions [93,102]. Alternatively, preventing the m6A-dependent interaction between LINC00941 and IGF2BP2 could be a valid strategy to impair pancreatic cancer cell migration and invasion [231]. Additionally, LINC00261 was shown to compete with the MYC mRNA for binding to IGF2BP1, suggesting that the overexpression of LINC00261, or maybe just parts of it, could be used to decrease oncogenic MYC protein expression in PDAC [232].
On the other hand, there is an urgent need for novel sensitive and specific biomarkers, since the only Food and Drug Administration (FDA)-approved blood biomarker CA 19-9 is limited in its applicability for surveillance due to its variability among different ethnic populations and its insufficient sensitivity and specificity [233,234]. Thus, prospective studies should explore several sources, e.g., blood, saliva, urine and exosomes, and analyze different molecular classes, as well as potentially combine multiple approaches, especially transcriptomics and proteomics, in order to evaluate lncRNAs and RBPs as early disease biomarkers for patient identification and stratification. In addition, high-risk populations, like patients diagnosed with chronic pancreatitis, should also be considered and thoroughly investigated in order to identify RBPs and lncRNAs that are associated with predisposing conditions [235,236].
With ongoing research and improvements in targeting and drug delivery, the inhibition of lncRNAs and RBPs holds great potential, and novel combinatorial approaches involving lncRNA- or RBP-targeting drugs as well as conventional (chemo)therapies or other targeted agents may enhance their therapeutic efficacy and pave the way for more effective treatments against one of the most challenging cancers.

Author Contributions

Conceptualization, A.S., N.B., S.H. and T.G.; investigation, P.P., F.W.O., H.E. and K.U.I.M.; writing—original draft preparation, P.P., F.W.O., H.E., K.U.I.M., N.B., S.H. and T.G.; writing—review and editing, N.B. and T.G.; visualization, N.B.; supervision, T.G.; funding acquisition, A.S., N.B., S.H. and T.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was conducted within the Research Training Group InCuPanc (RTG2751) funded by Deutsche Forschungsgemeinschaft (DFG), grant number 449501615.

Acknowledgments

The authors want to thank Sina Krehahn and Nico Hesselbarth for their administrative support within the RTG 2751.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  3. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Chone, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  4. Beutel, A.K.; Ekizce, M.; Ettrich, T.J.; Seufferlein, T.; Lindenmayer, J.; Gout, J.; Kleger, A. Organoid-based precision medicine in pancreatic cancer. United Eur. Gastroenterol. J. 2024, 13, 21–33. [Google Scholar] [CrossRef] [PubMed]
  5. Pishvaian, M.J.; Blais, E.M.; Brody, J.R.; Lyons, E.; DeArbeloa, P.; Hendifar, A.; Mikhail, S.; Chung, V.; Sahai, V.; Sohal, D.P.S.; et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: A retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 2020, 21, 508–518. [Google Scholar] [CrossRef] [PubMed]
  6. Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
  7. Anisetti, B.; Coston, T.W.; Ahmed, A.K.; Mahadevia, H.J.; Edgar, M.A.; Starr, J.S.; Babiker, H.M. An Excellent Response of Microsatellite Instability-High Pancreatic Adenocarcinoma to Pembrolizumab Treatment: The Role of Circulating Tumor DNA Testing. Cureus 2023, 15, e37239. [Google Scholar] [CrossRef]
  8. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  9. Hu, Z.I.; Shia, J.; Stadler, Z.K.; Varghese, A.M.; Capanu, M.; Salo-Mullen, E.; Lowery, M.A.; Diaz, L.A., Jr.; Mandelker, D.; Yu, K.H.; et al. Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin. Cancer Res. 2018, 24, 1326–1336. [Google Scholar] [CrossRef]
  10. Strickler, J.H.; Satake, H.; George, T.J.; Yaeger, R.; Hollebecque, A.; Garrido-Laguna, I.; Schuler, M.; Burns, T.F.; Coveler, A.L.; Falchook, G.S.; et al. Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N. Engl. J. Med. 2023, 388, 33–43. [Google Scholar] [CrossRef]
  11. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.M.; Gingras, M.C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef]
  12. Collisson, E.A.; Sadanandam, A.; Olson, P.; Gibb, W.J.; Truitt, M.; Gu, S.; Cooc, J.; Weinkle, J.; Kim, G.E.; Jakkula, L.; et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 2011, 17, 500–503. [Google Scholar] [CrossRef] [PubMed]
  13. Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178. [Google Scholar] [CrossRef]
  14. Raghavan, S.; Winter, P.S.; Navia, A.W.; Williams, H.L.; DenAdel, A.; Lowder, K.E.; Galvez-Reyes, J.; Kalekar, R.L.; Mulugeta, N.; Kapner, K.S.; et al. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 2021, 184, 6119–6137.e6126. [Google Scholar] [CrossRef] [PubMed]
  15. Glass, M.; Dorn, A.; Huttelmaier, S.; Haemmerle, M.; Gutschner, T. Comprehensive Analysis of LincRNAs in Classical and Basal-Like Subtypes of Pancreatic Cancer. Cancers 2020, 12, 2077. [Google Scholar] [CrossRef]
  16. Henninger, J.E.; Young, R.A. An RNA-centric view of transcription and genome organization. Mol. Cell 2024, 84, 3627–3643. [Google Scholar] [CrossRef] [PubMed]
  17. Singh, V.; Singh, A.; Liu, A.J.; Fuchs, S.Y.; Sharma, A.K.; Spiegelman, V.S. RNA Binding Proteins as Potential Therapeutic Targets in Colorectal Cancer. Cancers 2024, 16, 3502. [Google Scholar] [CrossRef]
  18. Weisse, J.; Rosemann, J.; Krauspe, V.; Kappler, M.; Eckert, A.W.; Haemmerle, M.; Gutschner, T. RNA-Binding Proteins as Regulators of Migration, Invasion and Metastasis in Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2020, 21, 6835. [Google Scholar] [CrossRef]
  19. Djebali, S.; Davis, C.A.; Merkel, A.; Dobin, A.; Lassmann, T.; Mortazavi, A.; Tanzer, A.; Lagarde, J.; Lin, W.; Schlesinger, F.; et al. Landscape of transcription in human cells. Nature 2012, 489, 101–108. [Google Scholar] [CrossRef]
  20. International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature 2004, 431, 931–945. [Google Scholar] [CrossRef]
  21. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef]
  22. Mattick, J.S. Challenging the dogma: The hidden layer of non-protein-coding RNAs in complex organisms. Bioessays 2003, 25, 930–939. [Google Scholar] [CrossRef] [PubMed]
  23. Melo, C.A.; Drost, J.; Wijchers, P.J.; van de Werken, H.; de Wit, E.; Oude Vrielink, J.A.; Elkon, R.; Melo, S.A.; Leveille, N.; Kalluri, R.; et al. eRNAs are required for p53-dependent enhancer activity and gene transcription. Mol. Cell 2013, 49, 524–535. [Google Scholar] [CrossRef]
  24. Zhao, L.; Wang, J.; Li, Y.; Song, T.; Wu, Y.; Fang, S.; Bu, D.; Li, H.; Sun, L.; Pei, D.; et al. NONCODEV6: An updated database dedicated to long non-coding RNA annotation in both animals and plants. Nucleic Acids Res. 2021, 49, D165–D171. [Google Scholar] [CrossRef] [PubMed]
  25. Deveson, I.W.; Brunck, M.E.; Blackburn, J.; Tseng, E.; Hon, T.; Clark, T.A.; Clark, M.B.; Crawford, J.; Dinger, M.E.; Nielsen, L.K.; et al. Universal Alternative Splicing of Noncoding Exons. Cell Syst. 2018, 6, 245–255.e245. [Google Scholar] [CrossRef]
  26. Iyer, M.K.; Niknafs, Y.S.; Malik, R.; Singhal, U.; Sahu, A.; Hosono, Y.; Barrette, T.R.; Prensner, J.R.; Evans, J.R.; Zhao, S.; et al. The landscape of long noncoding RNAs in the human transcriptome. Nat. Genet. 2015, 47, 199–208. [Google Scholar] [CrossRef] [PubMed]
  27. Gloss, B.S.; Dinger, M.E. The specificity of long noncoding RNA expression. Biochim. Biophys. Acta 2016, 1859, 16–22. [Google Scholar] [CrossRef]
  28. Quinn, J.J.; Zhang, Q.C.; Georgiev, P.; Ilik, I.A.; Akhtar, A.; Chang, H.Y. Rapid evolutionary turnover underlies conserved lncRNA-genome interactions. Genes. Dev. 2016, 30, 191–207. [Google Scholar] [CrossRef]
  29. Guttman, M.; Rinn, J.L. Modular regulatory principles of large non-coding RNAs. Nature 2012, 482, 339–346. [Google Scholar] [CrossRef]
  30. Bhan, A.; Mandal, S.S. LncRNA HOTAIR: A master regulator of chromatin dynamics and cancer. Biochim. Biophys. Acta 2015, 1856, 151–164. [Google Scholar] [CrossRef]
  31. Sahakyan, A.; Yang, Y.; Plath, K. The Role of Xist in X-Chromosome Dosage Compensation. Trends Cell Biol. 2018, 28, 999–1013. [Google Scholar] [CrossRef]
  32. Faghihi, M.A.; Modarresi, F.; Khalil, A.M.; Wood, D.E.; Sahagan, B.G.; Morgan, T.E.; Finch, C.E.; St Laurent, G., 3rd; Kenny, P.J.; Wahlestedt, C. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of beta-secretase. Nat. Med. 2008, 14, 723–730. [Google Scholar] [CrossRef] [PubMed]
  33. Gong, C.; Maquat, L.E. lncRNAs transactivate STAU1-mediated mRNA decay by duplexing with 3′ UTRs via Alu elements. Nature 2011, 470, 284–288. [Google Scholar] [CrossRef] [PubMed]
  34. Romero-Barrios, N.; Legascue, M.F.; Benhamed, M.; Ariel, F.; Crespi, M. Splicing regulation by long noncoding RNAs. Nucleic Acids Res. 2018, 46, 2169–2184. [Google Scholar] [CrossRef]
  35. Toki, N.; Takahashi, H.; Sharma, H.; Valentine, M.N.Z.; Rahman, F.M.; Zucchelli, S.; Gustincich, S.; Carninci, P. SINEUP long non-coding RNA acts via PTBP1 and HNRNPK to promote translational initiation assemblies. Nucleic Acids Res. 2020, 48, 11626–11644. [Google Scholar] [CrossRef] [PubMed]
