Skip to main content

Pancreatic cancer stemness: dynamic status in malignant progression

Abstract

Pancreatic cancer (PC) is one of the most aggressive malignancies worldwide. Increasing evidence suggests that the capacity for self-renewal, proliferation, and differentiation of pancreatic cancer stem cells (PCSCs) contribute to major challenges with current PC therapies, causing metastasis and therapeutic resistance, leading to recurrence and death in patients. The concept that PCSCs are characterized by their high plasticity and self-renewal capacities is central to this review. We focused specifically on the regulation of PCSCs, such as stemness-related signaling pathways, stimuli in tumor cells and the tumor microenvironment (TME), as well as the development of innovative stemness-targeted therapies. Understanding the biological behavior of PCSCs with plasticity and the molecular mechanisms regulating PC stemness will help to identify new treatment strategies to treat this horrible disease.

Introduction & background

Pancreatic cancer (PC) represents a major cause of cancer-related death worldwide and differs from other cancers, the incidence rate of PC patients has continued to increase over the past few years, with little improvement in survival rates [1]. Alarmingly, PC has a poor 5-year survival rate, less than 11% in the US [2], and it has been projected that the mortality of PC will become the second leading cause of cancer-related deaths in the US by 2030 [3].

Many tumor entities have made considerable advances in the diagnosis and treatment during the past decade; however, this is not the case for PC. From a clinical point of view, the comparatively low success rate of therapy for PC compared with other cancers is attributable to the deep location of the pancreas causing a lack of appropriate screening and diagnostic modalities and challenges in performing a tissue biopsy, aggressive clinical course, and low response rate of PC to chemo- and radiotherapy [4]. From the characteristics of the PC itself, in addition to the highly heterogeneous tumor immunosuppressive microenvironment, PC stem cells (PCSCs) are also functionally important in tumor progression and therapeutic resistance [5]. Tumors are comprised of a limited number of distinct cells known as cancer stem cells (CSCs), also referred to as tumor-initiating cells (TICs). They possess the ability of tumorigenesis reconstitution with unlimited proliferative potential and inherently higher chemo- and radioresistance, have increased metastatic and invasive potential and show higher disease recurrence compared with their differentiated cancer cell counterparts [5, 6]. Therefore, a deeper understanding of CSCs is necessary for the improved management of cancer patients.

The presence of CSCs was first demonstrated in acute myelogenous leukemia in 1994 [7, 8] and subsequently confirmed in breast [9] and brain tumors [10]. Three different studies using mouse models of the brain, skin, and intestine gave the first convincing data to demonstrate the involvement of CSCs in malignancies progression [11,12,13]. PCSCs were first identified in 2007 and represent less than 1% of all PC cells [14]. These cells were first discovered as CD44 + CD24 + ESA + cells with the ability to develop tumors at a significantly higher frequency than the bulk tumor [14]. However, later studies have shown that PCSCs can express multiple markers, including CD9, CD24, CD34, CD44, CD133, ABCB1, ABCG2, ALDH1, CXCR4, DCLK-1, ESA, EZH2, GLRX3, NANOG, OCT4, SOX2, NOTCH-1, c-MET, LGR5, alpha6beta4, tetraspanin-8 and nestin [15,16,17,18,19,20]. The expression of these factors reprograms cells to CSCs and promotes plasticity, thereby allowing tumor cells to adapt to changes in their environment and survive. Furthermore, the interactions and connections among CSC markers in PC are quite complicated. Specifically, these markers are poor prognostic indicators linked to tumor clinical progression and recurrence. CSCs promote tumorigenesis, chemical resistance and metastasis. Theoretically, eliminating CSCs may be a promising approach for PC treatment. However, an increasing number of studies have shown that CSCs exhibit strong plasticity and this plasticity allows them to be successfully adapted to targeted therapies [21, 22]. In 2008, Patrick C. Hermann et al. first proposed that PCSCs are in a plastic state rather than a hardwired defined state [23]. In general, the plasticity of CSCs can be defined as the ability of cells to differentiate across lineages and hierarchies and refers to the ability of cancer cells to generate more differentiated bulk tumor cells, as well as cells’ phenotypic potential——the capacity of cells to adopt a new identity or fate in response to changing circumstances and environmental factors, leading to increased tumor heterogeneity and promoting tumor progression [23, 24].

Herein, we summarize current knowledge of PCSCs from an oncology perspective, discuss developments in the field of PCSCs and, more importantly, focus on elucidating that PCSCs exist as a plastic state, influenced by multiple factors inside and outside the tumor cell, influenced by multiple factors inside and outside the tumor cell and, highlight the role of PCSCs in contributing to the malignant behavior of tumor and their potential clinical applications, which provide a comprehensive understanding of the plasticity of PCSCs and its roles in cancer progression. Figure 1 shows the milestones and discovery timeline related to PCSCs.

Fig. 1
figure 1

Early studies using FACs and xenotransplantation techniques identified markers of pancreatic cancer stem cells, which laid an important foundation for further research. Following the identification of these markers, a great deal of work has been devoted to exploring the intrinsic and extrinsic regulators of pancreatic cancer stemness

Classical markers and signaling pathways of PCSCs

Markers

Early research on PCSCs mainly focused on identifying their markers through flow cytometry and xenotransplantation assays in immunocompromised mice. Although it is not completely convincing to rely only on markers to identify PCSCs, these early studies have laid a solid foundation for later research on PC stemness. These markers do not exist independently, instead, interacting with each other and with stemness-related pathways to promote tumor progression. The triplet combination of CD24 + CD44 + ESA + and the binary CD133 + CXCR4 + combination represent the earliest identified PCSC surface markers [25, 26]. In addition to classical cell surface markers, side population and drug efflux markers, as well as intracellular markers have also attracted researchers' attention. High expression of ATP-binding cassette (ABC) transporters was found on the plasma membrane of many CSCs, which is responsible for transporting small molecules from the cytoplasm to the extracellular using the energy generated by ATP hydrolysis. It has been proved that the expression of ABCB1 and ABCG2 in PCSCs is increased [27]. The increase of ABC transporters in CSCs enhances their ability to excrete dyes and drugs. With this characteristic, PCSCs in large tumor samples can be identified by Hoechst 33,342 and flow cytometry. In addition, the enhanced efflux ability of drugs also increases chemotherapy resistance. Different ABC transporters exhibit different efflux abilities for different compounds. For example, ABCG2 mediates resistance to 5-flurorouracil and irinotecan—chemotherapeutic agents for PC [28]. PCSC markers, the associated pathways and their effect on tumor progression are summarized in Table 1.

Table 1 Pancreatic cancer stem cell markers, the associated pathways and their effect on tumor progression

Signaling pathways and targeted therapy

Major signaling pathways

Developmental pathways including Sonic Hedgehog (SHH), NOTCH and WNT signaling are the most activated pathways in PC cells, which have been experimentally demonstrated to be mechanistically connected with the cancer stemness features of PC and promote PC invasion, metastasis, and drug resistance [29]. Embryonic development and stemness regulation are two fundamental mechanisms regulated by the SHH signaling pathway [30]. SHH signaling usually ceases after embryogenesis; however, the signaling pathway is reactivated during the initial progression phase of PC [31]. Additionally, studies based on RNA sequencing data suggested that compared with pancreatic ductal epithelial cells and normal pancreatic stemness, SHH and other SHH components are significantly overexpressed in CD44 + CD24 + ESA + cells, further supporting the key role of SHH in PCSCs [14]. Moreover, signal transduction of the NOTCH signaling pathway is independent of the second messenger and only occurs between cells that are in contact with each other [32]. NOTCH is activated when NOTCH receptors bind to NOTCH ligands of adjacent cells, which transmits the signals from the neighboring cell to the nucleus, starting the expression of downstream transcription factors. There are four different types of NOTCH receptors (NOTCH-1, NOTCH-2, NOTCH-3, and NOTCH-4) [33] and five kinds of NOTCH ligands (DLL-1, DLL-3, DLL-4, Jagged-1, and Jagged-2) [34]. Upregulation of several NOTCH pathway components in PCSCs has been demonstrated previously. For instance, the overexpression of Hes1 promotes PCSC self-renewal and tumorigenicity [35], and NOTCH promotes apoptotic resistance in PCSCs potentially through activation of the nuclear factor of NF-κB [36].In addition, in the TME, NOTCH signaling cascades interact with fibroblast growth factor and WNT signaling cascades to maintain cancer stemness and reshape TME [37]. WNT signaling is also essential for the maintenance of cancer stemness [38]. Aberrant activation of the canonical WNT/β-catenin signaling pathway facilitates cancer stemness renewal, thus playing vital roles in tumorigenesis and the therapeutic response of a wide range of malignancies, including PC [39]. Figures 2 and 3 shows the specific regulatory mechanisms of the three classic pathways on PCSCs.

