Next Article in Journal
Addictions, Social Deprivation and Cessation Failure in Head and Neck Squamous Cell Carcinoma Survivors
Next Article in Special Issue
Urokinase-Type Plasminogen Activator Receptor (uPAR) Cooperates with Mutated KRAS in Regulating Cellular Plasticity and Gemcitabine Response in Pancreatic Adenocarcinomas
Previous Article in Journal
Anatomic Region of Cutaneous Melanoma Impacts Survival and Clinical Outcomes: A Population-Based Analysis
Previous Article in Special Issue
Development of a Clinical–Biological Model to Assess Tumor Progression in Metastatic Pancreatic Cancer: Post Hoc Analysis of the PRODIGE4/ACCORD11 Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

microRNAs Associated with Gemcitabine Resistance via EMT, TME, and Drug Metabolism in Pancreatic Cancer

Department of Hepatobiliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(4), 1230; https://doi.org/10.3390/cancers15041230
Submission received: 12 January 2023 / Revised: 13 February 2023 / Accepted: 13 February 2023 / Published: 15 February 2023
(This article belongs to the Special Issue Drug Resistance in Gastrointestinal Cancer)

Abstract

:

Simple Summary

We herein reviewed the current evidence for the role of microRNAs (miRNAs) in the mechanism of chemoresistance in pancreatic cancer. Pancreatic cancer has an extremely poor prognosis due to its late discovery, aggressive nature, and chemoresistance. Recent accumulated reports proved that aberrant miRNAs could induce chemoresistance in pancreatic cancer. However, the exact underlying molecular mechanisms remain poorly understood. In this review, we discuss recently available and novel knowledge about overcoming chemoresistance in pancreatic cancer.

Abstract

Despite extensive research, pancreatic cancer remains a lethal disease with an extremely poor prognosis. The difficulty in early detection and chemoresistance to therapeutic agents are major clinical concerns. To improve prognosis, novel biomarkers, and therapeutic strategies for chemoresistance are urgently needed. microRNAs (miRNAs) play important roles in the development, progression, and metastasis of several cancers. During the last few decades, the association between pancreatic cancer and miRNAs has been extensively elucidated, with several miRNAs found to be correlated with patient prognosis. Moreover, recent evidence has revealed that miRNAs are intimately involved in gemcitabine sensitivity and resistance through epithelial-to-mesenchymal transition, the tumor microenvironment, and drug metabolism. Gemcitabine is the gold standard drug for pancreatic cancer treatment, but gemcitabine resistance develops easily after chemotherapy initiation. Therefore, in this review, we summarize the gemcitabine resistance mechanisms associated with aberrantly expressed miRNAs in pancreatic cancer, especially focusing on the mechanisms associated with epithelial-to-mesenchymal transition, the tumor microenvironment, and metabolism. This novel evidence of gemcitabine resistance will drive further research to elucidate the mechanisms of chemoresistance and improve patient outcomes.

Graphical Abstract

1. Introduction

Pancreatic cancer remains a lethal disease and is the third leading cause of cancer-related deaths in the United States, with an incidence of 62,210 new cases, and an exceptionally high mortality rate of 49,830 deaths (80.1%) in 2022 [1]. Pancreatic cancer is expected to become the second leading cause of cancer-related deaths in the future [2]. Despite accumulated knowledge regarding pancreatic cancer etiology, patient prognosis has not significantly improved over the last decade [3]. The high mortality rate is rooted in the lack of specific symptoms, diagnostic tools, and effective chemotherapeutics, along with the increasing incidence of chemoresistance. Among these, chemoresistance is a well-known factor that contributes to a poor prognosis.
Compared with 5-fluorouracil, gemcitabine was found to be superior in relieving symptoms caused by progression in patients with pancreatic cancer [4]. This made gemcitabine the key drug for pancreatic cancer in combination with other chemotherapeutic agents such as nab-paclitaxel [5] or oxaliplatin [6]. However, the clinical benefits of these regimens are limited by chemoresistance. Thus, there is an urgent and crucial need to establish strategies for overcoming gemcitabine resistance to improve pancreatic cancer treatment. To date, the most clinically reliable marker for prognosis is CA19-9) (carbohydrate antigen 19-9 in pancreatic cancer [7]. Several other markers have been reported; however, they have not seen clinical use [8,9,10].
microRNAs (miRNAs) regulate gene expression by binding to the 3′-UTR (prime untranslated region) of their target mRNAs and inhibiting the production of those proteins. The association between miRNAs and chemoresistance has been reported in various types of cancer [11,12,13]. Thus, miRNAs are attractive molecules that retain the potential to overcome gemcitabine resistance. However, the scientific mechanism underlying this association remains unresolved despite extensive study. Recent evidence has shown that epithelial-to-mesenchymal transition (EMT) [14], tumor microenvironment (TME) [15,16], and drug metabolism [17] are major causes of the development of gemcitabine resistance via aberrantly expressed miRNAs. Therefore, it is important to identify innovative biomarkers and novel targets for intrinsic and acquired gemcitabine resistance in patients with pancreatic cancer, since CA19-9 is not a biomarker for gemcitabine resistance, though it is known to be a prognostic marker with high sensitivity and specificity (both 80%) [7]. EMT is a process that changes epithelial cells into mesenchymal-like cells, leading to the acquisition of cellular properties for invasiveness, proliferation, chemoresistance, and higher metastatic potential. Evidence has shown that expression of EMT-related genes such as those encoding vimentin, Snai1, and Zeb1 (zinc finger E-box binding homeobox 1) could further increase the risk of cancer cells acquiring a high-grade phenotype [18,19].
The TME is composed of interstitial tissues surrounding the pancreatic cancer cells and comprises immune cells, pancreatic stellate cells, stromal cells, blood vessels, fibroblasts, and the extracellular matrix (ECM). The TME can induce hypoxia, which confers several metabolic advantages for cancer cell protection and survival [20,21]. Recent review articles have shown that TME plays a pivotal role in the development of pancreatic cancer cells and gemcitabine resistance [22,23].
Finally, aberrant gemcitabine metabolism, including gemcitabine-related transporters and enzymes, is a cause of gemcitabine resistance [24,25,26,27]. Dysregulation of these transporters or enzymes results in gemcitabine resistance through complex, irregular factors and signaling pathways [28].
Based on these reports, the purpose of this review was to summarize the current evidence on the roles of miRNAs as potential biomarkers and therapeutic targets for gemcitabine resistance via EMT, TME, and drug metabolism in pancreatic cancer. Here, we systematically summarized recent published reports, including review articles, and focused on gemcitabine resistance in pancreatic cancer. A systematic search was performed using PubMed to identify original research studies mainly published in the last 20 years using the search terms pancreatic cancer and gemcitabine resistance. Finally, 1357 articles were chosen. Moreover, we targeted studies that satisfied the following criteria: (1) studied miRNA expression due to gemcitabine resistance in pancreatic cancer, (2) used human samples, and (3) were written in English. The full texts of the articles of interest were then evaluated. In this review, we focused on the mechanisms of gemcitabine resistance via EMT, TME, and gemcitabine metabolism.

2. Diverse Functions of miRNAs

miRNAs are short noncoding RNAs containing approximately 22 nucleotides, which can bind directly to specific targets within the 3′-untranslated regions of mRNAs [29,30]. In addition, miRNAs inhibit translation or enhance mRNA cleavage at the post-transcriptional level [31]. miRNAs are key regulators in the development, differentiation, and apoptosis of normal cells and may affect tumorigenesis and metastatic potential [32]. Remarkably, miRNAs exhibit tissue-specific and disease-specific expression, which suggests their potential as novel diagnostic and prognostic markers as well as therapeutic targets [33]. In addition, each miRNA has several different target genes, which results in the inhibition of the target genes for signaling pathways; therefore, miRNAs can function as both oncogenes and tumor suppressor genes [34]. Recent data have shown that frequently deregulated miRNAs are associated with detection, progression, and chemoresistance in pancreatic cancer [35,36]. Furthermore, several studies have documented that miRNAs influence gemcitabine resistance via EMT, TME, and drug metabolism [37,38,39].

3. Gemcitabine Resistance in Pancreatic Cancer

Gemcitabine remains the gold standard for the treatment of pancreatic cancer. Clinical studies, including combination treatment with gemcitabine, have been extensively performed; however, combination regimens were found to be insufficient compared with traditional regimens, such as gemcitabine plus nab-paclitaxel or erlotinib, or S-1 combination [40,41,42,43]. The major limitations in treatment effects include gemcitabine resistance and considerable adverse events [28]. An understanding of the mechanisms underlying gemcitabine resistance may lead to the discovery of promising targeted therapies. Numerous studies have reported the major molecular factors of gemcitabine resistance, such as: (1) cellular factors (cell signaling pathways, cancer stem cells, and EMT); (2) environmental factors (oxygen supply, stroma, ECM, and blood supply); and (3) drug metabolism (transporter, drug activation, and enzymes) in pancreatic cancer [22,44,45,46]. Moreover, these factors can result from molecular and cellular changes, including aberrant expression of mRNAs or miRNAs, and can interact in a complex manner [47]. It is therefore challenging to overcome gemcitabine resistance given the several different factors involved. Herein, we summarize the reported miRNA-mediated mechanisms that directly contribute to gemcitabine sensitivity and resistance in pancreatic cancer. Among them, we focus on EMT, TME, and metabolism-related mechanisms via miRNAs.

4. EMT-Mediated Mechanisms of Gemcitabine Resistance in Pancreatic Cancer

4.1. EMT and Gemcitabine Resistance

EMT is associated with gemcitabine resistance in pancreatic cancer [48,49]. It is characterized by the evolution of an epithelial phenotype into a mesenchymal phenotype, which can lead to cell proliferation, invasiveness, and metastasis [50,51]. This fundamental process is accompanied by morphological changes in the cancer cells. EMT is mediated by a variety of key genes and cellular signaling pathways; consequently, EMT results in higher proliferation, invasion, and chemoresistance. Molecular markers of EMT include increased expression of vimentin, Twist, Snail, Slug, and ZEB1 [52,53]. In contrast, mesenchymal-epithelial transition (MET) markers include Zo-1 and E-cadherin. Signaling pathways, such as the Notch and NFκB pathways, are critical for the induction of EMT [50]. Several studies have reported that gemcitabine-resistant cells show upregulated vimentin and downregulated E-cadherin expression associated with the activation of NFkB and c-MET tyrosine kinase, respectively [54,55,56]. Additionally, gemcitabine-resistant cells express higher SMAD2 or cancer stem cell (CSC) markers with EMT characteristics [57,58], because CSCs are evidently linked to the EMT process, which has been associated with gemcitabine resistance [59]. miR-34a recovers sensitivity to gemcitabine by inhibiting Notch 1, which is located upstream of the EMT pathways [60]. EMT can directly induce the CSC phenotype in pancreatic cancer. In contrast, gemcitabine induces EMT and CSC molecular marker expression [61]. Thus, EMT features overlap with molecular and morphological changes in CSC. In contrast, miR-17 is reduced in gemcitabine-resistant CSC by targeting the TGF-β1 signaling pathway and inhibiting the downstream targets p21, p57, and TBX3 [62]. Moreover, Ji et al. revealed that miR-34 may be involved in pancreatic cancer stem cell self-renewal, potentially via the direct modulation of downstream targets Bcl-2 and Notch [63]. In this review, we considered CSCs to be consistent with the EMT phenomenon. Conversely, inhibition of Notch signaling in gemcitabine-resistant cells could induce the MET phenotype from the EMT phenotype, indicating that gemcitabine resistance is reversible and associated with decreased EMT marker expression [58]. Hypoxia can also induce PLOD2-influenced gemcitabine resistance through EMT [64]. Moreover, ZEB1 can also mediate gemcitabine resistance and reduce E-cadherin expression. In contrast, reduced ZEB1 levels can restore gemcitabine sensitivity, indicating that ZEB1 is responsible for gemcitabine resistance [65]. Carrasco-Garcia et al. also showed that SOX9, which participates in the initiation of pancreatic cancer, correlated with EMT with high vimentin and low E-cadherin expression [66]. Recent evidence has shown that signaling pathways, including tumor necrosis factor (TNF) and hypoxia-inducible factor-1 (HIF-1), are associated with EMT in pancreatic cancer [66,67]. Moreover, Zhang et al. showed that DPEP1 induction enhanced gemcitabine sensitivity through the RAS-RAF-MEK-ERK and PI3K pathways [68]. EMT is also associated with tumor budding, which accounts for gemcitabine resistance; a study indicated that tumor budding with vimentin expression becomes a key process in pancreatic cancer and is responsible for progression and gemcitabine resistance [69]. Taken together, EMT induction is strongly correlated with the development of gemcitabine resistance in pancreatic cancer.