  36. Elguindy, M.M.; Mendell, J.T. NORAD-induced Pumilio phase separation is required for genome stability. Nature 2021, 595, 303–308. [Google Scholar] [CrossRef]
  37. Guo, C.J.; Ma, X.K.; Xing, Y.H.; Zheng, C.C.; Xu, Y.F.; Shan, L.; Zhang, J.; Wang, S.; Wang, Y.; Carmichael, G.G.; et al. Distinct Processing of lncRNAs Contributes to Non-conserved Functions in Stem Cells. Cell 2020, 181, 621–636.e622. [Google Scholar] [CrossRef]
  38. Liu, B.; Sun, L.; Liu, Q.; Gong, C.; Yao, Y.; Lv, X.; Lin, L.; Yao, H.; Su, F.; Li, D.; et al. A cytoplasmic NF-kappaB interacting long noncoding RNA blocks IkappaB phosphorylation and suppresses breast cancer metastasis. Cancer Cell 2015, 27, 370–381. [Google Scholar] [CrossRef]
  39. Lyu, Q.; Xu, S.; Lyu, Y.; Choi, M.; Christie, C.K.; Slivano, O.J.; Rahman, A.; Jin, Z.G.; Long, X.; Xu, Y.; et al. SENCR stabilizes vascular endothelial cell adherens junctions through interaction with CKAP4. Proc. Natl. Acad. Sci. USA 2019, 116, 546–555. [Google Scholar] [CrossRef]
  40. Yamazaki, T.; Souquere, S.; Chujo, T.; Kobelke, S.; Chong, Y.S.; Fox, A.H.; Bond, C.S.; Nakagawa, S.; Pierron, G.; Hirose, T. Functional Domains of NEAT1 Architectural lncRNA Induce Paraspeckle Assembly through Phase Separation. Mol. Cell 2018, 70, 1038–1053.e1037. [Google Scholar] [CrossRef]
  41. Gil, N.; Ulitsky, I. Regulation of gene expression by cis-acting long non-coding RNAs. Nat. Rev. Genet. 2020, 21, 102–117. [Google Scholar] [CrossRef] [PubMed]
  42. Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 2012, 22, 1775–1789. [Google Scholar] [CrossRef]
  43. Werner, M.S.; Ruthenburg, A.J. Nuclear Fractionation Reveals Thousands of Chromatin-Tethered Noncoding RNAs Adjacent to Active Genes. Cell Rep. 2015, 12, 1089–1098. [Google Scholar] [CrossRef]
  44. Arner, E.; Daub, C.O.; Vitting-Seerup, K.; Andersson, R.; Lilje, B.; Drablos, F.; Lennartsson, A.; Ronnerblad, M.; Hrydziuszko, O.; Vitezic, M.; et al. Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells. Science 2015, 347, 1010–1014. [Google Scholar] [CrossRef] [PubMed]
  45. Tan, J.Y.; Marques, A.C. The activity of human enhancers is modulated by the splicing of their associated lncRNAs. PLoS Comput. Biol. 2022, 18, e1009722. [Google Scholar] [CrossRef]
  46. Vucicevic, D.; Corradin, O.; Ntini, E.; Scacheri, P.C.; Orom, U.A. Long ncRNA expression associates with tissue-specific enhancers. Cell Cycle 2015, 14, 253–260. [Google Scholar] [CrossRef]
  47. Anderson, K.M.; Anderson, D.M.; McAnally, J.R.; Shelton, J.M.; Bassel-Duby, R.; Olson, E.N. Transcription of the non-coding RNA upperhand controls Hand2 expression and heart development. Nature 2016, 539, 433–436. [Google Scholar] [CrossRef] [PubMed]
  48. Han, X.; Zhang, J.; Liu, Y.; Fan, X.; Ai, S.; Luo, Y.; Li, X.; Jin, H.; Luo, S.; Zheng, H.; et al. The lncRNA Hand2os1/Uph locus orchestrates heart development through regulation of precise expression of Hand2. Development 2019, 146, dev176198. [Google Scholar] [CrossRef]
  49. Xiang, J.F.; Yin, Q.F.; Chen, T.; Zhang, Y.; Zhang, X.O.; Wu, Z.; Zhang, S.; Wang, H.B.; Ge, J.; Lu, X.; et al. Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 2014, 24, 513–531. [Google Scholar] [CrossRef]
  50. Cho, S.W.; Xu, J.; Sun, R.; Mumbach, M.R.; Carter, A.C.; Chen, Y.G.; Yost, K.E.; Kim, J.; He, J.; Nevins, S.A.; et al. Promoter of lncRNA Gene PVT1 Is a Tumor-Suppressor DNA Boundary Element. Cell 2018, 173, 1398–1412.e1322. [Google Scholar] [CrossRef]
  51. Penny, G.D.; Kay, G.F.; Sheardown, S.A.; Rastan, S.; Brockdorff, N. Requirement for Xist in X chromosome inactivation. Nature 1996, 379, 131–137. [Google Scholar] [CrossRef]
  52. Khalil, A.M.; Guttman, M.; Huarte, M.; Garber, M.; Raj, A.; Rivea Morales, D.; Thomas, K.; Presser, A.; Bernstein, B.E.; van Oudenaarden, A.; et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. USA 2009, 106, 11667–11672. [Google Scholar] [CrossRef] [PubMed]
  53. Long, Y.; Hwang, T.; Gooding, A.R.; Goodrich, K.J.; Rinn, J.L.; Cech, T.R. RNA is essential for PRC2 chromatin occupancy and function in human pluripotent stem cells. Nat. Genet. 2020, 52, 931–938. [Google Scholar] [CrossRef] [PubMed]
  54. Rinn, J.L.; Kertesz, M.; Wang, J.K.; Squazzo, S.L.; Xu, X.; Brugmann, S.A.; Goodnough, L.H.; Helms, J.A.; Farnham, P.J.; Segal, E.; et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007, 129, 1311–1323. [Google Scholar] [CrossRef]
  55. Roden, C.; Gladfelter, A.S. RNA contributions to the form and function of biomolecular condensates. Nat. Rev. Mol. Cell Biol. 2021, 22, 183–195. [Google Scholar] [CrossRef] [PubMed]
  56. Miyagawa, R.; Tano, K.; Mizuno, R.; Nakamura, Y.; Ijiri, K.; Rakwal, R.; Shibato, J.; Masuo, Y.; Mayeda, A.; Hirose, T.; et al. Identification of cis- and trans-acting factors involved in the localization of MALAT-1 noncoding RNA to nuclear speckles. RNA 2012, 18, 738–751. [Google Scholar] [CrossRef]
  57. Salmena, L.; Poliseno, L.; Tay, Y.; Kats, L.; Pandolfi, P.P. A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell 2011, 146, 353–358. [Google Scholar] [CrossRef]
  58. Denzler, R.; Agarwal, V.; Stefano, J.; Bartel, D.P.; Stoffel, M. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol. Cell 2014, 54, 766–776. [Google Scholar] [CrossRef]
  59. Cesana, M.; Cacchiarelli, D.; Legnini, I.; Santini, T.; Sthandier, O.; Chinappi, M.; Tramontano, A.; Bozzoni, I. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 2011, 147, 358–369. [Google Scholar] [CrossRef]
  60. Poliseno, L.; Salmena, L.; Zhang, J.; Carver, B.; Haveman, W.J.; Pandolfi, P.P. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010, 465, 1033–1038. [Google Scholar] [CrossRef]
  61. Vitiello, M.; Evangelista, M.; Zhang, Y.; Salmena, L.; Pandolfi, P.P.; Poliseno, L. PTENP1 is a ceRNA for PTEN: It’s CRISPR clear. J. Hematol. Oncol. 2020, 13, 73. [Google Scholar] [CrossRef]
  62. Khurana, E.; Fu, Y.; Colonna, V.; Mu, X.J.; Kang, H.M.; Lappalainen, T.; Sboner, A.; Lochovsky, L.; Chen, J.; Harmanci, A.; et al. Integrative annotation of variants from 1092 humans: Application to cancer genomics. Science 2013, 342, 1235587. [Google Scholar] [CrossRef] [PubMed]
  63. Gutschner, T.; Diederichs, S. The hallmarks of cancer: A long non-coding RNA point of view. RNA Biol. 2012, 9, 703–719. [Google Scholar] [CrossRef]
  64. Vancura, A.; Gutierrez, A.H.; Hennig, T.; Pulido-Quetglas, C.; Slack, F.J.; Johnson, R.; Haefliger, S. Is Evolutionary Conservation a Useful Predictor for Cancer Long Noncoding RNAs? Insights from the Cancer LncRNA Census 3. Noncoding RNA 2022, 8, 82. [Google Scholar] [CrossRef]
  65. Yang, M.; Lu, H.; Liu, J.; Wu, S.; Kim, P.; Zhou, X. lncRNAfunc: A knowledgebase of lncRNA function in human cancer. Nucleic Acids Res. 2022, 50, D1295–D1306. [Google Scholar] [CrossRef]
  66. Gutschner, T.; Richtig, G.; Haemmerle, M.; Pichler, M. From biomarkers to therapeutic targets-the promises and perils of long non-coding RNAs in cancer. Cancer Metastasis Rev. 2018, 37, 83–105. [Google Scholar] [CrossRef] [PubMed]
  67. Yan, X.; Hu, Z.; Feng, Y.; Hu, X.; Yuan, J.; Zhao, S.D.; Zhang, Y.; Yang, L.; Shan, W.; He, Q.; et al. Comprehensive Genomic Characterization of Long Non-coding RNAs across Human Cancers. Cancer Cell 2015, 28, 529–540. [Google Scholar] [CrossRef]
  68. Ashrafizadeh, M.; Rabiee, N.; Kumar, A.P.; Sethi, G.; Zarrabi, A.; Wang, Y. Long noncoding RNAs (lncRNAs) in pancreatic cancer progression. Drug Discov. Today 2022, 27, 2181–2198. [Google Scholar] [CrossRef] [PubMed]
  69. Duguang, L.; Jin, H.; Xiaowei, Q.; Peng, X.; Xiaodong, W.; Zhennan, L.; Jianjun, Q.; Jie, Y. The involvement of lncRNAs in the development and progression of pancreatic cancer. Cancer Biol. Ther. 2017, 18, 927–936. [Google Scholar] [CrossRef]
  70. Ghafouri-Fard, S.; Fathi, M.; Zhai, T.; Taheri, M.; Dong, P. LncRNAs: Novel Biomarkers for Pancreatic Cancer. Biomolecules 2021, 11, 1665. [Google Scholar] [CrossRef]
  71. Huang, X.; Zhi, X.; Gao, Y.; Ta, N.; Jiang, H.; Zheng, J. LncRNAs in pancreatic cancer. Oncotarget 2016, 7, 57379–57390. [Google Scholar] [CrossRef]
  72. Xu, J.; Xu, J.; Liu, X.; Jiang, J. The role of lncRNA-mediated ceRNA regulatory networks in pancreatic cancer. Cell Death Discov. 2022, 8, 287. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, K.; Jutooru, I.; Chadalapaka, G.; Johnson, G.; Frank, J.; Burghardt, R.; Kim, S.; Safe, S. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 2013, 32, 1616–1625. [Google Scholar] [CrossRef] [PubMed]
  74. Ma, C.; Nong, K.; Zhu, H.; Wang, W.; Huang, X.; Yuan, Z.; Ai, K. H19 promotes pancreatic cancer metastasis by derepressing let-7′s suppression on its target HMGA2-mediated EMT. Tumour Biol. 2014, 35, 9163–9169. [Google Scholar] [CrossRef] [PubMed]
  75. Pang, E.J.; Yang, R.; Fu, X.B.; Liu, Y.F. Overexpression of long non-coding RNA MALAT1 is correlated with clinical progression and unfavorable prognosis in pancreatic cancer. Tumour Biol. 2015, 36, 2403–2407. [Google Scholar] [CrossRef]