Fig. 2
figure 2

In the absence of ligands, the SHH signaling pathway is inactive. When hedgehog ligands activate the SHH pathway, these hedgehog ligands bind to PTCH and relieve the repressive effects on SMO, a seven-transmembrane protein, resulting in the translocation of the full length activated forms of GLI (GLIA) into the nucleus and activation of the expression of target genes, including PTCH and GLI themselves and many other cancer stemness-related genes. When the receptor activates the NOTCH pathway by binding with its ligand, the protein of the receptor is cleaved by a disintegrin and metalloproteinases (ADAMs) that mediate the cleavage of the NOTCH extracellular domain and γ-secretase, which mediates the cleavage of the NOTCH intracellular domain (NICD), which is then released. NICD is further translocated to the nucleus as a transcriptional effector and displaces the corepressor protein from CSL (CBF1, Suppressor of Hairless, LAG1)/RBPJ transcription factors, which leads to the activation of downstream signaling cascades to regulate target genes, including Hes1, Hey1, cyclinD1, c-myc, p21/Waf1 and nuclear factor-κB(NF- κB). WNT ligands are responsible for activating the WNT/β-catenin signaling pathway. In the absence of WNT ligands, β-catenin is phosphorylated by the destruction complex of β-catenin, which is a tertiary complex formed by the scaffolding proteins Axin and adenomatous polyposis coli (APC), the kinase proteins glycogen synthase kinase-3β (GSK-3β) and casein kinase 1α (Ck1α). This leads to β-catenin degradation via β-TrCP200 ubiquitination; therefore, β-catenin is failed to translocate from the cytoplasm to the nucleus. Contrastingly, in the presence of WNT ligands, the ligands bind to the FZD and the LRP receptor complex. Then the cytoplasmic domain of the LRP receptor is phosphorylated by GSK3β and CK1α, and the scaffolding protein Disheveled (Dvl) is recruited. The unphosphorylated β-catenin is released from the complex and enters the nucleus. In the cytoplasm, TAZ/YAP directly interacts with β-catenin and restricts its degradation. In the nucleus, β-catenin binds to TCF/LEF and enhances the recruitment of histone-modifying coactivators, such as BCL9, CBP/p300, and BRG1, to induce the transcription of WNT target genes. Many of these genes encode gene products capable of broadly upregulating cancer cell stemness, including CCND1, AXIN2, and the oncogene MYC

Fig. 3
figure 3

The extracellular matrix, cancer-associated fibroblasts, and immune cells are critical components in the regulation of pancreatic cancer stemness, influencing pancreatic cancer stemness-related pathways and stemness molecules through secretory factors, extracellular matrix and intercellular interactions

In addition, JAK/STAT3, TGF-β, PI3K/Akt/mTOR, and Hippo signaling are also involved in the maintenance of PC stemness [40,41,42,43,44]. Together, these signaling pathways interact with each other and with other oncogenic signaling pathways, which provides evidence for the molecular mechanism of the PC stemness and suggests a potential approach for targeting the cancer stemness in patients with PC.

Targeting stemness-related pathways in PC

A promising method for targeting PCSCs is to inhibit the developmental pathways, including SHH, NOTCH, and WNT pathways, which play significant roles in maintaining and promoting PC progression by regulating PC stemness signaling [45,46,47].

Both SMO and the GLI family of zinc finger transcription factors in the SHH signaling pathway are regarded as important targets for cancer therapy. Cyclopamine was the first SMO inhibitor to be discovered and it can induce gemcitabine sensitivity [48, 49]. Sonidegib is a highly effective SMO inhibitor, and it has been utilized as an SHH pathway antagonist, acting by binding to SMO and inhibiting the activation of downstream hedgehog target genes [50]. α‐Mangostin has been shown to inhibit the expression of stemness-related genes CD24, CD44, CD133, NANOG, OCT4, c‐Myc, SOX2, and KLF4 by inhibiting the GLI transcription, indicating that it can regulate cancer growth by inhibiting cancer stemness population [51]. Another GLI transcription factor inhibitor GANT-61 is able to block DNA binding of GLI and decreases transcriptional activity of pluripotency-promoting factors in PCSCs, thereby reducing cancer cell growth and proliferation [52, 53].

NOTCH signaling is triggered by γ-secretase-mediated cleavage of the NOTCH receptor, and thus γ-secretase is a central player in the NOTCH signaling pathway. Studies have shown that γ- secretase inhibitors (GSI), such as MK0752, PF-03084014, and MRK-003 induce cancer cell apoptosis and interfere with cancer cell proliferation and invasion in several human cancers including PC [54,55,56,57,58]. In conclusion, GSI has exhibited antitumor effects in human cancer in many preclinical models; however, GSI shows a variety of side effects, such as goblet cell metaplasia of the small intestine and diarrhea [59]. Some studies have used less toxic alternative therapies, such as quinomycin, to avoid the limitations of GSIs [60]. Some natural compounds inhibiting the NOTCH signaling pathway that are non-toxic to human cells have also been identified, such as genistein, curcumin (diferuloylmethane), sulforaphane, quercetin and cimigenoside [61,62,63,64,65,66], which are expected to become therapeutic agents targeting PCSCs.

Besides, there is substantial evidence that targeting WNT/β-catenin signaling pathway enhances the sensitivity of PC to chemotherapeutic agents [67]. As previously mentioned, the WNT pathway cannot be activated in the absence of WNT ligands. WNT ligands can be palmitoylated by Porcupine (PORCN), a membrane-bound member of the o-acyltransferase family of proteins, allowing them to secret and initiate cellular reactions [68]. Several inhibitors that target PORCN prevent WNT ligand proteins from being palmitoylated in the endoplasmic reticulum, which restricts their secretion subsequently. Thus, an effective treatment strategy is to abolish WNT secretion by blocking its acylation with a PORCN inhibitor. The small molecule inhibitor WNT974 (LGK974), which is accessible orally, inhibits tumor development in vivo and decreases the viability of epithelial ovarian cancer (EOC) cells in vitro. In EOC preclinical mouse models, WNT974 exhibits improved anticancer activity in combination with paclitaxel. Currently, there is a phase I clinical trial evaluating WNT974 monotherapy for patients with PC (NCT01351103). Vantictumab (OMP-18R5), a monoclonal antibody, can specifically target FZD. Further, OMP-18R5 inhibits tumor growth in xenograft mouse models of PC and many other malignancies and is now being investigated in a phase I trial for PC (NCT02005315) [69]. There are also some agents that target the β-catenin-destruction complex. For example, an existing FDA-approved medicine, pyrvinium, can bind all CK1 family members in vitro, and selectively enhance CK1α kinase activity. By decreasing β-catenin levels and blocking the transcription of β-catenin targeted genes, pyrvinium inhibits the WNT signal. Pyrvinium reduces the development of platinum-resistant tumor and promotes apoptosis in vitro and in vivo, and when combined with paclitaxel, these effects are strengthened. However, pyrvinium no longer has an effect on cancer cells with rising levels of β-catenin [70]. A phase 1 clinical trial investigating pyrvinium for PC that cannot be removed surgically is underway (NCT05055323). In addition, many potential compounds targeting PCSCs through inhibiting the WNT/β-catenin signaling pathway have been investigated in preclinical evaluations. For example, a preclinical evaluation revealed that FH535 inhibited β-catenin transcriptional activity [71] and suppressed the expression of stemness markers CD24 and CD44 [72]. Salinomycin showed a significant inhibitory effect on increasing the cytotoxic effects of traditional therapy of gemcitabine in PCSCs [73] and inhibiting tumor cell growth and migration by interfering with LPR phosphorylation and inducing its degradation in the xenograft model in vivo [74]. Besides, some studies have also demonstrated that some natural dietary compounds, including curcumin, sulforaphane, genistein, lycopene, and piperine, can inhibit the WNT/β-catenin signaling pathway, thus inhibiting cancer stemness [75, 76] A summary of drugs targeting these pathways is provided in Table 2.

Table 2 Summary of drugs targeting pancreatic cancer stemness related signaling pathways

Intracellular elements of tumor cells participate in the regulation of PCSC plasticity.

Many intrinsic regulators regulating PC stemness are converge into the above-mentioned stemness-related signaling pathways (WNT/β-catenin, NOTCH, SHH, JAK/STAT3, TGF-β, and Hippo). In addition, there are still many independent factors involved in the regulation of PC stemness. These factors can be categorized into several subclasses as listed in Table 3.

Table 3 Classification of intracellular elements of tumor cells that influence pancreatic cancer stemness plasticity

Transcription factors

In recent years, an increasing number of transcription factors that bind to the promoters of genes regulating PC stemness have been identified. RNA polymerase II-associated factor 1 (PAF1) promotes the expression of the stemness-associated genes CD44, NANOG, ABCG2, and ALDH1 [19]. ETS-homologous factor (EHF) binds to the promoter of CXCR4, thereby obstructing its transcription and resulting in altered crosstalk between PC cells and pancreatic stellate cells (PSCs) [77]. Snail family transcriptional repressor 2 (SNAI2) has been proved to promote the expression of CD44, while SNAI2 gene knockout significantly reduced the number of PCSCs, thus reducing the tumorigenicity and chemotherapy resistance of PC [78].

Epigenetic regulators

Fat mass and obesity-associated protein (FTO) is an RNA N6-methyladenosine demethylase. FTO depletion was shown to inhibit the spheroid formation in PC cells [79], and the absence of FTO in vitro significantly reduced the mRNA and protein expression of PCSC markers including CD44, ALDH1, SOX2, NANOG, and CD133 [79]. Overall, these results suggest that FTO is essential for spheroid formation, the maintenance of stemness marker expression, and the self-renewal potential of PC. Sirtuin 1 (SIRT1) and cullin 4B-ring E3 ligase (CRL4B) interact and cooperate as a functional unit, contributing to the epigenetic silencing of tumor suppressors, and playing an important role in regulating PCSC properties [80]. Methyl CpG binding domain 3 (MBD3) protein exhibits oncogenic effects in PC. MBD3 was proved to increase stemness markers level of OCT4, NANOG and SOX2 [81]. In addition, MBD3 binds to YAP to significantly inhibit stemness maintenance in PC cells via Hippo signaling. Ubiquitin carboxyl-terminal hydrolase isozyme L5 (UCHL5) directly deubiquitinates and stabilizes ELK3 protein to activate NOTCH-1 expression and signaling, enhancing self-renewal during PC development [82]. O-glycosyltransferases GALNT3 and B3GNT3 can promote the self-renewal of PCSCs [83]. Epigenetic regulation mediated by polycomb group (PcG) proteins, such as EZH2 and BMI-1, is also a major driver in PCSC pathogenesis [84, 85].