4.2. miRNAs Related to Gemcitabine and EMT

4.2.1. miRNAs Associated with Gemcitabine Sensitivity

With regard to the association between gemcitabine sensitivity and miRNAs mediating EMT, several tumor suppressor miRNAs have been identified to regulate EMT-related genes and improve gemcitabine sensitivity in pancreatic cancer [70,71]. (See Figure 1 and Table 1). Wang et al. showed that miR-30a reverses gemcitabine resistance in pancreatic cancer by targeting the Snail-AKT signaling pathway [72]. Funamizu et al. revealed that miR-200b could restore gemcitabine sensitivity by inhibiting ZEB1, thereby upregulating E-cadherin [73]. In addition, Li et al. proved that naturally occurring agent-induced miR-200 and let-7 expression could reverse MET from the EMT phenotype in gemcitabine-resistant cells [74]. Liu et al. reported that miR-125a-3p can restore gemcitabine sensitivity and inhibit EMT through targeting Fyn [75]. Fu et al. also revealed that NEAT1, mediated by miR-506, could control ZEB2 expression [76]. Additionally, Li et al. showed that miR-506 suppressed sphingosine kinase 1, which was significantly associated with poor survival in a large cohort [77]. Hiramoto et al. demonstrated that miR-509 and miR-1243 improve gemcitabine sensitivity via E-cadherin expression [78]. Furthermore, Yang et al. reported that miR-3656 plays a significant role in gemcitabine sensitivity by inhibiting vimentin and Twist expression [79]. Two reports have shown that miR-153 can enhance gemcitabine sensitivity by inhibiting Snail [80,81]. Wang et al. revealed that upregulation of miR-183 and miR-200b improved gemcitabine sensitivity via ZEB1 inhibition caused by KLF4 [82]. Chaudhary et al. also reported that miR-205 impairs gemcitabine resistance by inhibiting the EMT phenotype [83]. More recently, Yang et al. revealed that exosomal miR-210 in CSCs mediates gemcitabine resistance by activating the PI3K/Akt/mTOR pathway [84].

4.2.2. miRNAs Associated with Gemcitabine Resistance

The reported miRNAs associated with gemcitabine resistance in pancreatic cancer have been summarized. Hasegawa et al. revealed that miR-1246 contributes to gemcitabine resistance and induces CSC-like properties through CCNG2 [85]. Xiong et al. demonstrated that miR-10a contributes to gemcitabine resistance by targeting TFAP2C, thereby resulting in increased Snail 1 expression and EMT induction [86]. Zhang et al. showed that miR-15b degrades SMURF2 and promotes TGF-β-mediated EMT in pancreatic cancer [87]. In addition, Yang and Funamizu et al. demonstrated that miR-301b induced EMT and enhanced gemcitabine resistance by reducing E-cadherin expression [54,88]. Zhang et al. also showed that activation of the miR-301/TP63 axis caused by hypoxia-induced EMT contributes to gemcitabine resistance [89]. Okazaki et al. suggested that miR-296-5p induces EMT and gemcitabine resistance [90]. Meanwhile, Ma et al. demonstrated that miR-223 enhances gemcitabine resistance by targeting FBXW7 through activating Notch signaling-mediated EMT [91]. Yu et al. revealed that miR-1206 enhances gemcitabine resistance through ESRP1 inhibition [92]. Furthermore, Yu et al. showed that miR-497 could inhibit gemcitabine resistance in CSCs by targeting NFκB [93]. Ma et al. indicated that miR-200-3p attenuates gemcitabine resistance in CSCs by modulating EMT and stemness [94].
Table 1. miRNAs for EMT.
Table 1. miRNAs for EMT.
AuthorRef. NumbermiRNATarget Gene
Sensitivity
Li Y[74]let-7NA
Cioffi M[62]miR-17NODAL
Wang T[72]miR-30aSNAI1
Ji Q[63]miR-34Bcl-2
Liu G[75]miR-125aFyn
Liu F[80]miR-153Snail
Bai Z[81]miR-153Snail
Wang Z[82]miR-183ZEB1
Li Y[74]miR-200NA
Funamizu N[55]miR-200bZEB1
Wang Z[82]miR-200bZEB1
Ma C[94]miR-200cNA
Chaudhary AK[83]miR-205TUBB3
Yu Q[93]miR-497NFKB1
Fu X[76]miR-506NEAT1
Hiramoto H[78]miR-509NA
Yu S[92]miR-1206ESRP1
Hiramoto H[78]miR-1243NA
Yang RM[79]miR-3656EMT related genes
Resistance
Xiong G[86]miR-10aTFAP2C
Zhang WL[87]miR-15bSMURF2
Yang Z[84]miR-210Rapamycin
Ma J[91]miR-223FBXW7
Okazaki J[90]miR-296BOK
Zhang KD[89]miR-301TP63
Funamizu N[54]miR-301bTP63
Yang S[88]miR-301bNR3C2
Hasegawa S[85]miR-1246CyclinG2
Ref: reference, NA: not applicable.
Despite this evidence, the role of miRNAs in gemcitabine sensitivity and resistance remains controversial because the complex and interacting networks underlying the phenomenon are challenging to elucidate. However, accumulating evidence implicates that aberrant expression of miRNAs modulates the responsiveness of gemcitabine sensitization.

5. TME-Mediated Mechanisms of Gemcitabine Resistance in Pancreatic Cancer

5.1. TME and Gemcitabine Resistance in Pancreatic Cancer

Pancreatic cancer has unique features to survive therapeutic strategies, characterized by the presence of an extensive desmoplastic stroma composed of ECM, cancer-associated fibroblasts (CAFs), pancreatic stellate cells (PSCs), inflammatory cells, immune cells (including tumor-associated macrophages [TAMs]), and other cell types (such as endothelial cells). The dense stroma facilitates the compression of blood vessels and leads to a hypoxic environment, which reduces the supply of chemotherapeutic agents and supports cancer progression [95]. Under these circumstances, the TME can induce immunosuppression to escape the immune system. The desmoplastic stroma and hypoxic environment have also been reported to promote EMT and cause gemcitabine resistance [96,97]. Accumulated evidence has revealed that desmoplastic stroma simply does not develop a physical barrier to gemcitabine; moreover, the respective components in the TME act to resist gemcitabine [98,99,100,101].

5.2. Role of ECM in Gemcitabine Resistance

ECM is defined as the physical support and material that fills the extracellular space and functions as a scaffold for cell adhesion, including fibronectin, collagen, and proteoglycan. ECM components, including collagen, fibronectin, and laminin, are secreted by PSCs and CAFs [95]. Recent evidence has shown that the ECM functions as a supporting tissue, regulating cancer cell proliferation and EMT [102]. Fibronectin is a major component of the ECM that Miyamoto et al. have demonstrated to be a contributor to gemcitabine resistance [103]. In addition, fibronectin plays a key role in gemcitabine resistance by activating the ERK1/2 pathway [104]. Topalovski et al. indicated that cooperation between TGF-β and fibronectin may establish coordinated EMT induction [105]. Furthermore, fibronectin can support cancer progression and reduce gemcitabine response [105]. In contrast, Dangi-Garimella et al. demonstrated that membrane type 1 matrix metalloproteinase (MMP) contributes to gemcitabine resistance and suggested that targeting MMP could be a novel approach to improving gemcitabine sensitivity [106]. Thus, ECM-targeted agents combined with gemcitabine chemotherapy are promising strategies for overcoming gemcitabine resistance. However, two clinical studies involving anti-ECM inhibitors did not show significant efficacy of these drugs [107,108].

5.3. Role of CAFs in Gemcitabine Resistance

CAFs constitute a major part of the tumor mass in pancreatic cancer. CAFs can enhance chemoresistance through ECM remodeling and immunological reprogramming [109]. Richards et al. recently reported that CAFs have the potential for gemcitabine resistance. They also showed that exosomes produced by CAFs promote EMT and gemcitabine resistance by inducing Snail [110]. In addition, Zhang et al. showed that CAFs activate NFκB and IL1 receptor-associated kinase 4, which results in enhanced tumor fibrosis, cell proliferation, and gemcitabine resistance [111]. Recent reports have revealed that CAFs promote gemcitabine resistance via the LIF/STAT3 or the TGF-β1/SMAD2/3 pathway [112,113], suggesting that targeting these pathways may be a novel strategy to reverse gemcitabine resistance. Thus, therapeutic agents controlling CAF function may play a vital role in sensitizing gemcitabine.

5.4. Role of PSCs in Gemcitabine Resistance

PSCs provide a solid foundation for the production of collagenous stroma for cancer development and survival [114,115]. Unfortunately, the role of PSCs in gemcitabine resistance has not yet been fully elucidated. However, recent evidence suggests that PSCs act as drivers of gemcitabine resistance and cancer progression. Cao et al. showed that the Notch pathway activated by PSCs promotes gemcitabine resistance and induces EMT [116]. Interestingly, PSCs have no tolerance to glucose adjustment; therefore, an increased number of PSCs in the pancreas allows the development of type 2 diabetes. Moreover, EMT-mediated higher glucose levels promote malignant potential [117]. It is well known that PSCs promote EMT in pancreatic cancer cells [118]. These reports suggest that targeting the Notch pathway may be an effective strategy for recovering gemcitabine tolerance.

5.5. Role of TAMs in Gemcitabine Resistance

TAMs are also abundantly present in the TME. TAMs innately play a role in the phagocytosis of apoptotic cells and affect cancer progression and gemcitabine resistance by secreting numerous factors, such as growth factors, proteolytic enzymes, and inflammatory cytokines [119,120]. Weiseman et al. revealed that TAMs contribute to gemcitabine resistance by reducing apoptosis and upregulating cytidine deaminase (CDA) expression. These effects augment the response to gemcitabine by activating caspase-3 [121]. Moreover, Guo et al. showed that miR-222, delivered by TAM, suppresses TSC1 and activates the PI3K/AKT/mTOR pathway, which results in the development of gemcitabine-resistant cancer cells [122]. Additionally, Nagathihalli et al. showed that a STAT3 pathway inhibitor combined with gemcitabine can enhance gemcitabine delivery and response by remodeling the tumor stroma [123]. In contrast, TAMs directly activate the STAT3 pathway to regulate CSCs [124]. Thus, STAT3-targeted therapy with gemcitabine may be a promising therapeutic strategy for pancreatic cancer.