  76. Li, X.; Zhou, S.; Fan, T.; Feng, X. lncRNA DGCR 5/miR-27a-3p/BNIP3 promotes cell apoptosis in pancreatic cancer by regulating the p38 MAPK pathway. Int. J. Mol. Med. 2020, 46, 729–739. [Google Scholar] [CrossRef]
  77. Ma, L.; Wang, F.; Du, C.; Zhang, Z.; Guo, H.; Xie, X.; Gao, H.; Zhuang, Y.; Kornmann, M.; Gao, H.; et al. Long non-coding RNA MEG3 functions as a tumour suppressor and has prognostic predictive value in human pancreatic cancer. Oncol. Rep. 2018, 39, 1132–1140. [Google Scholar] [CrossRef]
  78. Wang, Y.; Ding, X.; Hu, H.; He, Y.; Lu, Z.; Wu, P.; Tian, L.; Xia, T.; Yin, J.; Yuan, H.; et al. Long non-coding RNA lnc-PCTST predicts prognosis through inhibiting progression of pancreatic cancer by downregulation of TACC-3. Int. J. Cancer 2018, 143, 3143–3154. [Google Scholar] [CrossRef]
  79. Liu, Y.; Feng, W.; Liu, W.; Kong, X.; Li, L.; He, J.; Wang, D.; Zhang, M.; Zhou, G.; Xu, W.; et al. Circulating lncRNA ABHD11-AS1 serves as a biomarker for early pancreatic cancer diagnosis. J. Cancer 2019, 10, 3746–3756. [Google Scholar] [CrossRef]
  80. Ou, Z.L.; Luo, Z.; Lu, Y.B. Long non-coding RNA HULC as a diagnostic and prognostic marker of pancreatic cancer. World J. Gastroenterol. 2019, 25, 6728–6742. [Google Scholar] [CrossRef]
  81. Permuth, J.B.; Chen, D.T.; Yoder, S.J.; Li, J.; Smith, A.T.; Choi, J.W.; Kim, J.; Balagurunathan, Y.; Jiang, K.; Coppola, D.; et al. Linc-ing Circulating Long Non-coding RNAs to the Diagnosis and Malignant Prediction of Intraductal Papillary Mucinous Neoplasms of the Pancreas. Sci. Rep. 2017, 7, 10484. [Google Scholar] [CrossRef] [PubMed]
  82. Mello, S.S.; Sinow, C.; Raj, N.; Mazur, P.K.; Bieging-Rolett, K.; Broz, D.K.; Imam, J.F.C.; Vogel, H.; Wood, L.D.; Sage, J.; et al. Neat1 is a p53-inducible lincRNA essential for transformation suppression. Genes. Dev. 2017, 31, 1095–1108. [Google Scholar] [CrossRef] [PubMed]
  83. Ingram, H.B.; Fox, A.H. Unveiling the intricacies of paraspeckle formation and function. Curr. Opin. Cell Biol. 2024, 90, 102399. [Google Scholar] [CrossRef] [PubMed]
  84. Cao, J.; Zhang, Y.; Yang, J.; He, S.; Li, M.; Yan, S.; Chen, Y.; Qu, C.; Xu, L. NEAT1 regulates pancreatic cancer cell growth, invasion and migration though mircroRNA-335-5p/c-met axis. Am. J. Cancer Res. 2016, 6, 2361–2374. [Google Scholar]
  85. Feng, Y.; Gao, L.; Cui, G.; Cao, Y. LncRNA NEAT1 facilitates pancreatic cancer growth and metastasis through stabilizing ELF3 mRNA. Am. J. Cancer Res. 2020, 10, 237–248. [Google Scholar]
  86. Huang, B.; Liu, C.; Wu, Q.; Zhang, J.; Min, Q.; Sheng, T.; Wang, X.; Zou, Y. Long non-coding RNA NEAT1 facilitates pancreatic cancer progression through negative modulation of miR-506-3p. Biochem. Biophys. Res. Commun. 2017, 482, 828–834. [Google Scholar] [CrossRef]
  87. Hingorani, S.R.; Petricoin, E.F.; Maitra, A.; Rajapakse, V.; King, C.; Jacobetz, M.A.; Ross, S.; Conrads, T.P.; Veenstra, T.D.; Hitt, B.A.; et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003, 4, 437–450. [Google Scholar] [CrossRef]
  88. Mueller, S.; Engleitner, T.; Maresch, R.; Zukowska, M.; Lange, S.; Kaltenbacher, T.; Konukiewitz, B.; Ollinger, R.; Zwiebel, M.; Strong, A.; et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 2018, 554, 62–68. [Google Scholar] [CrossRef]
  89. Guerra, C.; Schuhmacher, A.J.; Canamero, M.; Grippo, P.J.; Verdaguer, L.; Perez-Gallego, L.; Dubus, P.; Sandgren, E.P.; Barbacid, M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 2007, 11, 291–302. [Google Scholar] [CrossRef]
  90. Li, X.; Deng, S.J.; Zhu, S.; Jin, Y.; Cui, S.P.; Chen, J.Y.; Xiang, C.; Li, Q.Y.; He, C.; Zhao, S.F.; et al. Hypoxia-induced lncRNA-NUTF2P3-001 contributes to tumorigenesis of pancreatic cancer by derepressing the miR-3923/KRAS pathway. Oncotarget 2016, 7, 6000–6014. [Google Scholar] [CrossRef]
  91. Liu, P.; Yang, H.; Zhang, J.; Peng, X.; Lu, Z.; Tong, W.; Chen, J. The lncRNA MALAT1 acts as a competing endogenous RNA to regulate KRAS expression by sponging miR-217 in pancreatic ductal adenocarcinoma. Sci. Rep. 2017, 7, 5186. [Google Scholar] [CrossRef] [PubMed]
  92. Sun, C.C.; Li, L.; Jiang, Z.C.; Liu, Z.C.; Wang, L.; Wang, H.J. The Functional Role of LncRNA UCA1 in Pancreatic Cancer: A mini-review. J. Cancer 2023, 14, 275–280. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, Y.; Feng, W.; Gu, S.; Wang, H.; Zhang, Y.; Chen, W.; Xu, W.; Lin, C.; Gong, A.; Xu, M. The UCA1/KRAS axis promotes human pancreatic ductal adenocarcinoma stem cell properties and tumor growth. Am. J. Cancer Res. 2019, 9, 496–510. [Google Scholar]
  94. Barcelo, C.; Etchin, J.; Mansour, M.R.; Sanda, T.; Ginesta, M.M.; Sanchez-Arevalo Lobo, V.J.; Real, F.X.; Capella, G.; Estanyol, J.M.; Jaumot, M.; et al. Ribonucleoprotein HNRNPA2B1 interacts with and regulates oncogenic KRAS in pancreatic ductal adenocarcinoma cells. Gastroenterology 2014, 147, 882–892.e888. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, H.; Ding, Y.; He, Y.; Yu, Z.; Zhou, Y.; Gong, A.; Xu, M. LncRNA UCA1 promotes pancreatic cancer cell migration by regulating mitochondrial dynamics via the MAPK pathway. Arch. Biochem. Biophys. 2023, 748, 109783. [Google Scholar] [CrossRef]
  96. Liang, X.; Qi, M.; Wu, R.; Liu, A.; Chen, D.; Tang, L.; Chen, J.; Hu, X.; Li, W.; Zhan, L.; et al. Long non-coding RNA CUDR promotes malignant phenotypes in pancreatic ductal adenocarcinoma via activating AKT and ERK signaling pathways. Int. J. Oncol. 2018, 53, 2671–2682. [Google Scholar] [CrossRef]
  97. Wang, Y.; Chen, W.; Yang, C.; Wu, W.; Wu, S.; Qin, X.; Li, X. Long non-coding RNA UCA1a(CUDR) promotes proliferation and tumorigenesis of bladder cancer. Int. J. Oncol. 2012, 41, 276–284. [Google Scholar] [CrossRef]
  98. Qiao, X.; Lv, S.X.; Qiao, Y.; Li, Q.P.; Ye, B.; Wang, C.C.; Miao, L. Long noncoding RNA ABHD11-AS1 predicts the prognosis of pancreatic cancer patients and serves as a promoter by activating the PI3K-AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8630–8639. [Google Scholar] [CrossRef]
  99. Zhang, Y.; Zhang, R.; Luo, G.; Ai, K. Long noncoding RNA SNHG1 promotes cell proliferation through PI3K/AKT signaling pathway in pancreatic ductal adenocarcinoma. J. Cancer 2018, 9, 2713–2722. [Google Scholar] [CrossRef]
  100. Cao, W.; Zhang, H.F.; Ding, X.L.; Zhu, S.Z.; Zhou, G.X. The progression of pancreatic cancer cells is promoted by a long non-coding RNA LUCAT1 by activating AKT phosphorylation. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 738–748. [Google Scholar] [CrossRef]
  101. Nai, Y.; Pan, C.; Hu, X.; Ma, Y. LncRNA LUCAT1 contributes to cell proliferation and migration in human pancreatic ductal adenocarcinoma via sponging miR-539. Cancer Med. 2020, 9, 757–767. [Google Scholar] [CrossRef]
  102. Meng, L.D.; Shi, G.D.; Ge, W.L.; Huang, X.M.; Chen, Q.; Yuan, H.; Wu, P.F.; Lu, Y.C.; Shen, P.; Zhang, Y.H.; et al. Linc01232 promotes the metastasis of pancreatic cancer by suppressing the ubiquitin-mediated degradation of HNRNPA2B1 and activating the A-Raf-induced MAPK/ERK signaling pathway. Cancer Lett. 2020, 494, 107–120. [Google Scholar] [CrossRef] [PubMed]
  103. Shilo, A.; Ben Hur, V.; Denichenko, P.; Stein, I.; Pikarsky, E.; Rauch, J.; Kolch, W.; Zender, L.; Karni, R. Splicing factor hnRNP A2 activates the Ras-MAPK-ERK pathway by controlling A-Raf splicing in hepatocellular carcinoma development. RNA 2014, 20, 505–515. [Google Scholar] [CrossRef]
  104. Ishikawa, T.; Fukushige, S.; Saiki, Y.; Hirose, K.; Hiyoshi, T.; Ogawa, T.; Katori, Y.; Furukawa, T. Long Non-Coding RNAs Associated with Mitogen-Activated Protein Kinase in Human Pancreatic Cancer. Cancers 2023, 15, 303. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, J.; He, Z.; Liu, X.; Xu, J.; Jiang, X.; Quan, G.; Jiang, J. LINC00941 promotes pancreatic cancer malignancy by interacting with ANXA2 and suppressing NEDD4L-mediated degradation of ANXA2. Cell Death Dis. 2022, 13, 718. [Google Scholar] [CrossRef] [PubMed]
  106. Xu, M.; Cui, R.; Ye, L.; Wang, Y.; Wang, X.; Zhang, Q.; Wang, K.; Dong, C.; Le, W.; Chen, B. LINC00941 promotes glycolysis in pancreatic cancer by modulating the Hippo pathway. Mol. Ther. Nucleic Acids 2021, 26, 280–294. [Google Scholar] [CrossRef]
  107. Shen, W.; Tao, G.Q.; Zhang, Y.; Cai, B.; Sun, J.; Tian, Z.Q. TGF-beta in pancreatic cancer initiation and progression: Two sides of the same coin. Cell Biosci. 2017, 7, 39. [Google Scholar] [CrossRef]
  108. Hahn, S.A.; Schutte, M.; Hoque, A.T.; Moskaluk, C.A.; da Costa, L.T.; Rozenblum, E.; Weinstein, C.L.; Fischer, A.; Yeo, C.J.; Hruban, R.H.; et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996, 271, 350–353. [Google Scholar] [CrossRef]
  109. McCarthy, A.J.; Chetty, R. Smad4/DPC4. J. Clin. Pathol. 2018, 71, 661–664. [Google Scholar] [CrossRef]
  110. Tascilar, M.; Skinner, H.G.; Rosty, C.; Sohn, T.; Wilentz, R.E.; Offerhaus, G.J.; Adsay, V.; Abrams, R.A.; Cameron, J.L.; Kern, S.E.; et al. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2001, 7, 4115–4121. [Google Scholar]