Metabolic regulators

There is growing evidence that CSC metabolism has unique characteristics. The maintenance of the PCSC phenotype is mainly related to the mitochondrial regulation of redox homeostasis and energy metabolism. Firstly, mitochondrial fusion and fission represent the main events involved in mitochondrial dynamics, and both processes are mainly controlled by different members of the dynamin family, together with several bridging proteins [86, 87]. Optic atrophy 1 (OPA1) regulates mitochondrial function and stabilizes the respiratory chain supercomplex by participating in the formation of mitochondrial cristae junctions and driving mitochondrial fusion, controlling mitochondrial respiratory activity and thereby promoting PC stemness [88]. Mitofusin-1 and mitofusin-2 are also involved in mitochondrial fusion, but the specific mechanism of their role in PCSCs has not been elucidated [86]. Besides, emerging evidence indicates that mitochondrial fission enhances uncoupled respiration to avoid excessive ROS production, thereby preventing oxidative damage. Maintaining low mitochondrial ROS levels is essential for maintaining PCSC self-renewal and function. The GTPase of the kinesin superfamily of proteins, dynamin-related protein 1 (DRP1), is the primary enforcer of mitochondrial fission [89], and PCSCs exhibit increased DRP1 expression, which is positively correlated with the expression of PC stemness-related genes such as NANOG, OCT4 and SOX2 [86]. Inhibition of mitochondrial division by inhibition of DRP1 induces the accumulation of dysfunctional mitochondria, limiting the ability of PCSCs to activate alternative pathways of energy production [86]. In addition, peroxisome proliferator-activated receptor gamma co-activator 1 (PGC-1) is the pivotal regulator of mitochondrial activity. MYC binds to the PGC-1α promoter to directly inhibit PGC-1α, thereby suppressing mitochondrial respiration and reducing PC stemness. When MYC is inhibited, the subsequent increase in PGC-1α is critical for oxidative phosphorylation in PCSCs [90]. Liver kinase B1 (LKB1) is highly expressed in CD44 + PC cells [91]. It promotes the expression of PGC-1β, which further promotes the expression of pyruvate dehydrogenase (PDH), a key enzyme linking glycolysis and the tricarboxylic acid (TCA) cycle, as well as increasing the rate of mitochondrial fusion, thereby promoting PC stemness [91, 92]. Nutrient-deprivation autophagy factor-1 (NAF-1) is highly expressed in the endoplasmic reticulum and outer mitochondrial membrane of PCSCs, and is involved in maintaining mitochondrial homeostasis to promote mitochondrial respiration, thus promoting PC stemness [93].

Signaling pathway regulators

In addition to ligands of the stemness-related signaling pathways in PC, some intracellular and membrane proteins are also involved in the regulation of these pathways. Tetraspanin-8 expression enhances SHH signaling [94]. Tetraspanin-8 directly interacts with PTCH, and ATXN3 is subsequently recruited into the SHH-PTCH complex, which reduce ubiquitination of PTCH and inhibits the degradation of SHH-PTCH complex mediated by proteasome. Stable SHH and PTCH promotes the binding of GRK2 protein kinase to SMO, allowing GRK2 to enhance SMO phosphorylation, relieving the repressive effects of PTCH on SMO, and leading to the activation of GLI and subsequent downstream gene expression [94]. In addition, family with sequence similarity 83 member A (FAM83A) promotes the activation of WNT/β-catenin signaling [95, 96]. In the nucleus, FAM83A binds to TCF, which in turn promotes the transcription of WNT target genes [96]. Meanwhile, FAM83A tyrosine 138 phosphorylation promoted β-catenin binding to TCF, inhibited TCF recruitment to histone deacetylases and enhanced WNT/β-catenin-mediated transcriptional and oncogenic effects [96]. In the cytoplasm, the DUF1669 structural domain of FAM83A mediated the interaction between FAM83A and AXIN1, GSK3β, and β-catenin, which in turn inhibited the phosphorylation and degradation of β-catenin protein [96]. In vivo experiments further demonstrated that FAM83A overexpression enhances tumor-initiating capacity [95]. Retention in endoplasmic reticulum 1 (RER1) has been previously shown to promote the activation of the NOTCH signaling pathway by increasing the activity of the γ-secretase complex in the brain [97]. In PC, in vitro experiments have demonstrated that RER1 promotes stemness in a hypoxic environment, including enhancing tumorsphere formation ability and stemness markers expression such as CD133, SOX2, BMI-1, Lin28, and NANOG, but the specific effect of RER1 on NOTCH signaling in PC still needs to be further explored [98].

Noncoding RNAs

Noncoding RNAs (ncRNAs) represent a class of RNA molecules that do not encode proteins, including from long noncoding RNAs (lncRNAs) with more than 200 nucleotides to piwi-interacting RNAs (piRNAs) with only 20 nucleotides in terms of length [99]. This type of RNA can regulate the expression of protein-coding genes; therefore, it is essential to control cell function and identity, and is related to many pathological diseases, particularly cancer [100]. Recently, ncRNAs have been shown to be involved in the regulation of stemness in different types of cancers, including PC [101].

MicroRNAs (miRNAs) regulate gene expression and maintain cell homeostasis via recognizing cognate sequences and interfering with transcriptional, translational, or epigenetic processes, and their dysregulation is associated with the regulation of PC stemness features [102, 103]. Several PC stemness factors, including NANOG, SOX2, OCT4, and ALDH1, are critical for the maintenance of PC stemness pluripotency, and miRNAs control their expression. Different types of miRNAs play different roles in regulating PC stemness. Here, we divide the regulation modes of these miRNAs on PC stemness into two categories—the enrichment of oncomiRNAs that promote PC stemness and the down-regulation of tumor suppressor miRNAs. OncomiRNAs including miR-10b [104], miR-17-5p [105], miR-21 [106, 107], miR-27a [108], miR-221 [107, 109], miR-338-5p [110], miR-520 h [111], and miR-1246 [112] have been documented. Many of these miRNA dysregulations converge on regulating stemness-related signaling pathways and the expression of cancer-relating genes. For example, miR-10b facilitates EGF-TGF-β cross-talk and enhances the expression of EMT-promoting genes, whereas decreasing the expression of several metastasis-suppressing genes [104]. MiR-17-5p can reduce the expression of tumor suppressor gene PTEN [105]. MiR-338-5p is involved in promoting WNT/β-catenin signaling patwhway [110]. Tumor suppressor miRNAs that have been reported include miR-34a [113], miR-101 [114], miR-145 [115], miR-146a [116], miR-146b-3p [117], miR-183 [118],miR-200a/c [118], miR-203 [118], miR-429 [119]. The miR-34 and miR-200 families are two main tumor-suppressive miRNA families related to regulating cancer stemness. The miR-34 family can inhibit the expression of stemness-related pathways such as NOTCH, WNT/β-catenin, TGF/SMAD and EMT-related genes such as Snail, Slug, and ZEB [120]. CD44 + and CD133 + PCSC subsets show a decrease in miR-34a expression. Restoring miR-34a expression inhibits CD44 and CD133 expression in vitro and suppresses tumor formation in vivo. In addition, miR-34a sensitizes PC cells to 5-fluorouracil (5-FU), docetaxel, and gemcitabine treatment by inhibiting NOTCH signaling [121, 122]. The miR-200 family comprises five members, including miR-200a, miR-200b, miR-200c, miR-141, and miR-429. MiR-200a overexpression was reported to reduce the expression of CD24, CD44, and ESA [123]. It was also reported that miR-200c overexpression decreases colony formation, invasion, and chemoresistance of PCSCs [124]. Treatment of PCSCs with metformin could induce the re-expression of miR-200c which is frequently lost in PC and reduce the expression of the PC stemness factors CD44, EpCAM, EZH2, NOTCH-1, NANOG, and OCT4 [125]. The reexpression of miR-101 was sufficient to limit the expression of EZH2 and EpCAM [114].

Furthermore, by competitively sequestering miRNAs, lncRNAs can act as competing endogenous RNAs (ceRNAs) together with miRNAs and mRNAs to form ceRNA networks, which can modulate the expression levels of their downstream stemness-related target genes. The overexpression of Growth arrest‐specific 5 (GAS5) which was identified as a tumor suppressor repressed the stemness features of PC cells through directly binding the 3’UTR of miR‐221 to repress its expression and increasing the expression of suppressor of cytokine signaling 3 [126]. LncRNA HOTAIR sequesters miR-34a, activating the JAK2/STAT3 signaling pathway to promote PC stemness [127]. LncRNA XIST modulates PC stemness by acting as a sponge of miR-429 [119]. Lnc-DYNC2H1-4 promotes PCSC phenotypes by sponging miR-145 [115].

These findings provide new insights for targeting PCSCs. A brief summary of these intracellular regulators is tabulated in Table 3.

Stimuli in the TME participate in the regulation of PCSC plasticity

A growing number of studies have shown that the complex pancreatic TME is vital in supporting stemness phenotype. The TME comprises various components. The extracellular matrix (ECM), composed of collagen, proteoglycans and glycosaminoglycans, is a major component of the TME and mediates the interaction between tumor cells and stromal cells. Cancer-associated fibroblasts (CAFs) mediate the proliferation, angiogenesis, invasion and metastasis of tumor cells. Migration and proliferation of endothelial cells lead primarily to the formation of new capillaries that support tumor progression, invasion and metastasis. Immune cells can regulate tumor activity. A classic histological feature of PC is the tumor cell-induced pro-fibrous connective tissue microenvironment, which intertwines with the ECM to provide a dense physical protective barrier for PC cells. Activation of the PSCs to an activated CAFs phenotype is accompanied by increased production of ECM components, cumulatively termed as fibrosis. In this section, we discuss the emerging knowledge about the impact of the TME on PCSCs.

ECM

ECM is a highly dynamic structural component. In PC models, abnormal collagen cross-linking generates mechanical stress and increases ECM stiffness, which can create a favorable environment for PCSC survival and thus enhance their viability.