5.6. miRNAs Involved in TME-Mediated Gemcitabine Resistance

Hypoxia is an essential component of the TME for the survival of cancer cells in pancreatic cancer [125,126,127]. Extensive studies have revealed that miRNAs are strongly associated with TME function [128,129,130]. (Figure 2 and Table 2). Luo et al. revealed that miR-301a plays a critical role in gemcitabine resistance via the TME [131]. Liu et al. also showed that PVT1 and HIF-1 inhibition, mediated by miR-143, improves gemcitabine sensitivity [132]. Xin et al. proposed that nano-medically modified gemcitabine and miR-519c are effective therapeutic targets to treat desmoplasia and hypoxia-induced gemcitabine resistance in the TME [133]. In contrast, HIF-1 expression is a well-known inducible factor for gemcitabine resistance and EMT [134,135]. As such, HIF-1-targeted miRNA strategies have been described in the literature. Liu et al. showed that miR-3662 inhibits gemcitabine resistance by inhibiting HIF-1 in the TME [136]. In addition, Ni et al. showed that miR-210 controls HOXA9 expression and upregulates the HIF-1/NF-κB pathway, which promotes EMT and hypoxia [137].
A recent report showed that miR-21 affects CAFs accumulation, and subsequently, CAFs release miR-21 that can induce gemcitabine resistance by downregulating PTEN [111,138,139]. Notably, hypoxia induces miR-21 expression [115]. Moreover, tumor associated fibrosis produces miR-21 that targets PTEN [140]. miR-21 is a powerful oncogenic gene that enhances gemcitabine resistance by targeting PTEN or FasL [141]. In contrast, Park et al. reported that miR-21 inhibition or miR-221 induction enhanced gemcitabine sensitivity in pancreatic cancer due to increased PTEN expression [142]. Fang et al. showed that CAF-derived exosomal miR-106b plays a significant role in gemcitabine resistance [143]. Thus, miR-21-mediated CAF-targeted therapy may be a promising strategy to overcome chemoresistance in TME.
Table 2. miRNAs for TME.
Table 2. miRNAs for TME.
AuthorRef. NumbermiRNATarget Gene
Sensitivity
Liu YF[132]miR-143HIF-1
Ni J[137]miR-210HOXA9
Liu A[136]miR-3662HIF-1
Resistance
Zhang L[138]miR-21PDCD4
Kadera BE[139]miR-21NA
Fang Y[143]miR-106bTP53INP1
Masamune A[144]miR-221NA
Guo Y[122]miR-222TSC1
Luo G[131]miR-301aTP63
Xin X[133]miR-519cHIF-1
Ref: reference, NA: not applicable.
Finally, increased miR-221 expression in PSCs contributes to gemcitabine resistance by activating the MAPK signaling pathway [144]. These findings suggest the importance of miRNAs in the regulation of TME components and their potential role as novel targets for improving gemcitabine efficacy in pancreatic cancer.

6. Gemcitabine Metabolism-Mediated Mechanisms of Gemcitabine Resistance in Pancreatic Cancer

6.1. Gemcitabine Metabolism

Previously, numerous investigations regarding the transport- and metabolism-related genes for gemcitabine were performed to elucidate the mechanism of gemcitabine resistance in pancreatic cancer cells [145,146,147]. Gemcitabine (dFdC) is a deoxycytidine nucleoside analog. Its metabolic pathways are shown in Figure 3. Gemcitabine is transported into the cells by nucleoside transporters, including concentrative nucleoside transporters (hCNTs) and equilibrative nucleoside transporters (hENT1 and hENT2). hENT1 is the main transporter for gemcitabine uptake in cancer cells. Gemcitabine is a prodrug that requires intracellular phosphorylation for its activation. dFdC is first phosphorylated to dFdCMP by deoxycytidine kinase (dCK), a key enzyme in the rate-limiting process. Subsequently, double phosphorylation results in the formation of the triphosphate form (dFdCTP). Finally, dFdCTP is incorporated into the DNA chain to produce a therapeutic effect. However, most dFdCs are inactivated by CDA. Additionally, dFdCDP and dFdCTP can inhibit ribonucleotide reductase (RR), which is responsible for converting ribonucleosides to deoxyribonucleoside triphosphates (dNTPs). Moreover, the dCTP produced by RR acts as a competitive inhibitor of dFdCTP. Therefore, overt changes in gemcitabine metabolism may generate gemcitabine resistance [148,149,150,151,152,153].

6.2. Relation of Gemcitabine Metabolism to Gene Expression in Pancreatic Cancer

6.2.1. Transporters Associated with Gemcitabine Resistance

Alterations in hENT1, the major transporter for gemcitabine [153], contribute to the development of gemcitabine resistance [154]. Tsesmetzis et al. summarized that nucleoside analogs are principally transported by two membrane transporter families: hCNT1-3 and hENT1-4. Nucleoside analogs are transported by both hENTs and hCNTs, whereas nucleoside analogs are excreted by multidrug resistance proteins (MRP), also called ATP-binding cassette transporters (ABC transporters), which are classified into seven different subtypes (from ABCA- to ABCG) based on their gene structure [155]. Giovannetti et al. reported an association between hENT1 and gemcitabine sensitivity [156]. Mackey et al. also reported that hENT1 deficiency causes significant gemcitabine resistance [157]. In addition, recent data have shown that highly expressed hENT1 is a molecular and mechanistically relevant predictive marker for gemcitabine sensitivity [158,159]. In contrast, Hung et al. revealed that hCNT1 deficiency affects gemcitabine resistance [150]. Moreover, Skrypek et al. showed that MUC4-induced hcNT1 inhibition enhances gemcitabine resistance [160]. Qiu et al. described that inhibition of the PI3K/NFκB pathway could induce ABC transporter reduction by improving gemcitabine resistance [161]. Hagmann et al. also showed that ABC transporters are associated with gemcitabine resistance [149]. Because of the key roles played by hNT and ABC transporters in gemcitabine resistance, modulation of these transporters may lead to better efficacy of gemcitabine-based chemotherapeutic strategies.

6.2.2. CDA-Induced Gemcitabine Resistance

CDA is a catabolic enzyme that transforms gemcitabine into an inactivated metabolite [162]. Several studies have shown that the upregulation of CDA confers gemcitabine resistance and gemcitabine-induced toxicities [24,163,164,165,166]. Moreover, CDA expression levels can be used as a predictive marker for gemcitabine resistance [167]. Therefore, upregulated CDA expression may play an important role in gemcitabine resistance and patient prognosis in pancreatic cancer.

6.2.3. Relation of dCK and Gemcitabine Resistance

Gemcitabine is phosphorylated by dCK, resulting in its active product. dCK is the rate-limiting enzyme for nucleoside analogs [168,169]. Therefore, dCK inhibition is considered one of the causes of the development of gemcitabine resistance [25,170]. Ohmime et al. suggested that dCK expression is a prognostic factor in patients with pancreatic cancer [171]. Kamada et al. also reported that transduction of dCK could recover gemcitabine sensitivity in pancreatic cancer [172].

6.2.4. Relation of RR to Gemcitabine Resistance

Another potential factor in the mechanism for gemcitabine resistance is the overexpression of RR. RR is composed of the regulatory subunit M1 and the catalytic subunit M2, which are responsible for the conversion of ribonucleosides to deoxyribonucleoside triphosphates [173]. Duxbury et al. found that upregulated RRM2 closely participates in gemcitabine resistance by inhibiting the NF-κB signaling pathway [174]. Furthermore, previous studies have shown that increased RR is involved in the enhanced resistance to gemcitabine [24,175]. In contrast, Nakahira et al. revealed that RRM1 inhibition significantly reduces gemcitabine resistance [27]. Moreover, several studies have revealed that increased RR expression serves as a prognostic marker for patients with bile duct and pancreatic cancers [176,177].

6.3. Relation of Gemcitabine Metabolism to miRNAs

Amponsah et al. recently clarified the impact of miRNAs on gemcitabine metabolism, including transporters and gemcitabine-related enzymes. This study showed that miR-210 is responsible for gemcitabine sensitivity by targeting the ABCC5 gene [178]. Gu et al. indicated that miR-3178 promotes chemoresistance to gemcitabine by upregulating the ABC transporter-mediated RhoB/PI3K/Akt pathway [179]. Although Wang et al. did not mention gemcitabine resistance, they reported an association between miR-520h and ABCG2 [180]. Additionally, miR-93 enhances ABCB1 expression via targeting PTEN and increases Akt phosphorylation [181]. Furthermore, miR-331 induces ABCB1 expression by activating the Wnt/β-catenin pathway through ST7L in pancreatic cancer cells [182]. To the best of our knowledge, there are no reports regarding the association of hENT and hCNT with miR-mediated gemcitabine resistance. Recent data have shown that miR-101-3p and miR-211 enhance gemcitabine sensitivity by inhibiting RRM1 and RRM2, respectively [17,183]. Bhutia et al. revealed that let-7 negatively regulates RRM2 and sensitizes to gemcitabine [184]. Moreover, Rajabpour et al. showed that miR-608 leads to increased gemcitabine sensitivity, with decreased RRM1 and CDA expression [185]. Lu et al. suggested that miR-20a-5p upregulates gemcitabine sensitivity by targeting RRM2 [186]. Patel et al. also recently demonstrated that suppression of miR-155 induced dCK levels and restored gemcitabine sensitivity [187]. This demonstrates that miRNAs are connected with numerous targets involved in resistance to gemcitabine metabolism (Figure 3 and Table 3).

7. Conclusions

We believe gemcitabine plus miRNA-based therapeutics can be expected to overcome the poor prognosis of pancreatic cancer, although further studies are needed to elucidate the molecular mechanism of chemoresistance caused by aberrantly expressed miRNAs. One of the major limitations of miRNA-based therapeutics is the lack of a delivery system capable of targeting tumors. Recent clinical trials of miR-34a liposomal-based therapy have been performed for advanced solid cancers [188]. Unfortunately, the trial was halted due to serious adverse events. However, miRNA-based strategies have the potential to deliver unprecedented value in pancreatic cancer treatment, as these may be able to control several target genes and signaling pathways. Moreover, recent studies showed that secreted microvesicles such as exosomes contain miRNAs, which play an important role in gemcitabine chemoresistance [110,189,190]. Comandatore et al. reviewed that exosomes, including oncogenic miRNAs, can affect gemcitabine resistance [191]. Thus, we suggest that miRNA-based strategies, involving regulation of miRNA expression, might contribute to overcoming gemcitabine resistance in the future. In contrast, exosomes, including specific miRNAs, must be a biomarker for gemcitabine resistance due to liquid biopsy.

Author Contributions

Conceptualization, N.F. and Y.T.; methodology, N.F.; validation, M.H., K.T. and K.O.; investigation, N.F.; resources, M.H.; data curation, K.T.; writing—original draft preparation, N.F.; writing—review and editing, Y.T.; visualization, K.S.; supervision, K.O.; project administration, K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Acknowledgments

The authors thank Noriko Funamizu (Department of Internal Medicine, Hirose Hospital, Ehime, Japan) for the invaluable advice and discussions regarding the manuscript.

Conflicts of Interest

The authors declare no conflict of interest for this article.

Abbreviations

Bcl-2B cell lymphoma-2
ZEB1zinc finger E-box binding homeobox 1
TUBB3tubulin-beta3
NFKB1nuclear factor kappa B subunit 1
NEAT1nuclear-enriched abundant transcript 1
ESRP1epithelial splicing regulatory protein 1
TFAP2Ctranscription factor activating protein 2 gamma
SMURF2SMAD specific E3 ubiquitin protein ligase 2
FBXW7F-Box and WD Repeat Domain Containing 7
BOKB-cell lymphoma 2 (BCL-2) ovarian killer
TP63tumor protein p63
NR3C2nuclear receptor subfamily 3 group C member 2
SHC1src homology domain-containing transforming protein 1
Baxbcl-2 associated X protein
RRMribonucleotide reductase regulatory subunit M
ABCCATP binding cassette C
CDAcytidine deaminase
PTENphosphatase and tensin homolog
dCKdeoxycytidine kinase
ST7Lsuppression of tumorigenicity 7-like
HIF-1hypoxia-inducible factor 1
HOXA9homeobox A9
PDCD4programmed cell death 4
TP53INP1tumor protein 53-induced nuclear protein 1
TSC1tuberous sclerosis complex 1