  111. Yamada, S.; Fujii, T.; Shimoyama, Y.; Kanda, M.; Nakayama, G.; Sugimoto, H.; Koike, M.; Nomoto, S.; Fujiwara, M.; Nakao, A.; et al. SMAD4 expression predicts local spread and treatment failure in resected pancreatic cancer. Pancreas 2015, 44, 660–664. [Google Scholar] [CrossRef] [PubMed]
  112. Xia, X.; Wu, W.; Huang, C.; Cen, G.; Jiang, T.; Cao, J.; Huang, K.; Qiu, Z. SMAD4 and its role in pancreatic cancer. Tumour Biol. 2015, 36, 111–119. [Google Scholar] [CrossRef] [PubMed]
  113. Derderian, C.; Orunmuyi, A.T.; Olapade-Olaopa, E.O.; Ogunwobi, O.O. PVT1 Signaling Is a Mediator of Cancer Progression. Front. Oncol. 2019, 9, 502. [Google Scholar] [CrossRef]
  114. Li, R.; Wang, X.; Zhu, C.; Wang, K. lncRNA PVT1: A novel oncogene in multiple cancers. Cell Mol. Biol. Lett. 2022, 27, 84. [Google Scholar] [CrossRef]
  115. Zhang, X.; Feng, W.; Zhang, J.; Ge, L.; Zhang, Y.; Jiang, X.; Peng, W.; Wang, D.; Gong, A.; Xu, M. Long non-coding RNA PVT1 promotes epithelial-mesenchymal transition via the TGF-beta/Smad pathway in pancreatic cancer cells. Oncol. Rep. 2018, 40, 1093–1102. [Google Scholar] [CrossRef]
  116. Li, Z.; Ma, Z.; Wang, S.; Yan, Q.; Zhuang, H.; Zhou, Z.; Liu, C.; Chen, Y.; Han, M.; Wu, Z.; et al. LINC00909 up-regulates pluripotency factors and promotes cancer stemness and metastasis in pancreatic ductal adenocarcinoma by targeting SMAD4. Biol. Direct 2024, 19, 24. [Google Scholar] [CrossRef]
  117. Chen, G.; Xu, L.; Ye, G.; Lin, J.; Meng, Z.; Shen, Y. Overexpression of a Long Non-Coding RNA BC037916 is Associated with Pancreatic Tumorigenesis and Poor Prognosis. Onco Targets Ther. 2020, 13, 13451–13463. [Google Scholar] [CrossRef]
  118. Zhou, B.; Guo, W.; Sun, C.; Zhang, B.; Zheng, F. Linc00462 promotes pancreatic cancer invasiveness through the miR-665/TGFBR1-TGFBR2/SMAD2/3 pathway. Cell Death Dis. 2018, 9, 706. [Google Scholar] [CrossRef] [PubMed]
  119. Li, L.; Zhang, G.Q.; Chen, H.; Zhao, Z.J.; Chen, H.Z.; Liu, H.; Wang, G.; Jia, Y.H.; Pan, S.H.; Kong, R.; et al. Plasma and tumor levels of Linc-pint are diagnostic and prognostic biomarkers for pancreatic cancer. Oncotarget 2016, 7, 71773–71781. [Google Scholar] [CrossRef]
  120. Lu, H.; Yang, D.; Zhang, L.; Lu, S.; Ye, J.; Li, M.; Hu, W. Linc-pint inhibits early stage pancreatic ductal adenocarcinoma growth through TGF-beta pathway activation. Oncol. Lett. 2019, 17, 4633–4639. [Google Scholar] [CrossRef]
  121. Dorn, A.; Glass, M.; Neu, C.T.; Heydel, B.; Huttelmaier, S.; Gutschner, T.; Haemmerle, M. LINC00261 Is Differentially Expressed in Pancreatic Cancer Subtypes and Regulates a Pro-Epithelial Cell Identity. Cancers 2020, 12, 1227. [Google Scholar] [CrossRef]
  122. Wu, B.Q.; Jiang, Y.; Zhu, F.; Sun, D.L.; He, X.Z. Long Noncoding RNA PVT1 Promotes EMT and Cell Proliferation and Migration Through Downregulating p21 in Pancreatic Cancer Cells. Technol. Cancer Res. Treat. 2017, 16, 819–827. [Google Scholar] [CrossRef] [PubMed]
  123. Gebauer, F.; Schwarzl, T.; Valcarcel, J.; Hentze, M.W. RNA-binding proteins in human genetic disease. Nat. Rev. Genet. 2021, 22, 185–198. [Google Scholar] [CrossRef] [PubMed]
  124. Gerstberger, S.; Hafner, M.; Tuschl, T. A census of human RNA-binding proteins. Nat. Rev. Genet. 2014, 15, 829–845. [Google Scholar] [CrossRef]
  125. Hentze, M.W.; Castello, A.; Schwarzl, T.; Preiss, T. A brave new world of RNA-binding proteins. Nat. Rev. Mol. Cell Biol. 2018, 19, 327–341. [Google Scholar] [CrossRef]
  126. Pereira, B.; Billaud, M.; Almeida, R. RNA-Binding Proteins in Cancer: Old Players and New Actors. Trends Cancer 2017, 3, 506–528. [Google Scholar] [CrossRef]
  127. Glass, M.; Michl, P.; Huttelmaier, A.S. RNA Binding Proteins as Drivers and Therapeutic Target Candidates in Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020, 21, 4190. [Google Scholar] [CrossRef] [PubMed]
  128. Grammatikakis, I.; Abdelmohsen, K.; Gorospe, M. Posttranslational control of HuR function. Wiley Interdiscip. Rev. RNA 2017, 8, e1372. [Google Scholar] [CrossRef]
  129. Finan, J.M.; Sutton, T.L.; Dixon, D.A.; Brody, J.R. Targeting the RNA-Binding Protein HuR in Cancer. Cancer Res. 2023, 83, 3507–3516. [Google Scholar] [CrossRef]
  130. Srikantan, S.; Gorospe, M. HuR function in disease. Front. Biosci. 2012, 17, 189–205. [Google Scholar] [CrossRef]
  131. Richards, N.G.; Rittenhouse, D.W.; Freydin, B.; Cozzitorto, J.A.; Grenda, D.; Rui, H.; Gonye, G.; Kennedy, E.P.; Yeo, C.J.; Brody, J.R.; et al. HuR status is a powerful marker for prognosis and response to gemcitabine-based chemotherapy for resected pancreatic ductal adenocarcinoma patients. Ann. Surg. 2010, 252, 499–505; discussion 505–506. [Google Scholar] [CrossRef]
  132. Costantino, C.L.; Witkiewicz, A.K.; Kuwano, Y.; Cozzitorto, J.A.; Kennedy, E.P.; Dasgupta, A.; Keen, J.C.; Yeo, C.J.; Gorospe, M.; Brody, J.R. The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res. 2009, 69, 4567–4572. [Google Scholar] [CrossRef] [PubMed]
  133. Elebro, J.; Ben Dror, L.; Heby, M.; Nodin, B.; Jirstrom, K.; Eberhard, J. Prognostic effect of hENT1, dCK and HuR expression by morphological type in periampullary adenocarcinoma, including pancreatic cancer. Acta Oncol. 2016, 55, 286–296. [Google Scholar] [CrossRef] [PubMed]
  134. Zhou, H.; Telonis, A.G.; Jing, Y.; Xia, N.L.; Biederman, L.; Jimbo, M.; Blanco, F.; Londin, E.; Brody, J.R.; Rigoutsos, I. GPRC5A is a potential oncogene in pancreatic ductal adenocarcinoma cells that is upregulated by gemcitabine with help from HuR. Cell Death Dis. 2016, 7, e2294. [Google Scholar] [CrossRef]
  135. Lal, S.; Burkhart, R.A.; Beeharry, N.; Bhattacharjee, V.; Londin, E.R.; Cozzitorto, J.A.; Romeo, C.; Jimbo, M.; Norris, Z.A.; Yeo, C.J.; et al. HuR posttranscriptionally regulates WEE1: Implications for the DNA damage response in pancreatic cancer cells. Cancer Res. 2014, 74, 1128–1140. [Google Scholar] [CrossRef]
  136. Jain, A.; McCoy, M.; Coats, C.; Brown, S.Z.; Addya, S.; Pelz, C.; Sears, R.C.; Yeo, C.J.; Brody, J.R. HuR Plays a Role in Double-Strand Break Repair in Pancreatic Cancer Cells and Regulates Functional BRCA1-Associated-Ring-Domain-1(BARD1) Isoforms. Cancers 2022, 14, 1848. [Google Scholar] [CrossRef]
  137. Burkhart, R.A.; Pineda, D.M.; Chand, S.N.; Romeo, C.; Londin, E.R.; Karoly, E.D.; Cozzitorto, J.A.; Rigoutsos, I.; Yeo, C.J.; Brody, J.R.; et al. HuR is a post-transcriptional regulator of core metabolic enzymes in pancreatic cancer. RNA Biol. 2013, 10, 1312–1323. [Google Scholar] [CrossRef] [PubMed]
  138. Brown, S.Z.; McCarthy, G.A.; Carroll, J.R.; Di Niro, R.; Pelz, C.; Jain, A.; Sutton, T.L.; Holly, H.D.; Nevler, A.; Schultz, C.W.; et al. The RNA-Binding Protein HuR Posttranscriptionally Regulates the Protumorigenic Activator YAP1 in Pancreatic Ductal Adenocarcinoma. Mol. Cell Biol. 2022, 42, e0001822. [Google Scholar] [CrossRef]
  139. Pineda, D.M.; Rittenhouse, D.W.; Valley, C.C.; Cozzitorto, J.A.; Burkhart, R.A.; Leiby, B.; Winter, J.M.; Weber, M.C.; Londin, E.R.; Rigoutsos, I.; et al. HuR’s post-transcriptional regulation of Death Receptor 5 in pancreatic cancer cells. Cancer Biol. Ther. 2012, 13, 946–955. [Google Scholar] [CrossRef]
  140. Dong, R.; Chen, P.; Polireddy, K.; Wu, X.; Wang, T.; Ramesh, R.; Dixon, D.A.; Xu, L.; Aube, J.; Chen, Q. An RNA-Binding Protein, Hu-antigen R, in Pancreatic Cancer Epithelial to Mesenchymal Transition, Metastasis, and Cancer Stem Cells. Mol. Cancer Ther. 2020, 19, 2267–2277. [Google Scholar] [CrossRef]
  141. McCarthy, G.A.; Di Niro, R.; Finan, J.M.; Jain, A.; Guo, Y.; Wyatt, C.R.; Guimaraes, A.R.; Waugh, T.A.; Keith, D.; Morgan, T.K.; et al. Deletion of the mRNA stability factor ELAVL1 (HuR) in pancreatic cancer cells disrupts the tumor microenvironment integrity. NAR Cancer 2023, 5, zcad016. [Google Scholar] [CrossRef]
  142. Bell, J.L.; Wachter, K.; Muhleck, B.; Pazaitis, N.; Kohn, M.; Lederer, M.; Huttelmaier, S. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): Post-transcriptional drivers of cancer progression? Cell Mol. Life Sci. 2013, 70, 2657–2675. [Google Scholar] [CrossRef] [PubMed]
  143. Duan, M.; Liu, H.; Xu, S.; Yang, Z.; Zhang, F.; Wang, G.; Wang, Y.; Zhao, S.; Jiang, X. IGF2BPs as novel m(6)A readers: Diverse roles in regulating cancer cell biological functions, hypoxia adaptation, metabolism, and immunosuppressive tumor microenvironment. Genes. Dis. 2024, 11, 890–920. [Google Scholar] [CrossRef] [PubMed]
  144. Glass, M.; Misiak, D.; Bley, N.; Muller, S.; Hagemann, S.; Busch, B.; Rausch, A.; Huttelmaier, S. IGF2BP1, a Conserved Regulator of RNA Turnover in Cancer. Front. Mol. Biosci. 2021, 8, 632219. [Google Scholar] [CrossRef] [PubMed]
  145. Huang, H.; Weng, H.; Sun, W.; Qin, X.; Shi, H.; Wu, H.; Zhao, B.S.; Mesquita, A.; Liu, C.; Yuan, C.L.; et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 2018, 20, 285–295. [Google Scholar] [CrossRef]
  146. Muller, S.; Bley, N.; Glass, M.; Busch, B.; Rousseau, V.; Misiak, D.; Fuchs, T.; Lederer, M.; Huttelmaier, S. IGF2BP1 enhances an aggressive tumor cell phenotype by impairing miRNA-directed downregulation of oncogenic factors. Nucleic Acids Res. 2018, 46, 6285–6303. [Google Scholar] [CrossRef]