LOXL2 promotes collagen fibril cross-linking leading to ECM remodeling, thereby promoting PC stemness [128]. CD44 interacts with ezrin in the TME to regulate actin cytoskeletal rearrangements to promote PC stemness, and small molecule inhibitors of ezrin have been shown to reduce the self-renewal capacity of PCSCs [129]. The hyaluronic acid (HA)/CD44 axis creates a suitable ecological niche for PCSC survival by increasing centrosome abnormalities and micronucleation, as evidenced by increased expression of NANOG and SOX2 [130]. HA can also bind toll-like receptor 2 and 4 to promote inflammatory gene expression and exacerbate the inflammatory response at the tumor site. Subsequently, cytokines and inflammatory mediators secreted by tumor-associated immunosuppressive cells contribute to PC stemness [131, 132]. The most plentiful ECM protein, type I collagen, is the main scaffold for CD133 + and ALDH + PCSCs and increases PCSC enrichment by activating β-integrin and FAK [133]. Type I collagen activates β-integrin, which is required for FAK activation [133, 134]. The tyrosine protein kinase FAK is recruited by the activated β1-integrin to initiate signal transduction after activation [134]. The FAK domain autophosphorylates the Tyr 397 residue in response to integrin engagement, enlisting SRC family kinases, and then phosphorylates the Y576 and Y577 residues in the catalytic domain [134]. This represents the initial and major step in FAK activation, which further promotes ALDH1 expression. ECM can regulate the PCSC niche by creating a hypoxic environment that directly activates HIF and target genes, as well as regulate the cascade of stemness factors and pathways, such as OCT4 and NOTCH, and enhance the expression of stemness markers [135, 136]. Proteoglycans combine with various cytokines and chemokines in the TME to activate various signaling pathways in PCSCs, such as SHH, WNT/β-catenin, and NOTCH. For example, glypican-4 (GPC4) is a member of proteoglycans, which enhances stem cell–like properties via promotion of WNT/β-catenin pathway and decrease the sensitivity of PC cells to 5-FU [137].

CAFs

In the pancreatic TME, cancer-associated fibroblasts (CAFs) are abundant stromal-activated fibroblast cell types that can regulate PC stemness and are functionally important in tumor development and metastasis [138]. PC cells are capable of secreting different levels of signaling molecules, such as TGF-β, SHH, IL-6, and TNF-α, and then activating CAFs [139]. Activated CAFs, in return, release growth factors, chemokines or cytokines to directly affect cancer cells. CAFs form a paracrine niche for PCSCs, wherein paracrine signaling enhances PC stemness-like properties at the tumor-stroma interface [140]. In this section, we illustrate the effect of some cytokines secreted by CAFs on PC stemness.

A recent report suggested that the osteopontin (OPN) released by CAFs was regarded as a crucial driver of PC development by upregulating the plasticity of PCSCs [141]. OPN, a multifunctional secreted integrin-binding glycoprotein, is overexpressed in numerous cancers and can be identified as a prognostic factor clinically [142, 143]. OPN in the tumor microenvironment binds to CD44 expressed on PCSC properties, which subsequently promotes clonal growth, invasion, and metastasis. These effects require CD44 to bind to the protein encoded by the oncogene TIAM1, which activates Rac1 to induce membrane cytoskeleton-mediated cell adhesion, proliferation, and migration. Previous studies have shown that ovarian cancer cells stimulated mesothelial cells to promote OPN expression and release through TGF-β signaling. OPN promoted ovarian cancer stemness and chemoresistance via PI3K/AKT signaling, CD44 receptor activation, and ABC drug efflux transporter activity [144]. In addition, the glioma perivascular niche facilitates stemness characteristics via the OPN–CD44 signaling pathway was also demonstrated [145]. CAFs promotes PC stemness through the interaction between OPN–CD44 axis and tumor cells has also been confirmed [141]. The OPN secreted by CAFs acts on the CD44 receptor of PCSCs and promotes the stemness characteristics of PC by promoting the expression of stemness markers NANOG, OCT4, and ABCG2 [141].

Furthermore, hepatocyte growth factor (HGF), which is associated with the regulation of PC stemness, is also secreted by CAFs. When HGF binds to c-MET in cancer cells, they are stimulated to produce uPA, causing more pro-HGF to become active HGF, which then binds to c-MET in PC cells. Paracrine HGF induces YAP nuclear translocation by binding c-MET, resulting in the crosstalk between CAFs and cancer cells that HGF/c-MET-mediated induces the expression of stemness pluripotency markers such as NANOG, OCT4, and SOX2 in PC cells, as well as increased self-renewal ability [146]. In addition, YAP nuclear translocation is followed by binding to HIF-1α in the nucleus and maintains the stability of HIF-1α to promote glycolysis [147]. Glycolysis is a key characteristic of both normal stemness and cancer stemness, forming a novel metabostem property [148]. Even under aerobic conditions, tumor cells favor a metabolic transition to glycolysis, and this metabolic reprogramming is crucial in the development, maintenance, and differentiation of cancer stemness and is a hallmark of cancer stemness [148]. Cancer stemness is more dependent on glycolysis for bioenergy during the metabolic shift. Increased expression and stability of HIF-1α promote this metabolic reprogramming process [149]. Therefore, HIF is critical for promoting the stemness of PC. On the one hand, HIF-1α enhances the expression of hexokinase 2 (HK2), one of the rate-limiting enzymes in the glycolytic pathway [150]. On the other hand, HIF-1 directly stimulates the expression of c-MET. HIF-1α promotes c-MET signaling by inducing c-MET gene expression in response to metabolic stress, such as nutrient deficiency or hypoxia, thereby promoting PC stemness [151].

In addition, CAFs secrete the ligands of WNT and promotes PC stemness through the WNT/β- catenin linked protein pathway [152, 153]. IL-6 secreted by CAFs upregulates the expression of PCSC genes, including SOX2, NANOG, and OCT4 by activating the JAK2/STAT3 pathway [154]. CAFs also produce SDF-1, the ligand for CXCR4, to attract CXCR4 + PCSCs, causing micrometastases [155]. Nodal/Activin secreted by CAFs acts on Alk4/7 receptors on PCSCs to promote self-renewal [156]. Thus, CAFs play an important role in propagating PC stemness phenotype.

Immune cells and extracellular vesicles

The enhanced ability of PCSCs to promote tumor development suggests that these cells have an innate advantage for immune escape. The role of immune cells against tumors is two-fold, with both tumor-promoting and tumor-suppressing activities. Evidence is accumulating that several immune cell types have an important role in regulating PCSC properties. For example, in the context of PC cells, monocytes acquire an immunosuppressive phenotype by activating STAT3 and become myeloid-derived suppressor cells, while this STAT3 activation promotes ALDH1 + stem cell frequency in PC [157]. MiR-21a-5p is upregulation in M2 macrophage-derived extracellular vesicles (EVs), which promotes NANOG and OCT4 expression and sphere-forming, colony-forming, invasion, migration, and anti-apoptosis abilities of PCSCs in vitro and tumorigenic ability in vivo [158]. Targeting tumor-infiltrating macrophages decreased the number of PCSCs, relieved immunosuppression and improved chemotherapeutic responses in PC [159]. Circ_0030167 loaded in EVs derived from bone marrow mesenchymal stem cells inhibits the stemness of PC cells by sponging miR-338-5p and further regulating the WNT/β-catenin axis [110].

Metabolic plasticity of PCSCs and the impact of epigenetic regulation on it

Besides RNAs, cytokines, morphogens and growth factors, various metabolic pathways are also involved in stemness destiny control. Metabolic pathways can also transmit changing signals in the extrinsic environment to alter intrinsic cell fate. PCSC metabolism represents a combination and balance of intrinsic metabolic demands and extrinsic metabolic alterations. In this section, we describe the metabolic plasticity of PCSCs.

PCSCs are metabolised in a different way to other PC cells. For example, non-PCSCs are highly dependent on glycolytic metabolism, whereas PCSCs are strongly dependent on the mitochondrial oxidative phosphorylation (OXPHOS) pathway. This metabolic reprogramming of PCSCs is also plastic and can be regulated by the environment in which they are located. The use of the more energetically efficient metabolic pathway—OXPHOS in the presence of sufficient oxygen results in a higher number of ATP molecules per glucose molecule. Under hypoxia or stress, these stemness can revert to a glycolytic program, even in some cases using mitochondrial fatty acid oxidation. For example, when the mitochondrial inhibitor metformin was used in PC, metformin-resistant PCSCs reversed towards a non-stemness metabolic phenotype by increasing MYC expression to enhance glycolytic capacity [90]. Thus, disruption of mitochondrial metabolic dynamics is likely to attenuate the stemness phenotype of PC. Besides, in the process of dedifferentiation of PC cells, cells increase their oxidative metabolism by promoting pyruvate to enter the TCA cycle and improving the expression levels of citrate and citrate lyase in cells. The rapid transformation of this metabolic change indicates that it is closely related to epigenetics.

Mitochondrial dynamics and metabolism are mainly controlled by post-translational modifications (PTMs) of proteins, among which ubiquitination (Ub) and Ub-like (UbL) modifiers plays a major role in the regulation of PC stemness. This is very similar to the regulatory mechanism in normal stemness, where the ubiquitination and deubiquitination activities of the stemness pathways NOTCH, WNT, and SHH proteins are precisely regulated by ubiquitinating and deubiquitinating enzymes, resulting in the reprogramming of PC stemness metabolism. Among the UbL modifiers, the expression of interferon-stimulated gene 15 (ISG15), small ubiquitin-related modifier (SUMO), neural precursor cell expressed developmentally downregulated protein 8 (NEDD8), and human leukocyte antigen-F adjacent transcript 10 (FAT10) are significantly higher in CD133 + cells than in CD133- cells. A recent study showed that ISG15 and protein ISGylation are specifically enriched in PCSCs compared with non-PCSCs. Loss of ISG15/ISGylation alters the mitochondrial state and metabolism—manifested by an increase in the number of mitochondria but a severely impaired optical character recognition, that is, the accumulation of dysfunctional mitochondria. In addition, the glycolytic capacity of PCSCs is also significantly impaired in the absence of ISG15, indicating that the overall metabolic plasticity of PCSCs (aerobic and anaerobic respiration) is affected by the loss of ISG15/ISGylation. However, the regulation of OXPHOS by SUMO, NEDD8, and FAT10 in PCSCs has not been reported yet.