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA A Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [Green Version]
  3. Paulson, A.S.; Tran Cao, H.S.; Tempero, M.A.; Lowy, A.M. Therapeutic advances in pancreatic cancer. Gastroenterology 2013, 144, 1316–1326. [Google Scholar] [CrossRef]
  4. Burris, H.A.; Moore, M.J.; Andersen, J.; Green, M.R.; Rothenberg, M.L.; Modiano, M.R.; Cripps, M.C.; Portenoy, R.K.; Storniolo, A.M.; Tarassoff, P.; et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J. Clin. Oncol. 1997, 15, 2403–2413. [Google Scholar] [CrossRef] [Green Version]
  5. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
  6. Louvet, C.; Labianca, R.; Hammel, P.; Lledo, G.; Zampino, M.G.; André, T.; Zaniboni, A.; Ducreux, M.; Aitini, E.; Taïeb, J.; et al. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: Results of a GERCOR and GISCAD phase III trial. J. Clin. Oncol. 2005, 23, 3509–3516. [Google Scholar] [CrossRef]
  7. Barhli, A.; Cros, J.; Bartholin, L.; Neuzillet, C. Prognostic stratification of resected pancreatic ductal adenocarcinoma: Past, present, and future. Dig. Liver Dis. 2018, 50, 979–990. [Google Scholar] [CrossRef] [PubMed]
  8. Ohuchida, K.; Mizumoto, K.; Kayashima, T.; Fujita, H.; Moriyama, T.; Ohtsuka, T.; Ueda, J.; Nagai, E.; Hashizume, M.; Tanaka, M. MicroRNA expression as a predictive marker for gemcitabine response after surgical resection of pancreatic cancer. Ann. Surg. Oncol. 2011, 18, 2381–2387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Dhayat, S.; Mardin, W.A.; Mees, S.T.; Haier, J. Epigenetic markers for chemosensitivity and chemoresistance in pancreatic cancer—A review. Int. J. Cancer 2011, 129, 1031–1041. [Google Scholar] [CrossRef]
  10. Chiou, S.H.; Dorsch, M.; Kusch, E.; Naranjo, S.; Kozak, M.M.; Koong, A.C.; Winslow, M.M.; Grüner, B.M. Hmga2 is dispensable for pancreatic cancer development, metastasis, and therapy resistance. Sci. Rep. 2018, 8, 14008. [Google Scholar] [CrossRef]
  11. Mollah, F.; Varamini, P. Overcoming therapy resistance and relapse in TNBC: Emerging technologies to target breast cancer-associated fibroblasts. Biomedicines 2021, 9, 1921. [Google Scholar] [CrossRef] [PubMed]
  12. Campos, A.; Sharma, S.; Obermair, A.; Salomon, C. Extracellular vesicle-associated miRNAs and chemoresistance: A systematic review. Cancers 2021, 13, 4608. [Google Scholar] [CrossRef]
  13. Santos, P.; Almeida, F. Role of exosomal miRNAs and the tumor microenvironment in drug resistance. Cells 2020, 9, 1450. [Google Scholar] [CrossRef] [PubMed]
  14. Wellner, U.; Schubert, J.; Burk, U.C.; Schmalhofer, O.; Zhu, F.; Sonntag, A.; Waldvogel, B.; Vannier, C.; Darling, D.; zur Hausen, A.; et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat. Cell Biol. 2009, 11, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  15. Rice, A.J.; Cortes, E.; Lachowski, D.; Cheung, B.C.H.; Karim, S.A.; Morton, J.P.; Del Río Hernández, A. Matrix stiffness induces epithelial-mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis 2017, 6, e352. [Google Scholar] [CrossRef] [Green Version]
  16. Schoepp, M.; Ströse, A.J.; Haier, J. Dysregulation of miRNA expression in cancer associated fibroblasts (CAFs) and its consequences on the tumor microenvironment. Cancers 2017, 9, 54. [Google Scholar] [CrossRef] [Green Version]
  17. Maftouh, M.; Avan, A.; Funel, N.; Frampton, A.E.; Fiuji, H.; Pelliccioni, S.; Castellano, L.; Galla, V.; Peters, G.J.; Giovannetti, E. miR-211 modulates gemcitabine activity through downregulation of ribonucleotide reductase and inhibits the invasive behavior of pancreatic cancer cells. Nucleosides Nucleotides Nucleic Acids 2014, 33, 384–393. [Google Scholar] [CrossRef]
  18. Fazilaty, H.; Rago, L.; Kass Youssef, K.; Ocaña, O.H.; Garcia-Asencio, F.; Arcas, A.; Galceran, J.; Nieto, M.A. A gene regulatory network to control EMT programs in development and disease. Nat. Commun. 2019, 10, 5115. [Google Scholar] [CrossRef] [Green Version]
  19. Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.; Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [Google Scholar] [CrossRef]
  20. Lin, H.J.; Lin, J. Seed-in-soil: Pancreatic cancer influenced by tumor microenvironment. Cancers 2017, 9, 93. [Google Scholar] [CrossRef]
  21. Sunami, Y.; Böker, V.; Kleeff, J. Targeting and reprograming cancer-associated fibroblasts and the tumor microenvironment in pancreatic cancer. Cancers 2021, 13, 697. [Google Scholar] [CrossRef]
  22. Zeng, S.; Pöttler, M.; Lan, B.; Grützmann, R.; Pilarsky, C.; Yang, H. Chemoresistance in pancreatic cancer. Int. J. Mol. Sci. 2019, 20, 4504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chang, J.H.; Jiang, Y.; Pillarisetty, V.G. Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Medicine 2016, 95, e5541. [Google Scholar] [CrossRef] [PubMed]
  24. Funamizu, N.; Kamata, Y.; Misawa, T.; Uwagawa, T.; Lacy, C.R.; Yanaga, K.; Manome, Y. Hydroxyurea decreases gemcitabine resistance in pancreatic carcinoma cells with highly expressed ribonucleotide reductase. Pancreas 2012, 41, 107–113. [Google Scholar] [CrossRef] [PubMed]
  25. Funamizu, N.; Okamoto, A.; Kamata, Y.; Misawa, T.; Uwagawa, T.; Gocho, T.; Yanaga, K.; Manome, Y. Is the resistance of gemcitabine for pancreatic cancer settled only by overexpression of deoxycytidine kinase? Oncol. Rep. 2010, 23, 471–475. [Google Scholar] [CrossRef] [Green Version]
  26. Funamizu, N.; Lacy, C.R.; Fujita, K.; Furukawa, K.; Misawa, T.; Yanaga, K.; Manome, Y. Tetrahydrouridine inhibits cell proliferation through cell cycle regulation regardless of cytidine deaminase expression levels. PLoS ONE 2012, 7, e37424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Nakahira, S.; Nakamori, S.; Tsujie, M.; Takahashi, Y.; Okami, J.; Yoshioka, S.; Yamasaki, M.; Marubashi, S.; Takemasa, I.; Miyamoto, A.; et al. Involvement of ribonucleotide reductase M1 subunit overexpression in gemcitabine resistance of human pancreatic cancer. Int. J. Cancer. 2007, 120, 1355–1363. [Google Scholar] [CrossRef]
  28. Amrutkar, M.; Gladhaug, I.P. Pancreatic cancer chemoresistance to gemcitabine. Cancers 2017, 9, 157. [Google Scholar] [CrossRef] [Green Version]
  29. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  30. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  31. Bagga, S.; Bracht, J.; Hunter, S.; Massirer, K.; Holtz, J.; Eachus, R.; Pasquinelli, A.E. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell 2005, 122, 553–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Garofalo, M.; Romano, G.; Di Leva, G.; Nuovo, G.; Jeon, Y.J.; Ngankeu, A.; Sun, J.; Lovat, F.; Alder, H.; Condorelli, G.; et al. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat. Med. 2011, 18, 74–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Forterre, A.; Komuro, H.; Aminova, S.; Harada, M. A comprehensive review of cancer microRNA therapeutic delivery strategies. Cancers 2020, 12, 1852. [Google Scholar] [CrossRef] [PubMed]
  34. Khan, S.; Ansarullah, A.; Kumar, D.; Jaggi, M.; Chauhan, S.C. Targeting microRNAs in pancreatic cancer: Microplayers in the big game. Can. Res. 2013, 73, 6541–6547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Uddin, M.H.; Al-Hallak, M.N.; Philip, P.A.; Mohammad, R.M.; Viola, N.; Wagner, K.U.; Azmi, A.S. Exosomal microRNA in pancreatic cancer diagnosis, prognosis, and treatment: From bench to bedside. Cancers 2021, 13, 2777. [Google Scholar] [CrossRef] [PubMed]
  36. Kato, S.; Honda, K. Use of biomarkers and imaging for early detection of pancreatic cancer. Cancers 2020, 12, 1965. [Google Scholar] [CrossRef]
  37. Pavlíková, L.; Šereš, M.; Breier, A.; Sulová, Z. The roles of microRNAs in cancer multidrug resistance. Cancers 2022, 14, 1090. [Google Scholar] [CrossRef]
  38. Pan, G.; Liu, Y.; Shang, L.; Zhou, F.; Yang, S. EMT-associated microRNAs and their roles in cancer stemness and drug resistance. Cancer Commun. 2021, 41, 199–217. [Google Scholar] [CrossRef]
  39. Hwang, G.R.; Yuen, J.G.; Ju, J. Roles of microRNAs in gastrointestinal cancer stem cell resistance and therapeutic development. Int. J. Mol. Sci. 2021, 22, 1624. [Google Scholar] [CrossRef]
  40. Pusceddu, S.; Ghidini, M.; Torchio, M.; Corti, F.; Tomasello, G.; Niger, M.; Prinzi, N.; Nichetti, F.; Coinu, A.; Di Bartolomeo, M.; et al. Comparative effectiveness of gemcitabine plus nab-paclitaxel and FOLFIRINOX in the first-line setting of metastatic pancreatic cancer: A systematic review and meta-analysis. Cancers 2019, 11, 484. [Google Scholar] [CrossRef]
  41. Lee, Y.S.; Lee, J.C.; Kim, J.H.; Kim, J.; Hwang, J.H. Pharmacoethnicity of FOLFIRINOX versus gemcitabine plus nab-paclitaxel in metastatic pancreatic cancer: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 20152. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, Z.Y.; Yuan, J.Q.; Di, M.Y.; Zheng, D.Y.; Chen, J.Z.; Ding, H.; Wu, X.Y.; Huang, Y.F.; Mao, C.; Tang, J.L. Gemcitabine plus erlotinib for advanced pancreatic cancer: A systematic review with meta-analysis. PLoS ONE 2013, 8, e57528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Li, D.; Chen, C.; Zhou, Y.; Chen, R.; Fan, X.; Bi, Z.; Li, Z.; Liu, Y. Gemcitabine compared with gemcitabine and S-1 combination therapy in advanced pancreatic cancer: A systematic review and meta-analysis. Medicine 2015, 94, e1345. [Google Scholar] [CrossRef] [PubMed]
  44. Qin, C.; Yang, G.; Yang, J.; Ren, B.; Wang, H.; Chen, G.; Zhao, F.; You, L.; Wang, W.; Zhao, Y. Metabolism of pancreatic cancer: Paving the way to better anticancer strategies. Mol. Cancer 2020, 19, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Liang, C.; Shi, S.; Meng, Q.; Liang, D.; Ji, S.; Zhang, B.; Qin, Y.; Xu, J.; Ni, Q.; Yu, X. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: Where we are and where we are going. Exp. Mol. Med. 2017, 49, e406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Gaianigo, N.; Melisi, D.; Carbone, C. EMT and treatment resistance in pancreatic cancer. Cancers 2017, 9, 122. [Google Scholar] [CrossRef]
  47. Royam, M.M.; Ramesh, R.; Shanker, R.; Sabarimurugan, S.; Kumarasamy, C.; Gothandam, K.M.; Baxi, S.; Gupta, A.; Krishnan, S.; Jayaraj, R. miRNA predictors of pancreatic cancer chemotherapeutic response: A systematic review and meta-analysis. Cancers 2019, 11, 900. [Google Scholar] [CrossRef] [Green Version]