  147. Wan, B.S.; Cheng, M.; Zhang, L. Insulin-like growth factor 2 mRNA-binding protein 1 promotes cell proliferation via activation of AKT and is directly targeted by microRNA-494 in pancreatic cancer. World J. Gastroenterol. 2019, 25, 6063–6076. [Google Scholar] [CrossRef]
  148. Di Fusco, D.; Segreto, M.T.; Di Maggio, G.; Iannucci, A.; Maresca, C.; Di Grazia, A.; Colella, M.; Stolfi, C.; Monteleone, G.; Monteleone, I. Insulin-like Growth Factor II mRNA-Binding Protein 1 Regulates Pancreatic Cancer Cell Growth through the Surveillance of CDC25A mRNA. Cancers 2023, 15, 4983. [Google Scholar] [CrossRef]
  149. Zhang, Z.; Yu, H.; Yao, W.; Zhu, N.; Miao, R.; Liu, Z.; Song, X.; Xue, C.; Cai, C.; Cheng, M.; et al. RRP9 promotes gemcitabine resistance in pancreatic cancer via activating AKT signaling pathway. Cell Commun. Signal 2022, 20, 188. [Google Scholar] [CrossRef]
  150. Muller, S.; Bley, N.; Busch, B.; Glass, M.; Lederer, M.; Misiak, C.; Fuchs, T.; Wedler, A.; Haase, J.; Bertoldo, J.B.; et al. The oncofetal RNA-binding protein IGF2BP1 is a druggable, post-transcriptional super-enhancer of E2F-driven gene expression in cancer. Nucleic Acids Res. 2020, 48, 8576–8590. [Google Scholar] [CrossRef]
  151. Lin, C.; Wang, Y.; Dong, Y.; Lai, S.; Wang, L.; Weng, S.; Zhang, X. N6-methyladenosine-mediated SH3BP5-AS1 upregulation promotes GEM chemoresistance in pancreatic cancer by activating the Wnt signaling pathway. Biol. Direct 2022, 17, 33. [Google Scholar] [CrossRef]
  152. Cao, P.; Zhang, W.; Qiu, J.; Tang, Z.; Xue, X.; Feng, T. Gemcitabine Inhibits the Progression of Pancreatic Cancer by Restraining the WTAP/MYC Chain in an m6A-Dependent Manner. Cancer Res. Treat. 2024, 56, 259–271. [Google Scholar] [CrossRef] [PubMed]
  153. Zhu, S.; Wang, J.Z.; Chen, D.; He, Y.T.; Meng, N.; Chen, M.; Lu, R.X.; Chen, X.H.; Zhang, X.L.; Yan, G.R. An oncopeptide regulates m(6)A recognition by the m(6)A reader IGF2BP1 and tumorigenesis. Nat. Commun. 2020, 11, 1685. [Google Scholar] [CrossRef]
  154. Xu, X.; Yu, Y.; Zong, K.; Lv, P.; Gu, Y. Up-regulation of IGF2BP2 by multiple mechanisms in pancreatic cancer promotes cancer proliferation by activating the PI3K/Akt signaling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 497. [Google Scholar] [CrossRef] [PubMed]
  155. Dahlem, C.; Barghash, A.; Puchas, P.; Haybaeck, J.; Kessler, S.M. The Insulin-Like Growth Factor 2 mRNA Binding Protein IMP2/IGF2BP2 is Overexpressed and Correlates with Poor Survival in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 3204. [Google Scholar] [CrossRef] [PubMed]
  156. Peng, W.X.; Liu, F.; Jiang, J.H.; Yuan, H.; Zhang, Z.; Yang, L.; Mo, Y.Y. N6-methyladenosine modified LINC00901 promotes pancreatic cancer progression through IGF2BP2/MYC axis. Genes. Dis. 2023, 10, 554–567. [Google Scholar] [CrossRef]
  157. Yao, Y.; Luo, L.; Xiang, G.; Xiong, J.; Ke, N.; Tan, C.; Chen, Y.; Liu, X. The expression of m(6)A regulators correlated with the immune microenvironment plays an important role in the prognosis of pancreatic ductal adenocarcinoma. Gland. Surg. 2022, 11, 147–165. [Google Scholar] [CrossRef]
  158. Deng, H.; Yao, H.; Zhou, S.; He, C.; Huang, Y.; Li, Y.; Chen, H.; Shu, J. Pancancer analysis uncovers an immunological role and prognostic value of the m6A reader IGF2BP2 in pancreatic cancer. Mol. Cell Probes 2024, 73, 101948. [Google Scholar] [CrossRef]
  159. Huang, S.; Wu, Z.; Cheng, Y.; Wei, W.; Hao, L. Insulin-like growth factor 2 mRNA binding protein 2 promotes aerobic glycolysis and cell proliferation in pancreatic ductal adenocarcinoma via stabilizing GLUT1 mRNA. Acta Biochim. Biophys. Sin. 2019, 51, 743–752. [Google Scholar] [CrossRef]
  160. Liu, X.; Chen, J.; Chen, W.; Xu, Y.; Shen, Y.; Xu, X. Targeting IGF2BP3 in Cancer. Int. J. Mol. Sci. 2023, 24, 9423. [Google Scholar] [CrossRef]
  161. Mueller-Pillasch, F.; Lacher, U.; Wallrapp, C.; Micha, A.; Zimmerhackl, F.; Hameister, H.; Varga, G.; Friess, H.; Buchler, M.; Beger, H.G.; et al. Cloning of a gene highly overexpressed in cancer coding for a novel KH-domain containing protein. Oncogene 1997, 14, 2729–2733. [Google Scholar] [CrossRef]
  162. Wang, B.J.; Wang, L.; Yang, S.Y.; Liu, Z.J. Expression and clinical significance of IMP3 in microdissected premalignant and malignant pancreatic lesions. Clin. Transl. Oncol. 2015, 17, 215–222. [Google Scholar] [CrossRef]
  163. Senoo, J.; Mikata, R.; Kishimoto, T.; Hayashi, M.; Kusakabe, Y.; Yasui, S.; Yamato, M.; Ohyama, H.; Sugiyama, H.; Tsuyuguchi, T.; et al. Immunohistochemical analysis of IMP3 and p53 expression in endoscopic ultrasound-guided fine needle aspiration and resected specimens of pancreatic diseases. Pancreatology 2018, 18, 176–183. [Google Scholar] [CrossRef]
  164. Johnson, B.; Khalil, M.; Blansfield, J.; Lin, F.; Zhu, S.; Kirchner, H.L.; Weir, A.B., 3rd. Investigating the prognostic value of KOC (K homology domain containing protein overexpressed in cancer) overexpression after curative intent resection of pancreatic ductal adenocarcinoma. J. Gastrointest. Oncol. 2016, 7, E113–E117. [Google Scholar] [CrossRef] [PubMed]
  165. Li, H.; Wang, X.; Fang, Y.; Huo, Z.; Lu, X.; Zhan, X.; Deng, X.; Peng, C.; Shen, B. Integrated expression profiles analysis reveals novel predictive biomarker in pancreatic ductal adenocarcinoma. Oncotarget 2017, 8, 52571–52583. [Google Scholar] [CrossRef] [PubMed]
  166. Yantiss, R.K.; Cosar, E.; Fischer, A.H. Use of IMP3 in identification of carcinoma in fine needle aspiration biopsies of pancreas. Acta Cytol. 2008, 52, 133–138. [Google Scholar] [CrossRef]
  167. Wachter, D.L.; Schlabrakowski, A.; Hoegel, J.; Kristiansen, G.; Hartmann, A.; Riener, M.O. Diagnostic value of immunohistochemical IMP3 expression in core needle biopsies of pancreatic ductal adenocarcinoma. Am. J. Surg. Pathol. 2011, 35, 873–877. [Google Scholar] [CrossRef] [PubMed]
  168. Mikata, R.; Yasui, S.; Kishimoto, T.; Kouchi, Y.; Shingyoji, A.; Senoo, J.; Takahashi, K.; Nagashima, H.; Kusakabe, Y.; Ohyama, H.; et al. Diagnostic value of IMP3 and p53 immunohistochemical staining in EUS-guided fine-needle aspiration for solid pancreatic tumors. Sci. Rep. 2021, 11, 17257. [Google Scholar] [CrossRef]
  169. Ennajdaoui, H.; Howard, J.M.; Sterne-Weiler, T.; Jahanbani, F.; Coyne, D.J.; Uren, P.J.; Dargyte, M.; Katzman, S.; Draper, J.M.; Wallace, A.; et al. IGF2BP3 Modulates the Interaction of Invasion-Associated Transcripts with RISC. Cell Rep. 2016, 15, 1876–1883. [Google Scholar] [CrossRef]
  170. Guo, Z.; Zhang, X.; Lin, C.; Huang, Y.; Zhong, Y.; Guo, H.; Zheng, Z.; Weng, S. METTL3-IGF2BP3-axis mediates the proliferation and migration of pancreatic cancer by regulating spermine synthase m6A modification. Front. Oncol. 2022, 12, 962204. [Google Scholar] [CrossRef]
  171. Pasiliao, C.C.; Chang, C.W.; Sutherland, B.W.; Valdez, S.M.; Schaeffer, D.; Yapp, D.T.; Ng, S.S. The involvement of insulin-like growth factor 2 binding protein 3 (IMP3) in pancreatic cancer cell migration, invasion, and adhesion. BMC Cancer 2015, 15, 266. [Google Scholar] [CrossRef] [PubMed]
  172. Taniuchi, K.; Furihata, M.; Hanazaki, K.; Saito, M.; Saibara, T. IGF2BP3-mediated translation in cell protrusions promotes cell invasiveness and metastasis of pancreatic cancer. Oncotarget 2014, 5, 6832–6845. [Google Scholar] [CrossRef] [PubMed]
  173. Taniuchi, K.; Furihata, M.; Saibara, T. KIF20A-mediated RNA granule transport system promotes the invasiveness of pancreatic cancer cells. Neoplasia 2014, 16, 1082–1093. [Google Scholar] [CrossRef]
  174. Fu, W.; Lei, X.; Lu, Q.; Zhang, J.; Guo, J.; Zhao, J.; Tong, X.; Hu, X. UBE2K regulated by IGF2BP3 promotes cell proliferation and stemness in pancreatic ductal adenocarcinoma. Int. J. Oncol. 2023, 62, 52. [Google Scholar] [CrossRef] [PubMed]
  175. Dowdle, M.E.; Kanzler, C.R.; Harder, C.R.K.; Moffet, S.; Walker, M.N.; Sheets, M.D. Bicaudal-C Post-transcriptional regulator of cell fates and functions. Front. Cell Dev. Biol. 2022, 10, 981696. [Google Scholar] [CrossRef]
  176. Mahone, M.; Saffman, E.E.; Lasko, P.F. Localized Bicaudal-C RNA encodes a protein containing a KH domain, the RNA binding motif of FMR1. EMBO J. 1995, 14, 2043–2055. [Google Scholar] [CrossRef]
  177. Mohler, J.; Wieschaus, E.F. Dominant maternal-effect mutations of Drosophila melanogaster causing the production of double-abdomen embryos. Genetics 1986, 112, 803–822. [Google Scholar] [CrossRef]
  178. Saffman, E.E.; Styhler, S.; Rother, K.; Li, W.; Richard, S.; Lasko, P. Premature translation of oskar in oocytes lacking the RNA-binding protein bicaudal-C. Mol. Cell Biol. 1998, 18, 4855–4862. [Google Scholar] [CrossRef]
  179. Meng, F.; Hua, S.; Chen, X.; Meng, N.; Lan, T. Lymph node metastasis related gene BICC1 promotes tumor progression by promoting EMT and immune infiltration in pancreatic cancer. BMC Med. Genom. 2023, 16, 263. [Google Scholar] [CrossRef]
  180. Huang, C.; Li, H.; Xu, Y.; Xu, C.; Sun, H.; Li, Z.; Ge, Y.; Wang, H.; Zhao, T.; Gao, S.; et al. BICC1 drives pancreatic cancer progression by inducing VEGF-independent angiogenesis. Signal Transduct. Target. Ther. 2023, 8, 271. [Google Scholar] [CrossRef]