Besides, glutamine is a major mitochondrial reaction substrate and is also required for the maintenance of mitochondrial membrane potential and integrity. In the CD9 + PC stemness subpopulation, CD9 increases glutamine uptake and promotes mitochondrial OXPHOS by interacting with the glutamine transporter ASCT2 [160].

PCSCs and malignant phenotype

PCSCs and EMT promote each other significantrelationship

A crucial developmental program called EMT is frequently engaged during the invasion and metastasis of cancer. It is of fundamental importance in biology that the activation of the EMT process is related to the characteristics of stemness in neoplastic cells [161, 162]. During the shifts towards the mesenchymal phenotype, which represents a more invasive and aggressive disease phenotype, levels of non-invasive epithelial cell markers, including E-cadherin, α-catenin, and γ-catenin decrease, while the expression of vimentin, metalloproteinases MMP-2, MMP-9, fibronectin, and N-cadherin which are typically expressed on invasive epithelial cells increase [163]. A group of transcription factors called EMT-activating transcription factors, including Snail, Slug, Twist1, NF-κB, ZEB1, and ZEB2 control EMT by suppressing the expression of genes that code for epithelial markers, such as E-cadherin. ZEB1 is the most crucial promoter of EMT and links EMT with stemness-maintenance in PC as shown in the K-rasLSLG12D/+; Trp53R172H/+; Pdx-1-Cre (KPC) mouse models [164]. As a transcriptional repressor, ZEB1 binds to the E-box motif in the promoter regions of downstream target genes, such as E-cadherin and members of the miR-200 family, to decrease their production. It is also demonstrated that ZEB1 and stemness marker CD44 are mutually regulated. ZEB1 suppresses the epithelial splicing regulator ESRP1 in PC to promote CD44 isoforms (CD44s) splicing, causing the expression to change from the variant CD44v to the standard CD44s subtype. CD44 contains two isoforms, each with a distinct function: standard isoform (CD44s) and variant isoforms (CD44v). CD44s has been proven to be positively related with stemness gene features, whereas CD44v exhibits an inverse association [165]. Additionally, CD44s also contributes to PC lymph node and liver metastasis and advanced TNM staging [166]. An increase in CD44s level was shown to increase the expression of ZEB1, thus forming a positive feedback loop, leading to a self-sustaining ZEB1 and CD44s expression. The feedback loop between CD44s and ZEB1 influences the ability of cancer cells, including the increase of tumorsphere initiation and metastasis ability [167]. In addition, nestin, one of the PC stemness markers is also vital in PC cell metastasis, and the administration of nestin siRNA was reported to provide a novel therapeutic strategy for PC [168]. Nestin, NANOG, Slug, and MMP2 mRNA levels decreased, and E-cadherin expression levels increased in nestin shRNA-transfected PC cells [169]. An orthotopic implantation model using mice also showed that nestin knockdown significantly reduced primary and metastatic tumor development by human PC cells [168]. Besides, the expression of PC stemness marker CD133 reportedly induces EMT via the transcription factor NF-κB [170, 171]. Compared with CD133- cells, CD133 + cells showed increased NF-κB expression [172]. Mechanically, the overexpression of CD133 increases both mRNA and protein levels of IL-1β gene expression, and then IL-1β activates NF-κB, thus driving EMT and cell invasion [173].

Therapeutic resistance

Recently, a growing number of studies have revealed that the potential etiology of therapeutic resistance is related to stemness markers of PC. Herman et al. found that CD133 + cells were more resistant to gemcitabine than CD133 − cells isolated from PC patients, and prolonged exposure resulted in the selection of CD133 + cells [26]. Furthermore, they demonstrated in a xenograft model that animals given gemcitabine experienced a reduction in tumor size but an increase in the percentage of CD133 + cells. Several research exploring the molecular interaction between therapeutic resistance and CD133 + PC cells have highlighted the critical role of its metabolic plasticity, which is related to reactive oxygen species (ROS) [174]. Low levels of ROS are critical for maintaining cancer stemness and their resistance to therapy; however, the ROS regulating mechanisms in cancer stemness remain to be explored [175]. Studies have shown that ROS production is indeed lower in CD133 + cells compared with CD133- cells. When drugs typically associated with ROS production, such as gemcitabine, 5FU, and paclitaxel, were applied to CD133 + and CD133- cells, CD133 + stemness did not exhibit any increase of ROS, while CD133- cells had enhanced ROS generation [174]. These treatments further caused CD133- cancer cells to die whereas CD133 + cells were unaffected, which seems to provide CD133 + stemness a survival advantage [174]. In addition, although gemcitabine, a cytotoxic agent, can inhibit CD133 + stemness proliferation, it has little effect on the apoptosis of cancer stemness, allowing them to return to the stemness pool upon gemcitabine withdrawal [176]. In contrast, under gemcitabine treatment, the vast majority of the tumor cells — more differentiated cells — became apoptotic [177]. It is clear that traditional therapy can only target highly differentiated tumor cells, leaving undifferentiated cancer stemness resistant to therapy.

Other stemness markers are also associated with therapeutic resistance. It is reported that CD44 + PC stemness properties show higher malignancy and stronger resistance to chemotherapy and radiotherapy than CD44- cells [178,179,180]. Studies have also shown that PC with high CD44 expression is resistant to gemcitabine. Knocking out CD44 also leads to decreased invasiveness and increased sensitivity to gemcitabine [178, 181]. The influence of CD44 + stemness on therapeutic-resistance is mainly attributed to the ABC superfamily of transporter proteins in PC [182]. The overexpression of ABC superfamily of transporter proteins in PC limits the exposure to anticancer drugs. CD44 is also associated with the increased protein expression of the ABC transporter genes MDR1 and MRP1 [183,184,185]. Verapamil, an ABC transporter inhibitor, resensitized resistant cells to gemcitabine in a dose-dependent manner, and CD44 RNA interference inhibited the clonogenic activity of resistant cells [181].

Clinical perspective—Detection and prediction of prognosis in patients with PC via stemness properties

The analysis of PC stemness in surgical tissue specimens is anticipated to discover meaningful and reliable prognostic indicators and evaluate the effectiveness of anticancer therapy. Recently, agrin, an extracellular matrix protein, was reported to be enriched in the extracellular vesicles of PC stemness and act as a marker of poor prognosis in patients with PC [186]. In hepatocellular carcinoma, agrin promotes hepatocarcinogenesis by binding to the LRP-4 receptor and activating the YAP transcription factor [187]. Researchers have also verified the existence of this pathway in PC cells, proving that agrin in cancer stemness extracellular vesicles promotes YAP activation and cancer cell proliferation and inventory. It can translocate YAP, the core participant of the Hippo pathway, to the nucleus in order to alter the transcriptional program of cells, thus promoting tumor proliferation and metastasis [186]. In addition, researchers found that circulating agrin + extracellular vesicles (EVs) can be used as specific and sensitive biomarkers of disease progression in patients with PC, who did not undergo surgery, through the ROC curve analysis, and combination with CD133 + EVs improves the accuracy of disease progression prediction [186]. In addition, the expression of cancer stemness markers such as CD24, CD44, and CD133 has been linked to decreased survival in PC. For example, CD24 is overexpressed in high-grade tumors and more advanced PC stages, and lymphatic invasion and venous invasion are observed more frequently in the CD24 + PC, suggesting its role in the progression of PC [188]. CD24 is related to the recurrence of resectable PC and is an important factor that leads to a low survival rate in patients with PC [189]. Overexpression of CD44 or CD133 is significantly associated with clinical TNM stage, tumor differentiation, lymph node metastasis, and a decreased 5-year overall survival rate [190, 191].

Discussion

PC is a highly malignant tumor with a poor prognosis. Despite advancements in the treatment, late-stage diagnosis and other reasons result in poor prognosis, recurrence, and metastasis. Cancer stemness refers to the ability of a pool of self-sustaining cells in generating differentiated cancer cells and initiating tumor growth [176]. The term cancer stemness does not denote the origin, but rather the plasticity state of cancer cells. Increasing evidence supports the idea that cancer stemness exists in a highly plastic state rather than an absolute entity [192,193,194]. Cancer stemness have the ability to self-renew and regenerate, therefore, these cells are significantly resistant to chemo- and radiotherapy.

On the one hand, investigating cancer stemness from the standpoint of fundamental science helps improve the comprehension of tumor heterogeneity. On the other hand, refining our understanding of the plasticity of PCSCs may eventually lead to a better understanding of the clinical prospects of targeting PC stemness. In particular, targeting PC stemness has potential benefits for patients with PC. PC stemness heavily contributes to therapeutic resistance [195]. Accumulating evidence suggests that the combination of chemotherapy drugs and PC stemness inhibitors is more effective than monotherapy in vitro and in vivo [53, 196]. The relationship between PC stemness and tumor malignant phenotype demonstrates a new possibility of PC treatment based on PC stemness-targeting since PC stemness promotes tumor growth and metastasis [180]. Referring to the current state of research on PC stemness, there are several aspects in oncology that deserve further study. It is obvious that targeting PC stemness should be an integral part of the entire treatment scheme. Targeting PC stemness, however, provides substantial hurdles since therapy regimens may damage normal stemness in the human body. The challenge ahead is to specifically target PC stemness without unnecessarily affecting normal stemness. Thus, identifying of cancer stemness-specific signaling networks is critical for the improvement of anti-stemness cancer therapy [197]. Moreover, some studies have shown that the metabolism of cancer stemness rapidly transit under heterogeneous environmental circumstances. Identifying specific metabolic pathways, such as hypoxia, nutrient deficiency, and the low pH of cancer stemness may also be beneficial [198]. Another aspect worth exploring further is the characterization of markers that can identify circulating PCSCs in liquid biopsies for PC diagnosis, prediction of prognosis and assessment of treatment response.