  48. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, Z.; Li, Y.; Kong, D.; Banerjee, S.; Ahmad, A.; Azmi, A.S.; Ali, S.; Abbruzzese, J.L.; Gallick, G.E.; Sarkar, F.H. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009, 69, 2400–2407. [Google Scholar] [CrossRef] [Green Version]
  50. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell. Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef]
  51. Dhayat, S.A.; Traeger, M.M.; Rehkaemper, J.; Stroese, A.J.; Steinestel, K.; Wardelmann, E.; Kabar, I.; Senninger, N. Clinical impact of epithelial-to-mesenchymal transition regulating microRNAs in pancreatic ductal adenocarcinoma. Cancers 2018, 10, 328. [Google Scholar] [CrossRef] [Green Version]
  52. Wellner, U.; Brabletz, T.; Keck, T. ZEB1 in Pancreatic Cancer. Cancers 2010, 2, 1617–1628. [Google Scholar] [CrossRef] [Green Version]
  53. Polyak, K.; Weinberg, R.A. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar] [CrossRef] [PubMed]
  54. Funamizu, N.; Lacy, C.R.; Parpart, S.T.; Takai, A.; Hiyoshi, Y.; Yanaga, K. MicroRNA-301b promotes cell invasiveness through targeting TP63 in pancreatic carcinoma cells. Int. J. Oncol. 2014, 44, 725–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Funamizu, N.; Hu, C.; Lacy, C.; Schetter, A.; Zhang, G.; He, P.; Gaedcke, J.; Ghadimi, M.B.; Ried, T.; Yfantis, H.G.; et al. Macrophage migration inhibitory factor induces epithelial to mesenchymal transition, enhances tumor aggressiveness and predicts clinical outcome in resected pancreatic ductal adenocarcinoma. Int. J. Cancer 2013, 132, 785–794. [Google Scholar] [CrossRef] [Green Version]
  56. Shah, A.N.; Summy, J.M.; Zhang, J.; Park, S.I.; Parikh, N.U.; Gallick, G.E. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann. Surg. Oncol. 2007, 14, 3629–3637. [Google Scholar] [CrossRef]
  57. Wu, Q.; Wang, X.; Liu, J.; Zheng, J.; Liu, Y.; Li, Y.; Su, F.; Ou, W.; Wang, R. Nutlin-3 reverses the epithelial-mesenchymal transition in gemcitabine-resistant hepatocellular carcinoma cells. Oncol. Rep. 2016, 36, 1325–1332. [Google Scholar] [CrossRef] [Green Version]
  58. Quint, K.; Tonigold, M.; Di Fazio, P.; Montalbano, R.; Lingelbach, S.; Rückert, F.; Alinger, B.; Ocker, M.; Neureiter, D. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial-mesenchymal transition. Int. J. Oncol. 2012, 41, 2093–2102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Pan, Y.; Li, K.; Tao, X.; Zhao, Y.; Chen, Q.; Li, N.; Liu, J.; Go, V.L.W.; Guo, J.; Gao, G.; et al. MicroRNA-34a alleviates gemcitabine resistance in pancreatic cancer by repression of cancer stem cell renewal. Pancreas 2021, 50, 1260–1266. [Google Scholar] [CrossRef]
  61. Zhang, Y.; Wei, J.; Wang, H.; Xue, X.; An, Y.; Tang, D.; Yuan, Z.; Wang, F.; Wu, J.; Zhang, J.; et al. Epithelial mesenchymal transition correlates with CD24+CD44+ and CD133+ cells in pancreatic cancer. Oncol. Rep. 2012, 27, 1599–1605. [Google Scholar] [CrossRef] [Green Version]
  62. Cioffi, M.; Trabulo, S.M.; Sanchez-Ripoll, Y.; Miranda-Lorenzo, I.; Lonardo, E.; Dorado, J.; Reis Vieira, C.; Ramirez, J.C.; Hidalgo, M.; Aicher, A.; et al. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells. Gut 2015, 64, 1936–1948. [Google Scholar] [CrossRef]
  63. Ji, Q.; Hao, X.; Zhang, M.; Tang, W.; Yang, M.; Li, L.; Xiang, D.; Desano, J.T.; Bommer, G.T.; Fan, D.; et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE 2009, 4, e6816. [Google Scholar] [CrossRef]
  64. Okumura, Y.; Noda, T.; Eguchi, H.; Sakamoto, T.; Iwagami, Y.; Yamada, D.; Asaoka, T.; Wada, H.; Kawamoto, K.; Gotoh, K.; et al. Hypoxia-induced PLOD2 is a key regulator in epithelial-mesenchymal transition and chemoresistance in biliary tract cancer. Ann. Surg. Oncol. 2018, 25, 3728–3737. [Google Scholar] [CrossRef]
  65. Arumugam, T.; Ramachandran, V.; Fournier, K.F.; Wang, H.; Marquis, L.; Abbruzzese, J.L.; Gallick, G.E.; Logsdon, C.D.; McConkey, D.J.; Choi, W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009, 69, 5820–5828. [Google Scholar] [CrossRef] [Green Version]
  66. Carrasco-Garcia, E.; Lopez, L.; Moncho-Amor, V.; Carazo, F.; Aldaz, P.; Collado, M.; Bell, D.; Gaafar, A.; Karamitopoulou, E.; Tzankov, A.; et al. SOX9 triggers different epithelial to mesenchymal transition states to promote pancreatic cancer progression. Cancers 2022, 14, 916. [Google Scholar] [CrossRef]
  67. Singh, M.; Yelle, N.; Venugopal, C.; Singh, S.K. EMT: Mechanisms and therapeutic implications. Pharmacol. Ther. 2018, 182, 80–94. [Google Scholar] [CrossRef]
  68. Zhang, G.; Schetter, A.; He, P.; Funamizu, N.; Gaedcke, J.; Ghadimi, B.M.; Ried, T.; Hassan, R.; Yfantis, H.G.; Lee, D.H.; et al. DPEP1 inhibits tumor cell invasiveness, enhances chemosensitivity and predicts clinical outcome in pancreatic ductal adenocarcinoma. PLoS ONE 2012, 7, e31507. [Google Scholar] [CrossRef] [Green Version]
  69. Lawlor, R.T.; Veronese, N.; Nottegar, A.; Malleo, G.; Smith, L.; Demurtas, J.; Cheng, L.; Wood, L.D.; Silvestris, N.; Salvia, R.; et al. Prognostic role of high-grade tumor budding in pancreatic ductal adenocarcinoma: A systematic review and meta-analysis with a focus on epithelial to mesenchymal transition. Cancers 2019, 11, 113. [Google Scholar] [CrossRef] [Green Version]
  70. Krantz, S.B.; Shields, M.A.; Dangi-Garimella, S.; Bentrem, D.J.; Munshi, H.G. Contribution of epithelial-mesenchymal transition to pancreatic cancer progression. Cancers 2010, 2, 2084–2097. [Google Scholar] [CrossRef]
  71. Maier, H.J.; Wirth, T.; Beug, H. Epithelial-mesenchymal transition in pancreatic carcinoma. Cancers 2010, 2, 2058–2083. [Google Scholar] [CrossRef]
  72. Wang, T.; Chen, G.; Ma, X.; Yang, Y.; Chen, Y.; Peng, Y.; Bai, Z.; Zhang, Z.; Pei, H.; Guo, W. Mir-30a regulates cancer cell response to chemotherapy through snai1/irs1/akt pathway. Cell Death Dis. 2019, 10, 153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Funamizu, N.; Lacy, C.R.; Kamada, M.; Yanaga, K.; Manome, Y. MicroRNA-200b and −301 are associated with gemcitabine response as biomarkers in pancreatic carcinoma cells. Int. J. Oncol. 2019, 54, 991–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Li, Y.; Vandenboom, T.G.; Kong, D.; Wang, Z.; Ali, S.; Philip, P.A.; Sarkar, F.H. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009, 69, 6704–6712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Liu, G.; Ji, L.; Ke, M.; Ou, Z.; Tang, N.; Li, Y. miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn. Biomed. Pharmacother. 2018, 106, 523–531. [Google Scholar] [CrossRef]
  76. Fu, X.; Deng, X.; Xiao, W.; Huang, B.; Yi, X.; Zou, Y. Downregulation of NEAT1 sensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine through modulation of the miR-506-3p/ZEB2/EMT axis. Am. J. Cancer Res. 2021, 11, 3841–3856. [Google Scholar]
  77. Li, J.; Wu, H.; Li, W.; Yin, L.; Guo, S.; Xu, X.; Ouyang, Y.; Zhao, Z.; Liu, S.; Tian, Y.; et al. Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-κB signaling. Oncogene 2016, 35, 5501–5514. [Google Scholar] [CrossRef]
  78. Hiramoto, H.; Muramatsu, T.; Ichikawa, D.; Tanimoto, K.; Yasukawa, S.; Otsuji, E.; Inazawa, J. miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Sci. Rep. 2017, 7, 4002. [Google Scholar] [CrossRef] [Green Version]
  79. Yang, R.M.; Zhan, M.; Xu, S.W.; Long, M.M.; Yang, L.H.; Chen, W.; Huang, S.; Liu, Q.; Zhou, J.; Zhu, J.; et al. miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death Dis. 2017, 8, e3129. [Google Scholar] [CrossRef] [Green Version]
  80. Liu, F.; Liu, B.; Qian, J.; Wu, G.; Li, J.; Ma, Z. miR-153 enhances the therapeutic effect of gemcitabine by targeting Snail in pancreatic cancer. Acta Biochim. Biophys. Sin. 2017, 49, 520–529. [Google Scholar] [CrossRef] [Green Version]
  81. Bai, Z.; Sun, J.; Wang, X.; Wang, H.; Pei, H.; Zhang, Z. MicroRNA-153 is a prognostic marker and inhibits cell migration and invasion by targeting SNAI1 in human pancreatic ductal adenocarcinoma. Oncol. Rep. 2015, 34, 595–602. [Google Scholar] [CrossRef]
  82. Wang, Z.; Chen, Y.; Lin, Y.; Wang, X.; Cui, X.; Zhang, Z.; Xian, G.; Qin, C. Novel crosstalk between KLF4 and ZEB1 regulates gemcitabine resistance in pancreatic ductal adenocarcinoma. Int. J. Oncol. 2017, 51, 1239–1248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chaudhary, A.K.; Mondal, G.; Kumar, V.; Kattel, K.; Mahato, R.I. Chemosensitization and inhibition of pancreatic cancer stem cell proliferation by overexpression of microRNA-205. Cancer Lett. 2017, 402, 1–8. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, Z.; Zhao, N.; Cui, J.; Wu, H.; Xiong, J.; Peng, T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell. Oncol. 2020, 43, 123–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hasegawa, S.; Eguchi, H.; Nagano, H.; Konno, M.; Tomimaru, Y.; Wada, H.; Hama, N.; Kawamoto, K.; Kobayashi, S.; Nishida, N.; et al. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br. J. Cancer 2014, 111, 1572–1580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Xiong, G.; Huang, H.; Feng, M.; Yang, G.; Zheng, S.; You, L.; Zheng, L.; Hu, Y.; Zhang, T.; Zhao, Y. Mir-10a-5p targets tfap2c to promote gemcitabine resistance in pancreatic ductal adenocarcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 76. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, W.L.; Zhang, J.H.; Wu, X.Z.; Yan, T.; Lv, W. miR-15b promotes epithelial-mesenchymal transition by inhibiting SMURF2 in pancreatic cancer. Int. J. Oncol. 2015, 47, 1043–1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Yang, S.; He, P.; Wang, J.; Schetter, A.; Tang, W.; Funamizu, N.; Yanaga, K.; Uwagawa, T.; Satoskar, A.R.; Gaedcke, J.; et al. A novel MIF signaling pathway drives the malignant character of pancreatic cancer by targeting NR3C2. Cancer Res. 2016, 76, 3838–3850. [Google Scholar] [CrossRef] [Green Version]
  89. Zhang, K.D.; Hu, B.; Cen, G.; Yang, Y.H.; Chen, W.W.; Guo, Z.Y.; Wang, X.F.; Zhao, Q.; Qiu, Z.J. Mir-301a transcriptionally activated by hif-2alpha promotes hypoxia-induced epithelial-mesenchymal transition by targeting tp63 in pancreatic cancer. World J. Gastroenterol. 2020, 26, 2349–2373. [Google Scholar] [CrossRef]