  181. Sun, H.; Li, H.; Guan, Y.; Yuan, Y.; Xu, C.; Fu, D.; Xie, P.; Li, J.; Zhao, T.; Wang, X.; et al. BICC1 drives pancreatic cancer stemness and chemoresistance by facilitating tryptophan metabolism. Sci. Adv. 2024, 10, eadj8650. [Google Scholar] [CrossRef] [PubMed]
  182. Pu, J.; Zhang, X.; Luo, H.; Xu, L.; Lu, X.; Lu, J. Adrenaline promotes epithelial-to-mesenchymal transition via HuR-TGFbeta regulatory axis in pancreatic cancer cells and the implication in cancer prognosis. Biochem. Biophys. Res. Commun. 2017, 493, 1273–1279. [Google Scholar] [CrossRef] [PubMed]
  183. Hu, X.; Lei, X.; Guo, J.; Fu, W.; Sun, W.; Lu, Q.; Su, W.; Xu, Q.; Tu, K. The Emerging Role of RNA N6-Methyladenosine Modification in Pancreatic Cancer. Front. Oncol. 2022, 12, 927640. [Google Scholar] [CrossRef]
  184. Subramaniam, D.; Ramalingam, S.; Linehan, D.C.; Dieckgraefe, B.K.; Postier, R.G.; Houchen, C.W.; Jensen, R.A.; Anant, S. RNA binding protein CUGBP2/CELF2 mediates curcumin-induced mitotic catastrophe of pancreatic cancer cells. PLoS ONE 2011, 6, e16958. [Google Scholar] [CrossRef] [PubMed]
  185. Martinez-Useros, J.; Georgiev-Hristov, T.; Fernandez-Acenero, M.J.; Borrero-Palacios, A.; Indacochea, A.; Guerrero, S.; Li, W.; Cebrian, A.; Gomez Del Pulgar, T.; Puime-Otin, A.; et al. UNR/CDSE1 expression as prognosis biomarker in resectable pancreatic ductal adenocarcinoma patients: A proof-of-concept. PLoS ONE 2017, 12, e0182044. [Google Scholar] [CrossRef]
  186. Zhang, L.; Yang, L.; Du, K. Exosomal HSPB1, interacting with FUS protein, suppresses hypoxia-induced ferroptosis in pancreatic cancer by stabilizing Nrf2 mRNA and repressing P450. J. Cell Mol. Med. 2024, 28, e18209. [Google Scholar] [CrossRef]
  187. Yan-Sanders, Y.; Hammons, G.J.; Lyn-Cook, B.D. Increased expression of heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP) in pancreatic tissue from smokers and pancreatic tumor cells. Cancer Lett. 2002, 183, 215–220. [Google Scholar] [CrossRef]
  188. Chen, Z.Y.; Cai, L.; Zhu, J.; Chen, M.; Chen, J.; Li, Z.H.; Liu, X.D.; Wang, S.G.; Bie, P.; Jiang, P.; et al. Fyn requires HnRNPA2B1 and Sam68 to synergistically regulate apoptosis in pancreatic cancer. Carcinogenesis 2011, 32, 1419–1426. [Google Scholar] [CrossRef]
  189. Yang, N.; Liu, L.; Liu, X.; Chen, Y.; Lu, J.; Wang, Z. hnRNPC Promotes Malignancy in Pancreatic Cancer through Stabilization of IQGAP3. Biomed. Res. Int. 2022, 2022, 6319685. [Google Scholar] [CrossRef]
  190. Antal, C.E.; Oh, T.G.; Aigner, S.; Luo, E.C.; Yee, B.A.; Campos, T.; Tiriac, H.; Rothamel, K.L.; Cheng, Z.; Jiao, H.; et al. A super-enhancer-regulated RNA-binding protein cascade drives pancreatic cancer. Nat. Commun. 2023, 14, 5195. [Google Scholar] [CrossRef]
  191. Qiao, L.; Xie, N.; Bai, Y.; Li, Y.; Shi, Y.; Wang, J.; Liu, N. Identification of Upregulated HNRNPs Associated with Poor Prognosis in Pancreatic Cancer. Biomed. Res. Int. 2019, 2019, 5134050. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, Y.; Li, J.; Guo, S.; Ouyang, Y.; Yin, L.; Liu, S.; Zhao, Z.; Yang, J.; Huang, W.; Qin, H.; et al. Lin28B facilitates the progression and metastasis of pancreatic ductal adenocarcinoma. Oncotarget 2017, 8, 60414–60428. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, Y.; Wang, D.; Zhou, M.; Chen, H.; Wang, H.; Min, J.; Chen, J.; Wu, S.; Ni, X.; Zhang, Y.; et al. The KRAS/Lin28B axis maintains stemness of pancreatic cancer cells via the let-7i/TET3 pathway. Mol. Oncol. 2021, 15, 262–278. [Google Scholar] [CrossRef]
  194. Shu, Z.; Fan, M.; Tu, B.; Tang, Z.; Wang, H.; Li, H.; Li, H.; Yuan, M.; Bai, J.; Huo, S.; et al. The Lin28b/Wnt5a axis drives pancreas cancer through crosstalk between cancer associated fibroblasts and tumor epithelium. Nat. Commun. 2023, 14, 6885. [Google Scholar] [CrossRef]
  195. Panzeri, V.; Manni, I.; Capone, A.; Naro, C.; Sacconi, A.; Di Agostino, S.; de Latouliere, L.; Montori, A.; Pilozzi, E.; Piaggio, G.; et al. The RNA-binding protein MEX3A is a prognostic factor and regulator of resistance to gemcitabine in pancreatic ductal adenocarcinoma. Mol. Oncol. 2021, 15, 579–595. [Google Scholar] [CrossRef]
  196. Sheng, W.; Dong, M.; Chen, C.; Wang, Z.; Li, Y.; Wang, K.; Li, Y.; Zhou, J. Cooperation of Musashi-2, Numb, MDM2, and P53 in drug resistance and malignant biology of pancreatic cancer. FASEB J. 2017, 31, 2429–2438. [Google Scholar] [CrossRef] [PubMed]
  197. Guo, K.; Cui, J.; Quan, M.; Xie, D.; Jia, Z.; Wei, D.; Wang, L.; Gao, Y.; Ma, Q.; Xie, K. The Novel KLF4/MSI2 Signaling Pathway Regulates Growth and Metastasis of Pancreatic Cancer. Clin. Cancer Res. 2017, 23, 687–696. [Google Scholar] [CrossRef]
  198. Yang, H.; Hu, J.; Chen, J.; Chen, Z.; Jiao, F.; Cui, J.; Quan, M.; Wang, L. RNA-binding protein Musashi2 regulates Hippo signaling via SAV1 and MOB1 in pancreatic cancer. Med. Oncol. 2020, 37, 84. [Google Scholar] [CrossRef]
  199. Sheng, W.; Dong, M.; Chen, C.; Li, Y.; Liu, Q.; Dong, Q. Musashi2 promotes the development and progression of pancreatic cancer by down-regulating Numb protein. Oncotarget 2017, 8, 14359–14373. [Google Scholar] [CrossRef]
  200. Chu, L.; Hu, Y.; Jiang, Y.H.; Xu, C.; Liu, W.C.; Lu, Z.F. Effects of RNA binding protein QKI on pancreatic cancer ductal epithelial cells and surrounding activation fibroblasts. J. Cell Biochem. 2019, 120, 11551–11561. [Google Scholar] [CrossRef]
  201. Maurin, M.; Ranjouri, M.; Megino-Luque, C.; Newberg, J.Y.; Du, D.; Martin, K.; Miner, R.E., 3rd; Prater, M.S.; Wee, D.K.B.; Centeno, B.; et al. RBFOX2 deregulation promotes pancreatic cancer progression and metastasis through alternative splicing. Nat. Commun. 2023, 14, 8444. [Google Scholar] [CrossRef] [PubMed]
  202. Witkiewicz, A.K.; McMillan, E.A.; Balaji, U.; Baek, G.; Lin, W.C.; Mansour, J.; Mollaee, M.; Wagner, K.U.; Koduru, P.; Yopp, A.; et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 2015, 6, 6744. [Google Scholar] [CrossRef]
  203. Xiao, W.; Chen, X.; Li, X.; Deng, K.; Liu, H.; Ma, J.; Wang, Z.; Hu, Y.; Hou, J. RBM10 regulates human TERT gene splicing and inhibits pancreatic cancer progression. Am. J. Cancer Res. 2021, 11, 157–170. [Google Scholar] [PubMed]
  204. Iwata, T.; Kishikawa, T.; Seimiya, T.; Notoya, G.; Suzuki, T.; Shibata, C.; Miyakawa, Y.; Odawara, N.; Funato, K.; Tanaka, E.; et al. Satellite double-stranded RNA induces mesenchymal transition in pancreatic cancer by regulating alternative splicing. J. Biol. Chem. 2024, 300, 105742. [Google Scholar] [CrossRef] [PubMed]
  205. Zhao, X.; Chen, M.; Tan, J. Knockdown of ZFR suppresses cell proliferation and invasion of human pancreatic cancer. Biol. Res. 2016, 49, 26. [Google Scholar] [CrossRef]
  206. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef]
  207. Tani, H. Recent Advances and Prospects in RNA Drug Development. Int. J. Mol. Sci. 2024, 25, 12284. [Google Scholar] [CrossRef]
  208. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
  209. Liang, X.H.; Sun, H.; Nichols, J.G.; Crooke, S.T. RNase H1-Dependent Antisense Oligonucleotides Are Robustly Active in Directing RNA Cleavage in Both the Cytoplasm and the Nucleus. Mol. Ther. 2017, 25, 2075–2092. [Google Scholar] [CrossRef]
  210. Gutschner, T.; Hammerle, M.; Eissmann, M.; Hsu, J.; Kim, Y.; Hung, G.; Revenko, A.; Arun, G.; Stentrup, M.; Gross, M.; et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013, 73, 1180–1189. [Google Scholar] [CrossRef]
  211. Neil, E.E.; Bisaccia, E.K. Nusinersen: A Novel Antisense Oligonucleotide for the Treatment of Spinal Muscular Atrophy. J. Pediatr. Pharmacol. Ther. 2019, 24, 194–203. [Google Scholar] [CrossRef] [PubMed]
  212. Ammala, C.; Drury, W.J., 3rd; Knerr, L.; Ahlstedt, I.; Stillemark-Billton, P.; Wennberg-Huldt, C.; Andersson, E.M.; Valeur, E.; Jansson-Lofmark, R.; Janzen, D.; et al. Targeted delivery of antisense oligonucleotides to pancreatic beta-cells. Sci. Adv. 2018, 4, eaat3386. [Google Scholar] [CrossRef]
  213. Childs-Disney, J.L.; Yang, X.; Gibaut, Q.M.R.; Tong, Y.; Batey, R.T.; Disney, M.D. Targeting RNA structures with small molecules. Nat. Rev. Drug Discov. 2022, 21, 736–762. [Google Scholar] [CrossRef] [PubMed]
  214. Jaiswal, A.K.; Thaxton, M.L.; Scherer, G.M.; Sorrentino, J.P.; Garg, N.K.; Rao, D.S. Small molecule inhibition of RNA binding proteins in haematologic cancer. RNA Biol. 2024, 21, 276–289. [Google Scholar] [CrossRef]
  215. Li, Q.; Kang, C. Targeting RNA-binding proteins with small molecules: Perspectives, pitfalls and bifunctional molecules. FEBS Lett. 2023, 597, 2031–2047. [Google Scholar] [CrossRef]
  216. Nguyen, L.D.; Chau, R.K.; Krichevsky, A.M. Small Molecule Drugs Targeting Non-Coding RNAs as Treatments for Alzheimer’s Disease and Related Dementias. Genes 2021, 12, 2005. [Google Scholar] [CrossRef]
  217. Zhao, R.; Fu, J.; Zhu, L.; Chen, Y.; Liu, B. Designing strategies of small-molecule compounds for modulating non-coding RNAs in cancer therapy. J. Hematol. Oncol. 2022, 15, 14. [Google Scholar] [CrossRef] [PubMed]