In conclusion, a better understanding of PC stemness and its plasticity may provide crucial insights into novel and effective treatments and improve the prognosis of patients with PC.

Availability of data and material

Not applicable.

Abbreviations

PC:

Pancreatic cancer

PCSCs:

Pancreatic cancer stem cells

TME:

Tumor microenvironment

CSCs:

Cancer stem cells

TICs:

Tumor-initiating cells

ABC:

ATP-binding cassette

SHH:

Sonic Hedgehog

GSI:

γ- Secretase inhibitors

PORCN:

Porcupine

EOC:

Epithelial ovarian cancer

PAF1:

RNA polymerase II-associated factor 1

EHF:

ETS-homologous factor

PSCs:

Pancreatic stellate cells

SNAI:

Snail family transcriptional repressor 2

FTO:

Fat mass and obesity-associated protein

SIRT1:

Sirtuin 1

CRL4B:

Cullin 4B-ring E3 ligase

MBD3:

Methyl CpG binding domain 3

UCHL5:

Ubiquitin carboxyl-terminal hydrolase isozyme L5

PcG:

Polycomb group

OPA1:

Optic atrophy 1

DRP1:

Dynamin-related protein 1

PGC-1:

Proliferator-activated receptor gamma co-activator 1

LKB1:

Liver kinase B1

PDH:

Pyruvate dehydrogenase

TCA:

Tricarboxylic acid

NAF-1:

Nutrient-deprivation autophagy factor-1

FAM83A:

Family with sequence similarity 83 member A

RER1:

Retention in endoplasmic reticulum 1

ncRNAs:

Noncoding RNAs

lncRNAs:

Long noncoding RNAs

piRNAs:

Piwi-interacting RNAs

miRNAs:

MicroRNAs

ceRNAs:

Competing endogenous RNAs

GAS5:

Growth arrest‐specific 5

ECM:

Extracellular matrix

CAFs:

Cancer-associated fibroblasts

HA:

Hyaluronic acid

GPC4:

Glypican-4

OPN:

Osteopontin

HGF:

Hepatocyte growth factor

HK2:

Hexokinase 2

EVs:

Extracellular vesicles

OXPHOS:

Oxidative phosphorylation

PTMs:

Post-translational modifications

Ub:

Ubiquitination

UbL:

Ub-like

ISG15:

Interferon-stimulated gene 15

SUMO:

Small ubiquitin-related modifier

NEDD8:

Neural precursor cell expressed developmentally downregulated protein 8

FAT10:

F adjacent transcript 10

ROS:

Reactive oxygen species

References

  1. Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021;18(7):493–502.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hu J-X, et al. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J Gastroenterol. 2021;27(27):4298–321.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Rahib L, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Can Res. 2014;74(11):2913–21.

    Article  CAS  Google Scholar 

  4. Klose J, Ronellenfitsch U, Kleeff J. Management problems in patients with pancreatic cancer from a surgeon’s perspective. Semin Oncol. 2021;48(1):76–83.

    Article  PubMed  Google Scholar 

  5. Zeng S, et al. Chemoresistance in Pancreatic Cancer. Int J Mol Sci. 2019;20(18):4504.

  6. Patil K, et al. The plasticity of pancreatic cancer stem cells: implications in therapeutic resistance. Cancer Metastasis Rev. 2021;40(3):691–720.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–7.

    Article  CAS  PubMed  Google Scholar 

  8. Lapidot T, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–8.

    Article  CAS  PubMed  Google Scholar 

  9. Al-Hajj M, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100(7):3983–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Singh SK, et al. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401.

    Article  CAS  PubMed  Google Scholar 

  11. Driessens G, et al. Defining the mode of tumour growth by clonal analysis. Nature. 2012;488(7412):527–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schepers AG, et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science (New York, NY). 2012;337(6095):730–5.

    Article  CAS  PubMed  Google Scholar 

  13. Chen J, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li C, et al. Identification of pancreatic cancer stem cells. Can Res. 2007;67(3):1030–7.

    Article  CAS  Google Scholar 

  15. Wei H-J, et al. Expression of CD44, CD24 and ESA in pancreatic adenocarcinoma cell lines varies with local microenvironment. Hepatobiliary & Pancreatic Diseases International : HBPD INT. 2011;10(4):428–34.

    Article  Google Scholar 

  16. Xia P, Liu D-H. Cancer stem cell markers for liver cancer and pancreatic cancer. Stem Cell Research. 2022;60: 102701.

    Article  CAS  PubMed  Google Scholar 

  17. Jo JH, et al. GLRX3, a novel cancer stem cell-related secretory biomarker of pancreatic ductal adenocarcinoma. BMC Cancer. 2021;21(1):1241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gzil A, et al. Markers of pancreatic cancer stem cells and their clinical and therapeutic implications. Mol Biol Rep. 2019;46(6):6629–45.

    Article  CAS  PubMed  Google Scholar 

  19. Karmakar S, et al. RNA Polymerase II-Associated Factor 1 Regulates Stem Cell Features of Pancreatic Cancer Cells, Independently of the PAF1 Complex, via Interactions With PHF5A and DDX3. Gastroenterology. 2020;159(5):1898–915.e6.

  20. Ohike N, et al. Immunohistochemical analysis of nestin and c-kit and their significance in pancreatic tumors. Pathol Int. 2007;57(9):589–93.

    Article  CAS  PubMed  Google Scholar 

  21. de Sousa e Melo F, et al. A distinct role for Lgr5 stem cells in primary and metastatic colon cancer. Nature. 2017;543(7647):676–80.

    Article  CAS  PubMed  Google Scholar 

  22. Shimokawa M, et al. Visualization and targeting of LGR5 human colon cancer stem cells. Nature. 2017;545(7653):187–92.

    Article  CAS  PubMed  Google Scholar 

  23. Hermann PC, Sainz B. Pancreatic cancer stem cells: A state or an entity? Semin Cancer Biol. 2018;53:223–31.

    Article  CAS  PubMed  Google Scholar 

  24. Chiodi I, Mondello C. Life style factors, tumor cell plasticity and cancer stem cells. Mutat Res, Rev Mutat Res. 2020;784: 108308.

    Article  CAS  PubMed  Google Scholar 

  25. Li C, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67(3):1030–7.

    Article  CAS  PubMed  Google Scholar 

  26. Hermann PC, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1(3):313–23.

    Article  CAS  PubMed  Google Scholar 

  27. Zhou J, et al. Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J Gastroenterol. 2008;14(6):925–30.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kim EJ, et al. NRF2 Knockdown Resensitizes 5-Fluorouracil-Resistant Pancreatic Cancer Cells by Suppressing HO-1 and ABCG2 Expression. Int J Mol Sci. 2020;21(13):4646.

  29. Wang Z, et al. Targeting notch to eradicate pancreatic cancer stem cells for cancer therapy. Anticancer Res. 2011;31(4):1105–13.

    CAS  PubMed  Google Scholar 

  30. Gu D, Schlotman KE, Xie J. Deciphering the role of hedgehog signaling in pancreatic cancer. J Biomed Res. 2016;30(5):353–60.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Quatannens D, et al. Targeting hedgehog signaling in pancreatic ductal adenocarcinoma. Pharmacol Ther. 2022;236: 108107.

    Article  CAS  PubMed  Google Scholar 

  32. Li L, et al. Notch signaling pathway networks in cancer metastasis: a new target for cancer therapy. Medical Oncology (Northwood, London, England). 2017;34(10):180.

    Article  CAS  PubMed  Google Scholar 

  33. Mumm JS, Kopan R. Notch signaling: from the outside in. Dev Biol. 2000;228(2):151–65.

    Article  CAS  PubMed  Google Scholar 

  34. Lai EC. Notch signaling: control of cell communication and cell fate. Development (Cambridge, England). 2004;131(5):965–73.

    Article  CAS  PubMed  Google Scholar 

  35. Wang HC, et al. “Advances in pancreatic cancer stem cells, tumor- associated macrophages, and their interplay.” Cancer Cell Microenvironment 1. 2014. n. pag.

  36. Baker A, et al. Notch-1-PTEN-ERK1/2 signaling axis promotes HER2+ breast cancer cell proliferation and stem cell survival. Oncogene. 2018;37(33):4489–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Katoh M, Katoh M. Precision medicine for human cancers with Notch signaling dysregulation (Review). Int J Mol Med. 2020;45(2):279–97.

    CAS  PubMed  Google Scholar 

  38. Javed Z, et al. Wnt Signaling: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma. Asian Pac J Cancer Prev: APJCP. 2019;20(4):995–1003.

  39. Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol. 2020;13(1):165.

    Article  PubMed  PubMed Central  Google Scholar 

  40. He W, et al. IL22RA1/STAT3 Signaling Promotes Stemness and Tumorigenicity in Pancreatic Cancer. Can Res. 2018;78(12):3293–305.

    Article  CAS  Google Scholar 

  41. Datta J, et al. Combined MEK and STAT3 Inhibition Uncovers Stromal Plasticity by Enriching for Cancer-Associated Fibroblasts With Mesenchymal Stem Cell-Like Features to Overcome Immunotherapy Resistance in Pancreatic Cancer. Gastroenterology. 2022;163(6):1593–612.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang Z, Xu Y. FZD7 accelerates hepatic metastases in pancreatic cancer by strengthening EMT and stemness associated with TGF-β/SMAD3 signaling. Mol Med. 2022;28(1):82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Singh BN, et al. Rottlerin induces autophagy which leads to apoptotic cell death through inhibition of PI3K/Akt/mTOR pathway in human pancreatic cancer stem cells. Biochem Pharmacol. 2012;84(9):1154–63.