  90. Okazaki, J.; Tanahashi, T.; Sato, Y.; Miyoshi, J.; Nakagawa, T.; Kimura, T.; Miyamoto, H.; Fujino, Y.; Nakamura, F.; Takehara, M.; et al. MicroRNA-296-5p promotes cell invasion and drug resistance by targeting Bcl2-related ovarian killer, leading to a poor prognosis in pancreatic cancer. Digestion 2020, 101, 794–806. [Google Scholar] [CrossRef]
  91. Ma, J.; Fang, B.; Zeng, F.; Ma, C.; Pang, H.; Cheng, L.; Shi, Y.; Wang, H.; Yin, B.; Xia, J.; et al. Down-regulation of mir-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Oncotarget 2015, 6, 1740–1749. [Google Scholar] [CrossRef] [PubMed]
  92. Yu, S.; Wang, M.; Zhang, H.; Guo, X.; Qin, R. Circ_0092367 inhibits EMT and gemcitabine resistance in pancreatic cancer via regulating the miR-1206/ESRP1 axis. Genes 2021, 12, 1701. [Google Scholar] [CrossRef]
  93. Yu, Q.; Xiu, Z.; Jian, Y.; Zhou, J.; Chen, X.; Chen, X.; Chen, C.; Chen, H.; Yang, S.; Yin, L.; et al. microRNA-497 prevents pancreatic cancer stem cell gemcitabine resistance, migration, and invasion by directly targeting nuclear factor kappa B 1. Aging 2022, 14, 5908–5924. [Google Scholar] [CrossRef] [PubMed]
  94. Ma, C.; Huang, T.; Ding, Y.C.; Yu, W.; Wang, Q.; Meng, B.; Luo, S.X. Microrna-200c overexpression inhibits chemoresistance, invasion and colony formation of human pancreatic cancer stem cells. Int. J. Clin. Exp. Pathol. 2015, 8, 6533–6539. [Google Scholar]
  95. Feig, C.; Gopinathan, A.; Neesse, A.; Chan, D.S.; Cook, N.; Tuveson, D.A. The pancreas cancer microenvironment. Clin. Cancer Res. 2012, 18, 4266–4276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Deng, D.; Patel, R.; Chiang, C.Y.; Hou, P. Role of the tumor microenvironment in regulating pancreatic cancer therapy resistance. Cells 2022, 11, 2952. [Google Scholar] [CrossRef] [PubMed]
  97. Neesse, A.; Michl, P.; Frese, K.K.; Feig, C.; Cook, N.; Jacobetz, M.A.; Lolkema, M.P.; Buchholz, M.; Olive, K.P.; Gress, T.M.; et al. Stromal biology and therapy in pancreatic cancer. Gut 2011, 60, 861–868. [Google Scholar] [CrossRef]
  98. Deshmukh, S.K.; Tyagi, N.; Khan, M.A.; Srivastava, S.K.; Al-Ghadhban, A.; Dugger, K.; Carter, J.E.; Singh, S.; Singh, A.P. Gemcitabine treatment promotes immunosuppressive microenvironment in pancreatic tumors by supporting the infiltration, growth, and polarization of macrophages. Sci. Rep. 2018, 8, 12000. [Google Scholar] [CrossRef] [Green Version]
  99. Ahmad, I.M.; Dafferner, A.J.; O’Connell, K.A.; Mehla, K.; Britigan, B.E.; Hollingsworth, M.A.; Abdalla, M.Y. Heme Oxygenase-1 inhibition potentiates the effects of nab-paclitaxel-gemcitabine and modulates the tumor microenvironment in pancreatic ductal adenocarcinoma. Cancers 2021, 13, 2264. [Google Scholar] [CrossRef]
  100. Wei, L.; Ye, H.; Li, G.; Lu, Y.; Zhou, Q.; Zheng, S.; Lin, Q.; Liu, Y.; Li, Z.; Chen, R. Cancer-associated fibroblasts promote progression and gemcitabine resistance via the SDF-1/SATB-1 pathway in pancreatic cancer. Cell Death Dis. 2018, 9, 1065. [Google Scholar] [CrossRef] [Green Version]
  101. Gu, Z.; Du, Y.; Zhao, X.; Wang, C. Tumor microenvironment and metabolic remodeling in gemcitabine-based chemoresistance of pancreatic cancer. Cancer Lett. 2021, 521, 98–108. [Google Scholar] [CrossRef]
  102. Park, J.; Schwarzbauer, J.E. Mammary epithelial cell interactions with fibronectin stimulate epithelial-mesenchymal transition. Oncogene 2014, 33, 1649–1657. [Google Scholar] [CrossRef] [PubMed]
  103. Miyamoto, H.; Murakami, T.; Tsuchida, K.; Sugino, H.; Miyake, H.; Tashiro, S. Tumor-stroma interaction of human pancreatic cancer: Acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas 2004, 28, 38–44. [Google Scholar] [CrossRef] [PubMed]
  104. Amrutkar, M.; Aasrum, M.; Verbeke, C.S.; Gladhaug, I.P. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer 2019, 19, 596. [Google Scholar] [CrossRef] [Green Version]
  105. Topalovski, M.; Brekken, R.A. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett. 2016, 381, 252–258. [Google Scholar] [CrossRef] [Green Version]
  106. Dangi-Garimella, S.; Krantz, S.B.; Barron, M.R.; Shields, M.A.; Heiferman, M.J.; Grippo, P.J.; Bentrem, D.J.; Munshi, H.G. Three-dimensional collagen I promotes gemcitabine resistance in pancreatic cancer through MT1-MMP-mediated expression of HMGA2. Cancer Res. 2011, 71, 1019–1028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; et al. Randomized Phase III trial of Pegvorhyaluronidase Alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J. Clin. Oncol. 2020, 38, 3185–3194. [Google Scholar] [CrossRef]
  108. Ho, W.J.; Jaffee, E.M.; Zheng, L. The tumour microenvironment in pancreatic cancer—Clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 2020, 17, 527–540. [Google Scholar] [CrossRef]
  109. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  110. Richards, K.E.; Xiao, W.; Hill, R.; On Behalf Of The Usc Pancreas Research Team. Cancer-associated fibroblasts confer gemcitabine resistance to pancreatic cancer cells through PTEN-targeting miRNAs in exosomes. Cancers 2022, 14, 2812. [Google Scholar] [CrossRef]
  111. Zhang, D.; Li, L.; Jiang, H.; Li, Q.; Wang-Gillam, A.; Yu, J.; Head, R.; Liu, J.; Ruzinova, M.B.; Lim, K.H. Tumor-stroma IL1beta-IRAK4 feedforward circuitry drives tumor fibrosis, chemoresistance, and poor prognosis in pancreatic cancer. Cancer Res. 2018, 78, 1700–1712. [Google Scholar] [CrossRef] [Green Version]
  112. Hu, C.; Xia, R.; Zhang, X.; Li, T.; Ye, Y.; Li, G.; He, R.; Li, Z.; Lin, Q.; Zheng, S.; et al. circFARP1 enables cancer-associated fibroblasts to promote gemcitabine resistance in pancreatic cancer via the LIF/STAT3 axis. Mol. Cancer 2022, 21, 24. [Google Scholar] [CrossRef]
  113. Wei, L.; Lin, Q.; Lu, Y.; Li, G.; Huang, L.; Fu, Z.; Chen, R.; Zhou, Q. Cancer-associated fibroblasts-mediated ATF4 expression promotes malignancy and gemcitabine resistance in pancreatic cancer via the TGF-β1/SMAD2/3 pathway and ABCC1 transactivation. Cell Death Dis. 2021, 12, 334. [Google Scholar] [CrossRef]
  114. Ikenaga, N.; Ohuchida, K.; Mizumoto, K.; Akagawa, S.; Fujiwara, K.; Eguchi, D.; Kozono, S.; Ohtsuka, T.; Takahata, S.; Tanaka, M. Pancreatic cancer cells enhance the ability of collagen internalization during epithelial-mesenchymal transition. PLoS ONE 2012, 7, e40434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Zheng, M.; Li, H.; Sun, L.; Brigstock, D.R.; Gao, R. Interleukin-6 participates in human pancreatic stellate cell activation and collagen I production via TGF-β1/Smad pathway. Cytokine 2021, 143, 155536. [Google Scholar] [CrossRef] [PubMed]
  116. Cao, F.; Li, J.; Sun, H.; Liu, S.; Cui, Y.; Li, F. HES 1 is essential for chemoresistance induced by stellate cells and is associated with poor prognosis in pancreatic cancer. Oncol. Rep. 2015, 33, 1883–1889. [Google Scholar] [CrossRef] [Green Version]
  117. Lee, E.; Ryu, G.R.; Ko, S.H.; Ahn, Y.B.; Song, K.H. A role of pancreatic stellate cells in islet fibrosis and β-cell dysfunction in type 2 diabetes mellitus. Biochem. Biophys. Res. Commun. 2017, 485, 328–334. [Google Scholar] [CrossRef] [PubMed]
  118. Kikuta, K.; Masamune, A.; Watanabe, T.; Ariga, H.; Itoh, H.; Hamada, S.; Satoh, K.; Egawa, S.; Unno, M.; Shimosegawa, T. Pancreatic stellate cells promote epithelial-mesenchymal transition in pancreatic cancer cells. Biochem. Biophys. Res. Commun. 2010, 403, 380–384. [Google Scholar] [CrossRef] [PubMed]
  119. LaRue, M.M.; Parker, S.; Puccini, J.; Cammer, M.; Kimmelman, A.C.; Bar-Sagi, D. Metabolic reprogramming of tumor-associated macrophages by collagen turnover promotes fibrosis in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2022, 119, e2119168119. [Google Scholar] [CrossRef]
  120. Helm, O.; Held-Feindt, J.; Grage-Griebenow, E.; Reiling, N.; Ungefroren, H.; Vogel, I.; Krüger, U.; Becker, T.; Ebsen, M.; Röcken, C.; et al. Tumor-associated macrophages exhibit pro- and anti-inflammatory properties by which they impact on pancreatic tumorigenesis. Int. J. Cancer 2014, 135, 843–861. [Google Scholar] [CrossRef]
  121. Weizman, N.; Krelin, Y.; Shabtay-Orbach, A.; Amit, M.; Binenbaum, Y.; Wong, R.J.; Gil, Z. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene 2014, 33, 3812–3819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Guo, Y.; Wu, H.; Xiong, J.; Gou, S.; Cui, J.; Peng, T. miR-222-3p-containing macrophage-derived extracellular vesicles confer gemcitabine resistance via TSC1-mediated mTOR/AKT/PI3K pathway in pancreatic cancer. Cell Biol. Toxicol. 2022. [Google Scholar] [CrossRef]
  123. Nagathihalli, N.S.; Castellanos, J.A.; Shi, C.; Beesetty, Y.; Reyzer, M.L.; Caprioli, R.; Chen, X.; Walsh, A.J.; Skala, M.C.; Moses, H.L.; et al. Signal transducer and activator of transcription 3, mediated remodeling of the tumor microenvironment results in enhanced tumor drug delivery in a mouse model of pancreatic cancer. Gastroenterology 2015, 149, 1932–1943.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mitchem, J.B.; Brennan, D.J.; Knolhoff, B.L.; Belt, B.A.; Zhu, Y.; Sanford, D.E.; Belaygorod, L.; Carpenter, D.; Collins, L.; Piwnica-Worms, D.; et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013, 73, 1128–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Oba, T.; Sato, N.; Adachi, Y.; Amaike, T.; Kudo, Y.; Koga, A.; Kohi, S.; Hirata, K. Hypoxia increases KIAA1199/CEMIP expression and enhances cell migration in pancreatic cancer. Sci. Rep. 2021, 11, 18193. [Google Scholar] [CrossRef]
  126. Keleg, S.; Kayed, H.; Jiang, X.; Penzel, R.; Giese, T.; Büchler, M.W.; Friess, H.; Kleeff, J. Adrenomedullin is induced by hypoxia and enhances pancreatic cancer cell invasion. Int. J. Cancer. 2007, 121, 21–32. [Google Scholar] [CrossRef] [PubMed]
  127. Chee, N.T.; Carriere, C.H.; Miller, Z.; Welford, S.; Brothers, S.P. Activating transcription factor 4 regulates hypoxia inducible factor 1α in chronic hypoxia in pancreatic cancer cells. Oncol. Rep. 2023, 49, 14. [Google Scholar] [CrossRef]