  218. Bertoldo, J.B.; Muller, S.; Huttelmaier, S. RNA-binding proteins in cancer drug discovery. Drug Discov. Today 2023, 28, 103580. [Google Scholar] [CrossRef]
  219. Bekes, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200. [Google Scholar] [CrossRef]
  220. Gama-Brambila, R.A.; Chen, J.; Zhou, J.; Tascher, G.; Munch, C.; Cheng, X. A PROTAC targets splicing factor 3B1. Cell Chem. Biol. 2021, 28, 1616–1627.e1618. [Google Scholar] [CrossRef]
  221. Kaur, T.; Menon, A.; Garner, A.L. Synthesis of 7-benzylguanosine cap-analogue conjugates for eIF4E targeted degradation. Eur. J. Med. Chem. 2019, 166, 339–350. [Google Scholar] [CrossRef] [PubMed]
  222. Ghidini, A.; Clery, A.; Halloy, F.; Allain, F.H.T.; Hall, J. RNA-PROTACs: Degraders of RNA-Binding Proteins. Angew. Chem. Int. Ed. Engl. 2021, 60, 3163–3169. [Google Scholar] [CrossRef]
  223. Costales, M.G.; Matsumoto, Y.; Velagapudi, S.P.; Disney, M.D. Small Molecule Targeted Recruitment of a Nuclease to RNA. J. Am. Chem. Soc. 2018, 140, 6741–6744. [Google Scholar] [CrossRef] [PubMed]
  224. Costales, M.G.; Aikawa, H.; Li, Y.; Childs-Disney, J.L.; Abegg, D.; Hoch, D.G.; Pradeep Velagapudi, S.; Nakai, Y.; Khan, T.; Wang, K.W.; et al. Small-molecule targeted recruitment of a nuclease to cleave an oncogenic RNA in a mouse model of metastatic cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 2406–2411. [Google Scholar] [CrossRef]
  225. Meyer, S.M.; Tanaka, T.; Zanon, P.R.A.; Baisden, J.T.; Abegg, D.; Yang, X.; Akahori, Y.; Alshakarchi, Z.; Cameron, M.D.; Adibekian, A.; et al. DNA-Encoded Library Screening to Inform Design of a Ribonuclease Targeting Chimera (RiboTAC). J. Am. Chem. Soc. 2022, 144, 21096–21102. [Google Scholar] [CrossRef]
  226. Tong, Y.; Lee, Y.; Liu, X.; Childs-Disney, J.L.; Suresh, B.M.; Benhamou, R.I.; Yang, C.; Li, W.; Costales, M.G.; Haniff, H.S.; et al. Programming inactive RNA-binding small molecules into bioactive degraders. Nature 2023, 618, 169–179. [Google Scholar] [CrossRef] [PubMed]
  227. Cirino, N.M.; Li, G.; Xiao, W.; Torrence, P.F.; Silverman, R.H. Targeting RNA decay with 2′,5′ oligoadenylate-antisense in respiratory syncytial virus-infected cells. Proc. Natl. Acad. Sci. USA 1997, 94, 1937–1942. [Google Scholar] [CrossRef]
  228. Fang, Y.; Wu, Q.; Wang, F.; Liu, Y.; Zhang, H.; Yang, C.; Zhu, Z. Aptamer-RIBOTAC Strategy Enabling Tumor-Specific Targeted Degradation of MicroRNA for Precise Cancer Therapy. In Small Methods; Wiley: New York, NY, USA, 2024; p. e2400349. [Google Scholar] [CrossRef]
  229. Su, X.; Ma, W.; Feng, D.; Cheng, B.; Wang, Q.; Guo, Z.; Zhou, D.; Tang, X. Efficient Inhibition of SARS-CoV-2 Using Chimeric Antisense Oligonucleotides through RNase L Activation. Angew. Chem. Int. Ed. Engl. 2021, 60, 21662–21667. [Google Scholar] [CrossRef]
  230. Torrence, P.F.; Maitra, R.K.; Lesiak, K.; Khamnei, S.; Zhou, A.; Silverman, R.H. Targeting RNA for degradation with a (2′-5′)oligoadenylate-antisense chimera. Proc. Natl. Acad. Sci. USA 1993, 90, 1300–1304. [Google Scholar] [CrossRef]
  231. Lu, J.; Yu, L.; Xie, N.; Wu, Y.; Li, B. METTL14 Facilitates the Metastasis of Pancreatic Carcinoma by Stabilizing LINC00941 in an m6A-IGF2BP2-Dependent Manner. J. Cancer 2023, 14, 1117–1131. [Google Scholar] [CrossRef]
  232. Zhai, S.; Xu, Z.; Xie, J.; Zhang, J.; Wang, X.; Peng, C.; Li, H.; Chen, H.; Shen, B.; Deng, X. Epigenetic silencing of LncRNA LINC00261 promotes c-myc-mediated aerobic glycolysis by regulating miR-222-3p/HIPK2/ERK axis and sequestering IGF2BP1. Oncogene 2021, 40, 277–291. [Google Scholar] [CrossRef] [PubMed]
  233. Athanasiou, A.; Kureshi, N.; Wittig, A.; Sterner, M.; Huber, R.; Palma, N.A.; King, T.; Schiess, R. Biomarker Discovery for Early Detection of Pancreatic Ductal Adenocarcinoma (PDAC) Using Multiplex Proteomics Technology. J. Proteome Res. 2025, 24, 315–322. [Google Scholar] [CrossRef] [PubMed]
  234. Khomiak, A.; Brunner, M.; Kordes, M.; Lindblad, S.; Miksch, R.C.; Ohlund, D.; Regel, I. Recent Discoveries of Diagnostic, Prognostic and Predictive Biomarkers for Pancreatic Cancer. Cancers 2020, 12, 3234. [Google Scholar] [CrossRef] [PubMed]
  235. Bin, W.; Yuan, C.; Qie, Y.; Dang, S. Long non-coding RNAs and pancreatic cancer: A multifaceted view. Biomed. Pharmacother. 2023, 167, 115601. [Google Scholar] [CrossRef]
  236. von Widdern, J.C.; Rosendahl, J.; Ammer-Herrmenau, C. Chronic and Idiopathic Pancreatitis-A Personalized Treatment Approach. United Eur. Gastroenterol. J. 2025, 13, 116–124. [Google Scholar] [CrossRef]
Figure 1. Long non-coding RNA (lncRNA)-guided functions and regulatory mechanisms involved in PDAC development and progression. Identified mechanisms (a) and cellular functions (b) of selected lncRNAs in pancreatic cancer, specifically in the context of KRAS/MAPK and TGF-/SMAD signalling, have been described in the main text and are summarized here schematically, with implicated promoting (orange) or inhibitory (turquois) lncRNAs assigned to their respective function. For more details about the clinical relevance and the mode of action of these lncRNAs, see Table 2.
Figure 1. Long non-coding RNA (lncRNA)-guided functions and regulatory mechanisms involved in PDAC development and progression. Identified mechanisms (a) and cellular functions (b) of selected lncRNAs in pancreatic cancer, specifically in the context of KRAS/MAPK and TGF-/SMAD signalling, have been described in the main text and are summarized here schematically, with implicated promoting (orange) or inhibitory (turquois) lncRNAs assigned to their respective function. For more details about the clinical relevance and the mode of action of these lncRNAs, see Table 2.
Cancers 17 01601 g001
Figure 2. RNA-binding protein-guided functions and regulatory mechanisms involved in PDAC development and progression. Indicated cellular (a) and molecular functions (b) are depicted schematically with implicated promoting (orange) or inhibitory (turquois) RBPs assigned to their respective functions. Importantly, several RBPs have been shown to modulate cancer hallmarks by applying one or more modes of action to regulate their target transcripts.
Figure 2. RNA-binding protein-guided functions and regulatory mechanisms involved in PDAC development and progression. Indicated cellular (a) and molecular functions (b) are depicted schematically with implicated promoting (orange) or inhibitory (turquois) RBPs assigned to their respective functions. Importantly, several RBPs have been shown to modulate cancer hallmarks by applying one or more modes of action to regulate their target transcripts.
Cancers 17 01601 g002
Figure 3. Summary of RBP- and lncRNA-directed therapeutic strategies and their current application in pancreatic cancer. Most strategies are currently at the early stages and have been tested in cell lines and pre-clinical mouse models. The targeting strategies can be divided into two groups. RNA interference (RNAi) and antisense oligonucleotides (ASOs). Ribonuclease-targeting chimeras (RIBOTACs) are generally RNA-directed approaches and can be used to degrade RBP-encoding mRNAs as well as lncRNAs. In contrast, small molecules and proteolysis-targeting chimeras (PROTACs) aim to target the RNA-binding protein directly and can be used to either interfere with its molecular interactions, i.e., binding to RNA, or to recruit an E3 ubiquitin ligase to induce the destruction of the RBP via the ubiquitin–proteasome system. Small molecules can also be directed against structural elements in lncRNAs to interfere with molecular interactions. The identification of specific and high-affinity binding compounds and the tumour-specific delivery of these substances pose major challenges for their clinical application in PDAC patients.
Figure 3. Summary of RBP- and lncRNA-directed therapeutic strategies and their current application in pancreatic cancer. Most strategies are currently at the early stages and have been tested in cell lines and pre-clinical mouse models. The targeting strategies can be divided into two groups. RNA interference (RNAi) and antisense oligonucleotides (ASOs). Ribonuclease-targeting chimeras (RIBOTACs) are generally RNA-directed approaches and can be used to degrade RBP-encoding mRNAs as well as lncRNAs. In contrast, small molecules and proteolysis-targeting chimeras (PROTACs) aim to target the RNA-binding protein directly and can be used to either interfere with its molecular interactions, i.e., binding to RNA, or to recruit an E3 ubiquitin ligase to induce the destruction of the RBP via the ubiquitin–proteasome system. Small molecules can also be directed against structural elements in lncRNAs to interfere with molecular interactions. The identification of specific and high-affinity binding compounds and the tumour-specific delivery of these substances pose major challenges for their clinical application in PDAC patients.
Cancers 17 01601 g003
Table 1. Overview of common modes of action of long non-coding RNAs (lncRNAs).
Table 1. Overview of common modes of action of long non-coding RNAs (lncRNAs).