    Article  CAS  PubMed  Google Scholar 

  44. Wang C, Yin W, Liu H. MicroRNA-10a promotes epithelial-to-mesenchymal transition and stemness maintenance of pancreatic cancer stem cells via upregulating the Hippo signaling pathway through WWC2 inhibition. J Cell Biochem. 2020;121(11):4505–21.

    Article  CAS  PubMed  Google Scholar 

  45. Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene. 2009;28(40):3513–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Carballo GB, et al. A highlight on Sonic hedgehog pathway. Cell Commun Signal. 2018;16(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Jimeno A, et al. A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Mol Cancer Ther. 2009;8(2):310–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen JK, et al. Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev. 2002;16(21):2743–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Huang FT, et al. Inhibition of hedgehog signaling depresses self-renewal of pancreatic cancer stem cells and reverses chemoresistance. Int J Oncol. 2012;41(5):1707–14.

    Article  CAS  PubMed  Google Scholar 

  50. Li Y, Song Q, Day BW. Phase I and phase II sonidegib and vismodegib clinical trials for the treatment of paediatric and adult MB patients: a systemic review and meta-analysis. Acta Neuropathol Commun. 2019;7(1):123.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ma Y, et al. Inhibition of pancreatic cancer stem cell characteristics by α-Mangostin: Molecular mechanisms involving Sonic hedgehog and Nanog. J Cell Mol Med. 2019;23(4):2719–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lauth M, et al. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc Natl Acad Sci USA. 2007;104(20):8455–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Fu J, et al. GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft. Cancer Lett. 2013;330(1):22–32.

    Article  CAS  PubMed  Google Scholar 

  54. Plentz R, et al. Inhibition of gamma-secretase activity inhibits tumor progression in a mouse model of pancreatic ductal adenocarcinoma. Gastroenterology. 2009;136(5):1741-9.e6.

    Article  CAS  PubMed  Google Scholar 

  55. Du X, et al. Suppressive effects of gamma-secretase inhibitor DAPT on the proliferation of pancreatic cancer cells. Sichuan Da Xue Xue Bao. Yi Xue Ban = Journal of Sichuan University. Medical Science Edition. 2013;44(5):699–702.

    CAS  Google Scholar 

  56. Mizuma M, et al. The gamma secretase inhibitor MRK-003 attenuates pancreatic cancer growth in preclinical models. Mol Cancer Ther. 2012;11(9):1999–2009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yabuuchi S, et al. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013;335(1):41–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cook N, et al. A phase I trial of the γ-secretase inhibitor MK-0752 in combination with gemcitabine in patients with pancreatic ductal adenocarcinoma. Br J Cancer. 2018;118(6):793–801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Espinoza I, Miele L.  Notch inhibitors for cancer treatment. Pharmacol Ther. 2013;139(2):95–110.

  60. Ponnurangam S, et al. Quinomycin A targets Notch signaling pathway in pancreatic cancer stem cells. Oncotarget. 2016;7(3):3217–32.

    Article  PubMed  Google Scholar 

  61. Momtazi AA, Sahebkar A. Difluorinated Curcumin: A Promising Curcumin Analogue with Improved Anti-Tumor Activity and Pharmacokinetic Profile. Curr Pharm Des. 2016;22(28):4386–97.

    Article  CAS  PubMed  Google Scholar 

  62. Pham N-A, et al. The dietary isothiocyanate sulforaphane targets pathways of apoptosis, cell cycle arrest, and oxidative stress in human pancreatic cancer cells and inhibits tumor growth in severe combined immunodeficient mice. Mol Cancer Ther. 2004;3(10):1239–48.

    Article  CAS  PubMed  Google Scholar 

  63. Xia J, et al. Genistein inhibits cell growth and induces apoptosis through up-regulation of miR-34a in pancreatic cancer cells. Curr Drug Targets. 2012;13(14):1750–6.

    Article  CAS  PubMed  Google Scholar 

  64. Bao B, et al. Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett. 2011;307(1):26–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Nwaeburu CC, et al. Quercetin-induced miR-200b-3p regulates the mode of self-renewing divisions in pancreatic cancer. Mol Cancer. 2017;16(1):23.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Jia H, et al. Cimigenoside functions as a novel γ-secretase inhibitor and inhibits the proliferation or metastasis of human breast cancer cells by γ-secretase/Notch axis. Pharmacol Res. 2021;169: 105686.

    Article  CAS  PubMed  Google Scholar 

  67. Curtin JC, Lorenzi MV. Drug discovery approaches to target Wnt signaling in cancer stem cells. Oncotarget. 2010;1(7):563–77.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Yu J, et al. Structural model of human PORCN illuminates disease-associated variants and drug-binding sites. J Cell Sci. 2021;134(24):jcs259383.

  69. Flanagan DJ, et al. Is Required for Wnt Signaling in Gastric Tumors with and Without Mutations. Can Res. 2019;79(5):970–81.

    Article  CAS  Google Scholar 

  70. Wei W, et al. Targeting Wnt/β-catenin by anthelmintic drug niclosamide overcomes paclitaxel resistance in esophageal cancer. Fundam Clin Pharmacol. 2021;35(1):165–73.

    Article  CAS  PubMed  Google Scholar 

  71. Gustafson CT, et al. FH535 Suppresses Osteosarcoma Growth and Inhibits Wnt Signaling through Tankyrases. Front Pharmacol. 2017;8:285.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Razak S, et al. Growth inhibition and apoptosis in colorectal cancer cells induced by Vitamin D-Nanoemulsion (NVD): involvement of Wnt/β-catenin and other signal transduction pathways. Cell Biosci. 2019;9:15.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Zhang G-N, et al. Combination of salinomycin and gemcitabine eliminates pancreatic cancer cells. Cancer Lett. 2011;313(2):137–44.

    Article  CAS  PubMed  Google Scholar 

  74. He L, et al. Mechanism of action of salinomycin on growth and migration in pancreatic cancer cell lines. Pancreatology. 2013;13(1):72–8.

  75. Naujokat C, McKee DL. The “Big Five” Phytochemicals Targeting Cancer Stem Cells: Curcumin, EGCG, Sulforaphane. Resveratrol and Genistein Curr Med Chem. 2021;28(22):4321–42.

    Article  CAS  PubMed  Google Scholar 

  76. Li Y, et al. Implications of cancer stem cell theory for cancer chemoprevention by natural dietary compounds. J Nutr Biochem. 2011;22(9):799–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhou T, et al. ESE3/EHF, a promising target of rosiglitazone, suppresses pancreatic cancer stemness by downregulating CXCR4. Gut. 2022;71(2):357–71.

    Article  CAS  PubMed  Google Scholar 

  78. Masuo K, et al. SNAIL2 contributes to tumorigenicity and chemotherapy resistance in pancreatic cancer by regulating IGFBP2. Cancer Sci. 2021;112(12):4987–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Garg R, et al. Targeting FTO Suppresses Pancreatic Carcinogenesis via Regulating Stem Cell Maintenance and EMT Pathway. Cancers. 2022;14(23):5919.

  80. Leng S, et al. SIRT1 coordinates with the CRL4B complex to regulate pancreatic cancer stem cells to promote tumorigenesis. Cell Death Differ. 2021;28(12):3329–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Wang H, et al. Methyl-CpG-binding domain 3 inhibits stemness of pancreatic cancer cells via Hippo signaling. Exp Cell Res. 2020;393(1):112091.

    Article  CAS  PubMed  Google Scholar 

  82. Yang Y, et al. Deubiquitinase UCHL5 stabilizes ELK3 to potentiate cancer stemness and tumor progression in pancreatic adenocarcinoma (PAAD). Exp Cell Res. 2022;421(2):113402.

    Article  CAS  PubMed  Google Scholar 

  83. Barkeer S, et al. Novel role of O-glycosyltransferases GALNT3 and B3GNT3 in the self-renewal of pancreatic cancer stem cells. BMC Cancer. 2018;18(1):1157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. van Vlerken LE, et al. EZH2 is required for breast and pancreatic cancer stem cell maintenance and can be used as a functional cancer stem cell reporter. Stem Cells Transl Med. 2013;2(1):43–52.

    Article  PubMed  Google Scholar 

  85. Proctor E, et al. Bmi1 enhances tumorigenicity and cancer stem cell function in pancreatic adenocarcinoma. PLoS ONE. 2013;8(2): e55820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Courtois S, et al. Inhibition of Mitochondrial Dynamics Preferentially Targets Pancreatic Cancer Cells with Enhanced Tumorigenic and Invasive Potential. Cancers (Basel). 2021;13(4):698.

  87. Olichon A, et al. Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem. 2003;278(10):7743–6.

    Article  CAS  PubMed  Google Scholar 

  88. Carmona-Carmona CA, et al. Mitochondrial Elongation and OPA1 Play Crucial Roles during the Stemness Acquisition Process in Pancreatic Ductal Adenocarcinoma. Cancers (Basel). 2022;14(14):3432.

  89. Kamerkar SC, et al. Dynamin-related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat Commun. 2018;9(1):5239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Sancho P, et al. MYC/PGC-1α Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015;22(4):590–605.