  128. Fan, Y.; Xu, L.L.; Shi, C.Y.; Wei, W.; Wang, D.S.; Cai, D.F. MicroRNA-454 regulates stromal cell derived factor-1 in the control of the growth of pancreatic ductal adenocarcinoma. Sci. Rep. 2016, 6, 22793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Bao, B.; Ali, S.; Ahmad, A.; Azmi, A.S.; Li, Y.; Banerjee, S.; Kong, D.; Sethi, S.; Aboukameel, A.; Padhye, S.B.; et al. Hypoxia-induced aggressiveness of pancreatic cancer cells is due to increased expression of VEGF, IL-6 and miR-21, which can be attenuated by CDF treatment. PLoS ONE 2012, 7, e50165. [Google Scholar] [CrossRef] [Green Version]
  130. Nong, K.; Zhang, D.; Chen, C.; Yang, Y.; Yang, Y.; Liu, S.; Cai, H. MicroRNA-519 inhibits hypoxia-induced tumorigenesis of pancreatic cancer by regulating immune checkpoint PD-L1. Oncol. Lett. 2020, 19, 1427–1433. [Google Scholar] [CrossRef]
  131. Luo, G.; Xia, X.; Wang, X.; Zhang, K.; Cao, J.; Jiang, T.; Zhao, Q.; Qiu, Z. miR-301a plays a pivotal role in hypoxia-induced gemcitabine resistance in pancreatic cancer. Exp. Cell Res. 2018, 369, 120–128. [Google Scholar] [CrossRef] [PubMed]
  132. Liu, Y.F.; Luo, D.; Li, X.; Li, Z.Q.; Yu, X.; Zhu, H.W. PVT1 knockdown inhibits autophagy and improves gemcitabine sensitivity by regulating the MiR-143/HIF-1α/VMP1 axis in pancreatic cancer. Pancreas 2021, 50, 227–234. [Google Scholar] [CrossRef]
  133. Xin, X.; Kumar, V.; Lin, F.; Kumar, V.; Bhattarai, R.; Bhatt, V.R.; Tan, C.; Mahato, R.I. Redox-responsive nanoplatform for codelivery of miR-519c and gemcitabine for pancreatic cancer therapy. Sci. Adv. 2020, 6, eabd6764. [Google Scholar] [CrossRef] [PubMed]
  134. Kasuya, K.; Tsuchida, A.; Nagakawa, Y.; Suzuki, M.; Abe, Y.; Itoi, T.; Serizawa, H.; Nagao, T.; Shimazu, M.; Aoki, T. Hypoxia-inducible factor-1α expression and gemcitabine chemotherapy for pancreatic cancer. Oncol. Rep. 2011, 26, 1399–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Yang, X.; Yin, H.; Zhang, Y.; Li, X.; Tong, H.; Zeng, Y.; Wang, Q.; He, W. Hypoxia-induced autophagy promotes gemcitabine resistance in human bladder cancer cells through hypoxia-inducible factor 1α activation. Int. J. Oncol. 2018, 53, 215–224. [Google Scholar] [CrossRef] [Green Version]
  136. Liu, A.; Zhou, Y.; Zhao, T.; Tang, X.; Zhou, B.; Xu, J. MiRNA-3662 reverses the gemcitabine resistance in pancreatic cancer through regulating the tumor metabolism. Cancer Chemother. Pharmacol. 2021, 88, 343–357. [Google Scholar] [CrossRef]
  137. Ni, J.; Zhou, S.; Yuan, W.; Cen, F.; Yan, Q. Mechanism of miR-210 involved in epithelial-mesenchymal transition of pancreatic cancer cells under hypoxia. J. Recept. Signal Transduct. 2019, 39, 399–406. [Google Scholar] [CrossRef] [PubMed]
  138. Zhang, L.; Yao, J.; Li, W.; Zhang, C. Micro-RNA-21 regulates cancer-associated fibroblast-mediated drug resistance in pancreatic cancer. Oncol. Res. 2018, 26, 827–835. [Google Scholar] [CrossRef]
  139. Kadera, B.E.; Li, L.; Toste, P.A.; Wu, N.; Adams, C.; Dawson, D.W.; Donahue, T.R. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS ONE 2013, 8, e71978. [Google Scholar] [CrossRef] [Green Version]
  140. Mace, T.A.; Collins, A.L.; Wojcik, S.E.; Croce, C.M.; Lesinski, G.B.; Bloomston, M. Hypoxia induces the overexpression of microRNA-21 in pancreatic cancer cells. J. Surg. Res. 2013, 184, 855–860. [Google Scholar] [CrossRef] [Green Version]
  141. Wang, P.; Zhuang, L.; Zhang, J.; Fan, J.; Luo, J.; Chen, H.; Wang, K.; Liu, L.; Chen, Z.; Meng, Z. The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FASL. Mol. Oncol. 2013, 7, 334–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Park, J.K.; Lee, E.J.; Esau, C.; Schmittgen, T.D. Antisense inhibition of microRNA-21 or -221 arrests cell cycle, induces apoptosis, and sensitizes the effects of gemcitabine in pancreatic adenocarcinoma. Pancreas 2009, 38, e190–e199. [Google Scholar] [CrossRef]
  143. Fang, Y.; Zhou, W.; Rong, Y.; Kuang, T.; Xu, X.; Wu, W.; Wang, D.; Lou, W. Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp. Cell Res. 2019, 383, 111543. [Google Scholar] [CrossRef] [PubMed]
  144. Masamune, A.; Nakano, E.; Hamada, S.; Takikawa, T.; Yoshida, N.; Shimosegawa, T. Alteration of the microRNA expression profile during the activation of pancreatic stellate cells. Scand. J. Gastroenterol. 2014, 49, 323–331. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, M.; Zhang, Y.; Yang, J.; Cui, X.; Zhou, Z.; Zhan, H.; Ding, K.; Tian, X.; Yang, Z.; Fung, K.A.; et al. ZIP4 Increases Expression of transcription factor ZEB1 to Promote integrin α3β1 Signaling and Inhibit Expression of the gemcitabine Transporter ENT1 in Pancreatic Cancer Cells. Gastroenterology 2020, 158, 679–692.e1. [Google Scholar] [CrossRef]
  146. Hu, Q.; Qin, Y.; Zhang, B.; Liang, C.; Ji, S.; Shi, S.; Xu, W.; Xiang, J.; Liang, D.; Ni, Q.; et al. FBW7 increases the chemosensitivity of pancreatic cancer cells to gemcitabine through upregulation of ENT1. Oncol. Rep. 2017, 38, 2069–2077. [Google Scholar] [CrossRef] [Green Version]
  147. Michalski, C.W.; Erkan, M.; Sauliunaite, D.; Giese, T.; Stratmann, R.; Sartori, C.; Giese, N.A.; Friess, H.; Kleeff, J. Ex vivo chemosensitivity testing and gene expression profiling predict response towards adjuvant gemcitabine treatment in pancreatic cancer. Br. J. Cancer 2008, 99, 760–767. [Google Scholar] [CrossRef] [Green Version]
  148. Saiki, Y.; Hirota, S.; Horii, A. Attempts to remodel the pathways of gemcitabine metabolism: Recent approaches to overcoming tumours with acquired chemoresistance. Cancer Drug Resist. 2020, 3, 819–831. [Google Scholar] [CrossRef]
  149. Hagmann, W.; Faissner, R.; Schnölzer, M.; Löhr, M.; Jesnowski, R. Membrane drug transporters and chemoresistance in human pancreatic carcinoma. Cancers 2010, 3, 106–125. [Google Scholar] [CrossRef] [Green Version]
  150. Hung, S.W.; Marrache, S.; Cummins, S.; Bhutia, Y.D.; Mody, H.; Hooks, S.B.; Dhar, S.; Govindarajan, R. Defective hCNT1 transport contributes to gemcitabine chemoresistance in ovarian cancer subtypes: Overcoming transport defects using a nanoparticle approach. Cancer Lett. 2015, 359, 233–240. [Google Scholar] [CrossRef] [Green Version]
  151. Bhutia, Y.D.; Hung, S.W.; Patel, B.; Lovin, D.; Govindarajan, R. CNT1 expression influences proliferation and chemosensitivity in drug-resistant pancreatic cancer cells. Cancer Res. 2011, 71, 1825–1835. [Google Scholar] [CrossRef] [Green Version]
  152. Maréchal, R.; Mackey, J.R.; Lai, R.; Demetter, P.; Peeters, M.; Polus, M.; Cass, C.E.; Young, J.; Salmon, I.; Devière, J.; et al. Human equilibrative nucleoside transporter 1 and human concentrative nucleoside transporter 3 predict survival after adjuvant gemcitabine therapy in resected pancreatic adenocarcinoma. Clin. Cancer Res. 2009, 15, 2913–2919. [Google Scholar] [CrossRef]
  153. García-Manteiga, J.; Molina-Arcas, M.; Casado, F.J.; Mazo, A.; Pastor-Anglada, M. Nucleoside transporter profiles in human pancreatic cancer cells: Role of hCNT1 in 2′,2′-difluorodeoxycytidine- induced cytotoxicity. Clin. Cancer Res. 2003, 9, 5000–5008. [Google Scholar] [PubMed]
  154. Rauchwerger, D.R.; Firby, P.S.; Hedley, D.W.; Moore, M.J. Equilibrative-sensitive nucleoside transporter and its role in gemcitabine sensitivity. Cancer Res. 2000, 60, 6075–6079. [Google Scholar]
  155. Tsesmetzis, N.; Paulin, C.B.J.; Rudd, S.G.; Herold, N. Nucleobase and nucleoside analogues: Resistance and re-sensitisation at the level of pharmacokinetics, pharmacodynamics and metabolism. Cancers 2018, 10, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Giovannetti, E.; Del Tacca, M.; Mey, V.; Funel, N.; Nannizzi, S.; Ricci, S.; Orlandini, C.; Boggi, U.; Campani, D.; Del Chiaro, M.; et al. Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine. Cancer Res. 2006, 66, 3928–3935. [Google Scholar] [CrossRef] [Green Version]
  157. Mackey, J.R.; Mani, R.S.; Selner, M.; Mowles, D.; Young, J.D.; Belt, J.A.; Crawford, C.R.; Cass, C.E. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res. 1998, 58, 4349–4357. [Google Scholar]
  158. Aughton, K.; Elander, N.O.; Evans, A.; Jackson, R.; Campbell, F.; Costello, E.; Halloran, C.M.; Mackey, J.R.; Scarfe, A.G.; Valle, J.W.; et al. hENT1 predicts benefit from gemcitabine in pancreatic cancer but only with low CDA mRNA. Cancers 2021, 13, 5758. [Google Scholar] [CrossRef]
  159. Farrell, J.J.; Elsaleh, H.; Garcia, M.; Lai, R.; Ammar, A.; Regine, W.F.; Abrams, R.; Benson, A.B.; Macdonald, J.; Cass, C.E.; et al. Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterology 2009, 136, 187–195. [Google Scholar] [CrossRef] [PubMed]
  160. Skrypek, N.; Duchêne, B.; Hebbar, M.; Leteurtre, E.; van Seuningen, I.; Jonckheere, N. The MUC4 mucin mediates gemcitabine resistance of human pancreatic cancer cells via the Concentrative nucleoside Transporter family. Oncogene 2013, 32, 1714–1723. [Google Scholar] [CrossRef] [Green Version]
  161. Qiu, F.; Chen, J.; Cao, J.; Diao, F.; Huang, P. Low-intensity low-frequency ultrasound enhances the chemosensitivity of gemcitabine-resistant ASPC-1 cells via PI3K/AKT/NF-κB pathway-mediated ABC transporters. Oncol. Rep. 2020, 44, 1158–1168. [Google Scholar] [CrossRef]
  162. Mini, E.; Nobili, S.; Caciagli, B.; Landini, I.; Mazzei, T. Cellular pharmacology of gemcitabine. Ann. Oncol. 2006, 17 (Suppl. S5), v7–v12. [Google Scholar] [CrossRef] [PubMed]
  163. Neff, T.; Blau, C.A. Forced expression of cytidine deaminase confers resistance to cytosine arabinoside and gemcitabine. Exp. Hematol. 1996, 24, 1340–1346. [Google Scholar] [PubMed]
  164. Eliopoulos, N.; Cournoyer, D.; Momparler, R.L. Drug resistance to 5-aza-2′-deoxycytidine, 2′,2′-difluorodeoxycytidine, and cytosine arabinoside conferred by retroviral-mediated transfer of human cytidine deaminase cDNA into murine cells. Cancer Chemother. Pharmacol. 1998, 42, 373–378. [Google Scholar] [CrossRef]
  165. Kanno, S.; Hiura, T.; Ohtake, T.; Koiwai, K.; Suzuki, H.; Ujibe, M.; Ishikawa, M. Characterization of resistance to cytosine arabinoside (ara-C) in NALM-6 human B leukemia cells. Clin. Chim. Acta 2007, 377, 144–149. [Google Scholar] [CrossRef] [PubMed]
  166. Ohta, T.; Hori, H.; Ogawa, M.; Miyahara, M.; Kawasaki, H.; Taniguchi, N.; Komada, Y. Impact of cytidine deaminase activity on intrinsic resistance to cytarabine in carcinoma cells. Oncol. Rep. 2004, 12, 1115–1120. [Google Scholar] [CrossRef] [PubMed]