FunctionMode of ActionDescriptionExamples
Enhancer
action
cis/transA total of 30–60% of lncRNAs are transcribed from enhancer regions, and enhancer activity is modulated by the active transcription and splicing of enhancer lncRNAs [44,45,46]. These transcripts may potentiate enhancer activity by recruiting proteins that participate in the formation of chromatin loops between distal enhancers and their promoters or direct the recruitment of transcriptional activators to gene-proximal enhancers [47,48,49]. Similar mechanisms can also result in the repression of enhancer activity when lncRNA loci compete for enhancers with protein-coding genes [50].Hand2os1 [47,48]
CCAT1-L [49]
PVT1 [50]
Chromatin
architecture
modulation
cis/transThe process of X chromosome inactivation is dependent on the cis-acting lncRNA XIST, which spreads along and coats the inactive chromosome, ensuring dosage compensation [31,51]. Additionally, association with chromatin remodelling complexes is a common feature among lncRNAs [52]. The polycomp repressive complex 2 (PRC2) in particular is functionally dependent on RNAs and has been shown to interact with ~20% of human lncRNAs [52,53]. HOTAIR [30,54]
XIST [31,51]
Biomolecular
condensate
formation
transIntracellular condensates are formed from RNA and protein interactions through liquid–liquid phase separation. Many of these compartments depend on architectural lncRNAs for their structures, compositions and functions [55].NORAD [36]
NEAT1 [40]
MALAT1 [56]
miRNA
sponge
transThe target sites of miRNAs are found in mRNAs but are also present in many types of ncRNAs including lncRNAs. This has led to the hypothesis that all transcripts sharing binding sites for a particular miRNA may regulate each other by competing against each other [57]. An extensive number of publications have focused on the action of lncRNAs as competing endogenous RNAs (ceRNAs), but the hypothesis has also received criticism relating to the stoichiometries needed to support ceRNA regulatory networks [58].LINCMD1 [59],
PTENP1 [60,61]
Table 2. LncRNAs associated with KRAS/MAPK and TGF-β signalling in pancreatic cancer.
Table 2. LncRNAs associated with KRAS/MAPK and TGF-β signalling in pancreatic cancer.
LncRNAExpression in PDACClinical AssociationFunction and MechanismReferences
NUTF2P3-01upregulatedhigh expression correlates with tumour size; poor tumour differentiation; the tumour, node and metastasis (TNM) stage; lymphatic invasion; distant metastasis; and shorter overall survival (OS)promotes proliferation, invasion, xenograft tumour growth and hepatic metastasis and acts as a ceRNA to sequester miR-3923 and increase KRAS expression[90]
MALAT1upregulatedhigh expression is associated with shorter OSpromotes proliferation, migration, invasion, and xenograft growth; reduces apoptosis; and acts as a ceRNA to sequester miR-217 and increase KRAS expression[91]
UCA1/CUDRupregulatedhigh expression correlates with an advanced T and N stage and is associated with poor OSpromotes proliferation, migration, drug resistance and xenograft tumour growth; reduces apoptosis; positively regulates MAPK/ERK and AKT/FAK signalling; enhances the interaction between KRAS and hnRNPA2/B1; and acts as a ceRNA to sequester miR-590-3p and increase KRAS expression [93,95,96]
ABHD11-AS1upregulatedexpression correlates with poor OS and disease-free survival (DFS), an advanced TNM stage, increased distant metastasis and poor tumour differentiationpromotes proliferation and migration, reduces apoptosis and positively regulates PI3K/AKT signalling and EMT[98]
SNHG1upregulatedexpression correlates with an advanced TNM stage and a larger tumour sizepromotes proliferation, invasion and xenograft tumour growth; reduces apoptosis; and positively regulates PI3K/AKT signalling [99]
LUCAT1upregulatedincreased expression linked to a larger tumour size and lymphatic invasionpromotes proliferation, invasion and xenograft tumour growth; reduces apoptosis; and positively regulates AKT and p38-MAPK signalling and the sponge of miR-539[100,101]
LINC01232upregulatedexpression is positively correlated with nerve invasion, and high LINC01232 levels are associated with worse outcomespromotes migration and invasion and regulates the alternative splicing of ARAF by stabilizing hnRNPA2/B1, thereby activating MAPK/ERK signalling[102]
LINC00941upregulatedhigh expression is associated with a poor prognosis promotes proliferation, is a downstream target of MAPK/ETS-1 signalling and increases E2F7[104]
PVT1upregulatedhigh expression is associated with an advanced clinical stage and lymph node metastasis (LNM)promotes cell adhesion, viability, migration and invasion; enhances EMT via TGF-β/SMAD signalling; and downregulates SMAD4 and p21[115,122]
LINC00909upregulatedhigh expression is associated with poor OS and DFS and correlates with an advanced TNM stage, a larger tumour size, poor differentiation and LNMpromotes cell viability, colony formation, migration and xenograft tumour growth and metastasis; inhibits apoptosis; upregulates the expression of pluripotency factors; promotes the pancreatic cancer stem cell phenotype; activates the MAPK/JNK pathway; and destabilizes the SMAD4 mRNA[116]
BC037916upregulatedincreased expression correlates with a clinical stage and advanced T and N stages and is associated with poor OSpromotes proliferation, invasion and xenograft tumour growth; reduces apoptosis; positively regulates EMT via SMAD2/3 signalling; and upregulates JAK/STAT signalling[117]
LINC00462upregulatedexpression is associated with a larger tumour size, poor tumour differentiation, a TNM stage and distant metastasispromotes proliferation, migration, invasion, xenograft growth and invasion; reduces apoptosis and cellular adhesion; induces EMT through the activation of the TGF-β pathway via the upregulation of TGFBR1/2; and acts as a ceRNA to sequester miR-665[118]
LINC-PINTdownregulatedLow plasma levels correlate with tumour recurrence and are associated with tumour size; a low LINC-PINT level in tumour tissues correlates with a poor prognosis after pancreatectomyreduces cell proliferation and increases TGF-β1 expression in a pancreatic cancer cell line [119,120]
LINC00261downregulatedexpression is subtype-dependent and shows an inverse correlation with tumour grade and stage, and high expression is associated with better OSinhibits cell migration and invasion; TGF-β stimulation decreases LINC00261 levels, and its genetic ablation leads to the downregulation of the E-cadherin mRNA and protein and the induction of an EMT programme[121]
Table 3. RNA-binding proteins and their roles in pancreatic cancer.
Table 3. RNA-binding proteins and their roles in pancreatic cancer.
RBPRoles in PDACReferences
ELAVL1 (HuR)upregulated in PDAC; associated with a poor prognosis; promotes proliferation, the cell cycle, migration and invasion; inhibits apoptosis; and might mediate gemcitabine
sensitivity induced by promoting DCK expression; stabilized mRNAs in PDAC:
BARD1, DCK, DR5/TNFRSF10B, GPRC5A, IDH1, PRPS2, PDGFA, SNAI1, WEE1 and YAP1
[128,131,132,133,134,135,136,137,138,139,140,141,182]
IGF2BP1upregulated in PDAC; associated with a poor prognosis; promotes proliferation, cell cycle progression, EMT, metastasis and gemcitabine resistance; is an m6A reader; and
stabilized RNAs in PDAC: SH3BP5-AS, E2F1 and CDC25A
[142,143,144,146,147,148,149,150,183]
IGF2BP2upregulated in PDAC; associated with a poor prognosis; promotes proliferation and glucose metabolism; associated with an immune-suppressive microenvironment; is an m6A reader; and stabilized mRNAs in PDAC: SLC2A1 and CD274[142,143,154,155,156,157,158,159,183]
IGF2BP3upregulated in PDAC; associated with a poor prognosis; frequently used as a biomarker (needle biopsy); promotes proliferation, adhesion, migration and invasion; associated with an immune suppressive microenvironment; is an m6A reader; and
stabilized mRNAs in PDAC: ARF6, ARHGEF4, CD44, SMS and UBE2K
[142,143,157,161,162,164,165,166,167,168,169,170,171,172,173,174,183]
BICC1upregulated in PDAC; expression associated with EMT and immune infiltration; and supports angiogenesis and chemoresistance by promoting LCN2 and CXCL1
expression
[176,177,178,179,180,181]
CELF2 (CUGBP2)enhances stability but represses the translation of VEGF and COX2 mRNAs and
induced by curcumin
[184]
CSDE1 (UNR)biomarker associated with a favourable prognosis and the immunogenic subtype[185]
FUSupregulated in PDAC, stabilizes the NRF2 mRNA and suppresses oxidative stress and ferroptosis[186]
hnRNPA2/B1upregulated in early PDAC stages; inhibits apoptosis by suppressing BCL-X(S) splicing; regulates splicing of ARAF; and interacts with KRAS[93,102,187,188]
hnRNPCcontrols the mRNA stability of IQGAP3 to promote cell proliferation, migration,
invasion and metastasis
[189]
hnRNPFpart of a druggable super-enhancer network that promotes PDAC cell and tumour growth and stabilizes the PRMT1 mRNA to promote MYC expression[190]
hnRNPLupregulated in PDAC, associated with a poor prognosis and promotes migration and EMT[191]
LIN28Bupregulated in PDAC, associated with metastasis and a poor prognosis; promotes proliferation, stemness, migration and EMT;
targets in PDAC (via de-regulating let-7): MYC, HMGA2, KRAS, TET3, WNT5A and PCSK9
[192,193,194]
MEX3Aupregulated in PDAC, expression correlates with disease progression in human and mice; promotes cell cycle and gemcitabine resistance and
stabilized mRNAs in PDAC: CDK6
[195]
MSI2upregulated in PDAC; associated with metastasis and a poor prognosis; promotes proliferation, metastasis and chemoresistance; transcriptionally suppressed by KLF4; and controls HIPPO signalling by suppressing SAV1 and MOB1 translation; mRNA targets in PDAC: NUMB, MOB1, SAV1[196,197,198,199]
QKIupregulated in PDAC tumour cells and fibroblasts and promotes proliferation, EMT and metastasis[200]
RBFOX2prevents PDAC progression and metastasis and controls the splicing of ABI1 mRNA to
inhibit migration
[201]
RBM10downregulated in PDAC; the RBM10 mutation in advanced-stage tumours is associated with favourable survival; regulates the splicing of hTERT[202,203]
STRBPBinds to the HSATII RNA to prevent EMT and invasion and controls isoform switching of CLSTN1[204]
ZFRupregulated in PDAC and promotes proliferation, cell cycle progression and invasion[205]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Preckwinkel, P.; Mir, K.U.I.; Otto, F.W.; Elrewany, H.; Sinz, A.; Hüttelmaier, S.; Bley, N.; Gutschner, T. Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression. Cancers 2025, 17, 1601. https://doi.org/10.3390/cancers17101601

AMA Style

Preckwinkel P, Mir KUI, Otto FW, Elrewany H, Sinz A, Hüttelmaier S, Bley N, Gutschner T. Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression. Cancers. 2025; 17(10):1601. https://doi.org/10.3390/cancers17101601

Chicago/Turabian Style

Preckwinkel, Pit, Khursheed Ul Islam Mir, Florian W. Otto, Hend Elrewany, Andrea Sinz, Stefan Hüttelmaier, Nadine Bley, and Tony Gutschner. 2025. "Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression" Cancers 17, no. 10: 1601. https://doi.org/10.3390/cancers17101601

APA Style

Preckwinkel, P., Mir, K. U. I., Otto, F. W., Elrewany, H., Sinz, A., Hüttelmaier, S., Bley, N., & Gutschner, T. (2025). Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression. Cancers, 17(10), 1601. https://doi.org/10.3390/cancers17101601

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