  167. Ludovini, V.; Floriani, I.; Pistola, L.; Minotti, V.; Meacci, M.; Chiari, R.; Garavaglia, D.; Tofanetti, F.R.; Flacco, A.; Siggillino, A.; et al. Association of cytidine deaminase and xeroderma pigmentosum group D polymorphisms with response, toxicity, and survival in cisplatin/gemcitabine-treated advanced non-small cell lung cancer patients. J. Thorac. Oncol. 2011, 6, 2018–2026. [Google Scholar] [CrossRef] [Green Version]
  168. Blackstock, A.W.; Lightfoot, H.; Case, L.D.; Tepper, J.E.; Mukherji, S.K.; Mitchell, B.S.; Swarts, S.G.; Hess, S.M. Tumor uptake and elimination of 2′, 2. Clin. Cancer Res. 2001, 7, 3263–3268. [Google Scholar]
  169. Manome, Y.; Wen, P.Y.; Dong, Y.; Tanaka, T.; Mitchell, B.S.; Kufe, D.W.; Fine, H.A. Viral vector transduction of the human deoxycytidine kinase cDNA sensitizes glioma cells to the cytotoxic effects of cytosine arabinoside in vitro and in vivo. Nat. Med. 1996, 2, 567–573. [Google Scholar] [CrossRef]
  170. Ohhashi, S.; Ohuchida, K.; Mizumoto, K.; Fujita, H.; Egami, T.; Yu, J.; Toma, H.; Sadatomi, S.; Nagai, E.; Tanaka, M. Down-regulation of deoxycytidine kinase enhances acquired resistance to gemcitabine in pancreatic cancer. Anticancer Res. 2008, 28, 2205–2212. [Google Scholar]
  171. Ohmine, K.; Kawaguchi, K.; Ohtsuki, S.; Motoi, F.; Ohtsuka, H.; Kamiie, J.; Abe, T.; Unno, M.; Terasaki, T. Quantitative targeted proteomics of pancreatic cancer: Deoxycytidine kinase protein level correlates to progression-free survival of patients receiving gemcitabine treatment. Mol. Pharm. 2015, 12, 3282–3291. [Google Scholar] [CrossRef] [PubMed]
  172. Kamada, M.; Akiyoshi, K.; Akiyama, N.; Funamizu, N.; Watanabe, M.; Fujioka, K.; Ikeda, K.; Manome, Y. Cholangiocarcinoma cell line TK may be useful for the pharmacokinetic study of the chemotherapeutic agent gemcitabine. Oncol. Rep. 2014, 32, 829–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Elledge, S.J.; Zhou, Z.; Allen, J.B. Ribonucleotide reductase: Regulation, regulation, regulation. Trends Biochem. Sci. 1992, 17, 119–123. [Google Scholar] [CrossRef]
  174. Duxbury, M.S.; Ito, H.; Benoit, E.; Zinner, M.J.; Ashley, S.W.; Whang, E.E. Retrovirally mediated RNA interference targeting the M2 subunit of ribonucleotide reductase: A novel therapeutic strategy in pancreatic cancer. Surgery 2004, 136, 261–269. [Google Scholar] [CrossRef] [PubMed]
  175. Vena, F.; Li Causi, E.; Rodriguez-Justo, M.; Goodstal, S.; Hagemann, T.; Hartley, J.A.; Hochhauser, D. The MEK1/2 inhibitor Pimasertib enhances gemcitabine efficacy in pancreatic cancer models by altering ribonucleotide reductase subunit-1 (RRM1). Clin. Cancer Res. 2015, 21, 5563–5577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Nakamura, J.; Kohya, N.; Kai, K.; Ohtaka, K.; Hashiguchi, K.; Hiraki, M.; Kitajima, Y.; Tokunaga, O.; Noshiro, H.; Miyazaki, K. Ribonucleotide reductase subunit M1 assessed by quantitative double-fluorescence immunohistochemistry predicts the efficacy of gemcitabine in biliary tract carcinoma. Int. J. Oncol. 2010, 37, 845–852. [Google Scholar]
  177. Sierzega, M.; Pach, R.; Kulig, P.; Legutko, J.; Kulig, J. Prognostic implications of expression profiling for gemcitabine-related genes (hENT1, dCK, RRM1, RRM2) in patients with resectable pancreatic adenocarcinoma receiving adjuvant chemotherapy. Pancreas 2017, 46, 684–689. [Google Scholar] [CrossRef]
  178. Amponsah, P.S.; Fan, P.; Bauer, N.; Zhao, Z.; Gladkich, J.; Fellenberg, J.; Herr, I. MicroRNA, 210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer. Cancer Lett. 2017, 388, 107–117. [Google Scholar] [CrossRef]
  179. Gu, J.; Huang, W.; Wang, X.; Zhang, J.; Tao, T.; Zheng, Y.; Liu, S.; Yang, J.; Chen, Z.S.; Cai, C.Y.; et al. Hsa-miR-3178/RhoB/PI3K/Akt, a novel signaling pathway regulates ABC transporters to reverse gemcitabine resistance in pancreatic cancer. Mol. Cancer 2022, 21, 112. [Google Scholar] [CrossRef]
  180. Wang, F.; Xue, X.; Wei, J.; An, Y.; Yao, J.; Cai, H.; Wu, J.; Dai, C.; Qian, Z.; Xu, Z.; et al. hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br. J. Cancer 2010, 103, 567–574. [Google Scholar] [CrossRef] [Green Version]
  181. Wu, Y.; Xu, W.; Yang, Y.; Zhang, Z. miRNA-93-5p promotes gemcitabine resistance in pancreatic cancer cells by targeting the PTEN-mediated PI3K/Akt signaling pathway. Ann. Clin. Lab. Sci. 2021, 51, 310–320. [Google Scholar] [PubMed]
  182. Zhan, T.; Chen, X.; Tian, X.; Han, Z.; Liu, M.; Zou, Y.; Huang, S.; Chen, A.; Cheng, X.; Deng, J.; et al. MiR-331-3p links to drug resistance of pancreatic cancer cells by activating WNT/β-catenin signal via ST7L. Technol. Cancer Res. Treat. 2020, 19, 1533033820945801. [Google Scholar] [CrossRef]
  183. Fan, P.; Liu, L.; Yin, Y.; Zhao, Z.; Zhang, Y.; Amponsah, P.S.; Xiao, X.; Bauer, N.; Abukiwan, A.; Nwaeburu, C.C.; et al. MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett. 2016, 373, 130–137. [Google Scholar] [CrossRef] [PubMed]
  184. Bhutia, Y.D.; Hung, S.W.; Krentz, M.; Patel, D.; Lovin, D.; Manoharan, R.; Thomson, J.M.; Govindarajan, R. Differential processing of let-7a precursors influences RRM2 expression and chemosensitivity in pancreatic cancer: Role of LIN-28 and SET oncoprotein. PLoS ONE 2013, 8, e53436. [Google Scholar] [CrossRef] [Green Version]
  185. Rajabpour, A.; Afgar, A.; Mahmoodzadeh, H.; Radfar, J.E.; Rajaei, F.; Teimoori-Toolabi, L. MiR-608 regulating the expression of ribonucleotide reductase M1 and cytidine deaminase is repressed through induced gemcitabine chemoresistance in pancreatic cancer cells. Cancer Chemother. Pharmacol. 2017, 80, 765–775. [Google Scholar] [CrossRef] [PubMed]
  186. Lu, H.; Lu, S.; Yang, D.; Zhang, L.; Ye, J.; Li, M.; Hu, W. MiR-20a-5p regulates gemcitabine chemosensitivity by targeting RRM2 in pancreatic cancer cells and serves as a predictor for gemcitabine-based chemotherapy. Biosci. Rep. 2019, 39, BSR20181374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Patel, G.K.; Khan, M.A.; Bhardwaj, A.; Srivastava, S.K.; Zubair, H.; Patton, M.C.; Singh, S.; Khushman, M.; Singh, A.P. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br. J. Cancer 2017, 116, 609–619. [Google Scholar] [CrossRef]
  188. Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef] [PubMed]
  189. Richards, K.E.; Zeleniak, K.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 30, 1770–1778. [Google Scholar] [CrossRef] [Green Version]
  190. Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; et al. MicroRNA-155 controls exosome synthess and promotes gemcitabine resistance in pancreatic ductal adenocarcinoma. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef]
  191. Comanedatore, A.; Immordino, B.; Balsano, M.; Capula, M.; Garajova, I.; Ciccolini, J.; Giovannetti, E.; Morelli, L. Potenital role of exosomes in the chemoresistance to gemcitabine and nab-paclitaxel in pancreatic cancer. Diagnostics 2022, 12, 286. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Association between microRNAs related to EMT (Epithelial–mesenchymal transition) and gemcitabine resistance. EMT is well known to increase drug resistance, cell proliferation, and invasiveness through acquiring properties like cancer stem cells.
Figure 1. Association between microRNAs related to EMT (Epithelial–mesenchymal transition) and gemcitabine resistance. EMT is well known to increase drug resistance, cell proliferation, and invasiveness through acquiring properties like cancer stem cells.
Cancers 15 01230 g001
Figure 2. Association of microRNAs with TME (tumor microenvironment) and gemcitabine resistance. TME works to the advantage of cancer cell survival by, for example, inducing gemcitabine resistance and enhancing cell proliferation.
Figure 2. Association of microRNAs with TME (tumor microenvironment) and gemcitabine resistance. TME works to the advantage of cancer cell survival by, for example, inducing gemcitabine resistance and enhancing cell proliferation.
Cancers 15 01230 g002
Figure 3. Association of miRNAs with metabolism and gemcitabine resistance. Mainly, transporter and deoxycytidine kinase (dCK) are associated with the activation of gemcitabine. Conversely, cytidine deaminase (CDA) and ribonucleotide reductase (RR) are associated with the inactivation of gemcitabine. These transporters and enzymes are controlled by miRNAs, which results in gemcitabine resistance in pancreatic cancer.
Figure 3. Association of miRNAs with metabolism and gemcitabine resistance. Mainly, transporter and deoxycytidine kinase (dCK) are associated with the activation of gemcitabine. Conversely, cytidine deaminase (CDA) and ribonucleotide reductase (RR) are associated with the inactivation of gemcitabine. These transporters and enzymes are controlled by miRNAs, which results in gemcitabine resistance in pancreatic cancer.
Cancers 15 01230 g003
Table 3. miRNAs for metabolism.
Table 3. miRNAs for metabolism.
AuthorRef. NumbermiRNATarget Gene
Sensitivity
Bhutia YD [184]let-7aRRM2
Lu H[186]miR-20aRRM2
Fan P[183]miR-101RRM1
Amponsah PS[178]miR-210ABCC5
Maftouh M[17]miR-211RRM2
Rajabpour A[185]miR-608CDA, RRM1
Gu J[179]miR-3178ABC transporter
Resistance
Wu Y[181]miR-93PTEN
Patel GK[187]miR-155dCK
Zhan T[182]miR-331ST7L
Xin X[133]miR-519cABCG2
Ref: reference.
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

Funamizu, N.; Honjo, M.; Tamura, K.; Sakamoto, K.; Ogawa, K.; Takada, Y. microRNAs Associated with Gemcitabine Resistance via EMT, TME, and Drug Metabolism in Pancreatic Cancer. Cancers 2023, 15, 1230. https://doi.org/10.3390/cancers15041230

AMA Style

Funamizu N, Honjo M, Tamura K, Sakamoto K, Ogawa K, Takada Y. microRNAs Associated with Gemcitabine Resistance via EMT, TME, and Drug Metabolism in Pancreatic Cancer. Cancers. 2023; 15(4):1230. https://doi.org/10.3390/cancers15041230

Chicago/Turabian Style

Funamizu, Naotake, Masahiko Honjo, Kei Tamura, Katsunori Sakamoto, Kohei Ogawa, and Yasutsugu Takada. 2023. "microRNAs Associated with Gemcitabine Resistance via EMT, TME, and Drug Metabolism in Pancreatic Cancer" Cancers 15, no. 4: 1230. https://doi.org/10.3390/cancers15041230

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