    Article  CAS  PubMed  Google Scholar 

  91. Kumazoe M, et al. The FOXO3/PGC-1β signaling axis is essential for cancer stem cell properties of pancreatic ductal adenocarcinoma. J Biol Chem. 2017;292(26):10813–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Liesa M, et al. Mitochondrial fusion is increased by the nuclear coactivator PGC-1beta. PLoS ONE. 2008;3(10):e3613.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Qin T, et al. NAF-1 Inhibition by Resveratrol Suppresses Cancer Stem Cell-Like Properties and the Invasion of Pancreatic Cancer. Front Oncol. 2020;10:1038.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Zhu R, et al. TSPAN8 promotes cancer cell stemness via activation of sonic Hedgehog signaling. Nat Commun. 2019;10(1):2863.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Chen S, et al. FAM83A is amplified and promotes cancer stem cell-like traits and chemoresistance in pancreatic cancer. Oncogenesis. 2017;6(3):e300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhou C, et al. B-lymphoid tyrosine kinase-mediated FAM83A phosphorylation elevates pancreatic tumorigenesis through interacting with β-catenin. Signal Transduct Target Ther. 2023;8(1):66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Hara T, et al. Rer1-mediated quality control system is required for neural stem cell maintenance during cerebral cortex development. PLoS Genet. 2018;14(9):e1007647.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Chen S, et al. RER1 enhances carcinogenesis and stemness of pancreatic cancer under hypoxic environment. J Exp Clin Cancer Res. 2019;38(1):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Cable J, et al. Noncoding RNAs: biology and applications-a Keystone Symposia report. Ann N Y Acad Sci. 2021;1506(1):118–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. French R, Pauklin S. Epigenetic regulation of cancer stem cell formation and maintenance. Int J Cancer. 2021;148(12):2884–97.

    Article  CAS  PubMed  Google Scholar 

  101. Peng J-F, et al. Noncoding RNAs and pancreatic cancer. World J Gastroenterol. 2016;22(2):801–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Chen L, et al. Trends in the development of miRNA bioinformatics tools. Brief Bioinform. 2019;20(5):1836–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Mishra S, Yadav T, Rani V. Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit Rev Oncol Hematol. 2016;98:12–23.

    Article  PubMed  Google Scholar 

  104. Ouyang H, et al. microRNA-10b enhances pancreatic cancer cell invasion by suppressing TIP30 expression and promoting EGF and TGF-β actions. Oncogene. 2014;33(38):4664–74.

    Article  CAS  PubMed  Google Scholar 

  105. Gu J, et al. GFRα2 prompts cell growth and chemoresistance through down-regulating tumor suppressor gene PTEN via Mir-17-5p in pancreatic cancer. Cancer Lett. 2016;380(2):434–41.

    Article  CAS  PubMed  Google Scholar 

  106. Moriyama T, et al. MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol Cancer Ther. 2009;8(5):1067–74.

    Article  CAS  PubMed  Google Scholar 

  107. Zhao Y, et al. Antisense inhibition of microRNA-21 and microRNA-221 in tumor-initiating stem-like cells modulates tumorigenesis, metastasis, and chemotherapy resistance in pancreatic cancer. Target Oncol. 2015;10(4):535–48.

    Article  PubMed  Google Scholar 

  108. Ma Y, et al. miR-27a regulates the growth, colony formation and migration of pancreatic cancer cells by targeting Sprouty2. Cancer Lett. 2010;298(2):150–8.

    Article  CAS  PubMed  Google Scholar 

  109. Liu B, et al. lncRNA GAS5 Reverses EMT and Tumor Stem Cell-Mediated Gemcitabine Resistance and Metastasis by Targeting miR-221/SOCS3 in Pancreatic Cancer. Mol Ther Nucleic Acids. 2018;13:472–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Yao X, et al. Exosomal circ_0030167 derived from BM-MSCs inhibits the invasion, migration, proliferation and stemness of pancreatic cancer cells by sponging miR-338-5p and targeting the Wif1/Wnt8/β-catenin axis. Cancer Lett. 2021;512:38–50.

    Article  CAS  PubMed  Google Scholar 

  111. Wang F, et al. hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br J Cancer. 2010;103(4):567–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hasegawa S, et al. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br J Cancer. 2014;111(8):1572–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Nalls D, et al. Targeting epigenetic regulation of miR-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE. 2011;6(8):e24099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Bao B, et al. Curcumin analogue CDF inhibits pancreatic tumor growth by switching on suppressor microRNAs and attenuating EZH2 expression. Cancer Res. 2012;72(1):335–45.

    Article  CAS  PubMed  Google Scholar 

  115. Gao Y, et al. Linc-DYNC2H1-4 promotes EMT and CSC phenotypes by acting as a sponge of miR-145 in pancreatic cancer cells. Cell Death Dis. 2017;8(7):e2924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ali S, et al. Inactivation of Ink4a/Arf leads to deregulated expression of miRNAs in K-Ras transgenic mouse model of pancreatic cancer. J Cell Physiol. 2012;227(10):3373–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Zhou M, et al. MiR-146b-3p regulates proliferation of pancreatic cancer cells with stem cell-like properties by targeting MAP3K10. J Cancer. 2021;12(12):3726–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Wellner U, et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 2009;11(12):1487–95.

    Article  CAS  PubMed  Google Scholar 

  119. Shen J, et al. LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. Int J Biochem Cell Biol. 2019;113:17–26.

    Article  CAS  PubMed  Google Scholar 

  120. Ji Q, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE. 2009;4(8):e6816.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Pan Y, et al. MicroRNA-34a Alleviates Gemcitabine Resistance in Pancreatic Cancer by Repression of Cancer Stem Cell Renewal. Pancreas. 2021;50(9):1260–6.

    Article  CAS  PubMed  Google Scholar 

  122. Long LM, et al. The Clinical Significance of miR-34a in Pancreatic Ductal Carcinoma and Associated Molecular and Cellular Mechanisms. Pathobiology. 2017;84(1):38–48.

    Article  CAS  PubMed  Google Scholar 

  123. Lu Y, et al. MiR-200a inhibits epithelial-mesenchymal transition of pancreatic cancer stem cell. BMC Cancer. 2014;14:85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Singh SK, et al. Antithetical NFATc1-Sox2 and p53-miR200 signaling networks govern pancreatic cancer cell plasticity. Embo j. 2015;34(4):517–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Bao B, et al. Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells. Cancer Prevention Research (Philadelphia, Pa). 2012;5(3):355–64.

    Article  CAS  PubMed  Google Scholar 

  126. Liu B, et al. lncRNA GAS5 Reverses EMT and Tumor Stem Cell-Mediated Gemcitabine Resistance and Metastasis by Targeting miR-221/SOCS3 in Pancreatic Cancer. Molecular Therapy Nucleic Acids. 2018;13:472–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Deng S, et al. LncRNA HOTAIR Promotes Cancer Stem-Like Cells Properties by Sponging miR-34a to Activate the JAK2/STAT3 Pathway in Pancreatic Ductal Adenocarcinoma. Onco Targets Ther. 2021;14:1883–93.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Alonso-Nocelo M, et al. Macrophages direct cancer cells through a LOXL2-mediated metastatic cascade in pancreatic ductal adenocarcinoma. Gut. 2023;72(2):345–59.

    Article  CAS  PubMed  Google Scholar 

  129. Penchev VR, et al. Ezrin Promotes Stem Cell Properties in Pancreatic Ductal Adenocarcinoma. Mol Cancer Res. 2019;17(4):929–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kesharwani P, et al. Hyaluronic Acid Engineered Nanomicelles Loaded with 3,4-Difluorobenzylidene Curcumin for Targeted Killing of CD44+ Stem-Like Pancreatic Cancer Cells. Biomacromol. 2015;16(9):3042–53.

    Article  CAS  Google Scholar 

  131. Yang M, et al. The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment. Bioact Mater. 2021;6(7):1973–87.

    Article  CAS  PubMed  Google Scholar 

  132. Simpson DS, et al. Interferon-γ primes macrophages for pathogen ligand-induced killing via a caspase-8 and mitochondrial cell death pathway. Immunity. 2022;55(3):423-441.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Begum A, et al. Direct Interactions With Cancer-Associated Fibroblasts Lead to Enhanced Pancreatic Cancer Stem Cell Function. Pancreas. 2019;48(3):329–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Begum A, et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS ONE. 2017;12(7):e0180181.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Chen H, et al. Smart hypoxia-responsive transformable and charge-reversible nanoparticles for the deep penetration and tumor microenvironment modulation of pancreatic cancer. Biomaterials. 2022;287:121599.

    Article  CAS  PubMed  Google Scholar 

  136. Lee IC, Wu YC, Hung WS. Hyaluronic Acid-Based Multilayer Films Regulate Hypoxic Multicellular Aggregation of Pancreatic Cancer Cells with Distinct Cancer Stem-Cell-like Properties. ACS Appl Mater Interfaces. 2018;10(45):38769–79.

    Article  CAS  PubMed  Google Scholar 

  137. Cao J, et al. Targeting glypican-4 overcomes 5-FU resistance and attenuates stem cell-like properties via suppression of Wnt/β-catenin pathway in pancreatic cancer cells. J Cell Biochem. 2018;119(11):9498–512.

    Article  CAS  PubMed  Google Scholar 

  138. Chan T-S, Shaked Y, Tsai KK. Targeting the Interplay Between Cancer Fibroblasts, Mesenchymal Stem Cells, and Cancer Stem Cells in Desmoplastic Cancers. Front Oncol. 2019;9:688.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Steele NG, et al. Inhibition of Hedgehog Signaling Alters Fibroblast Composition in Pancreatic Cancer. Clin Cancer Re. 2021;27(7):2023–37.

    Article  CAS  Google Scholar 

  140. Vennin C, et al. CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan. Nat Commun. 2019;10(1):3637.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Nallasamy P, et al. Pancreatic Tumor Microenvironment Factor Promotes Cancer Stemness via SPP1-CD44 Axis. Gastroenterology. 2021;161(6):1998–2013.e7.

  142. Zhao K, Ma Z, Zhang W. Comprehensive Analysis to Identify as a Prognostic Biomarker in Cervical Cancer. Front Genet. 2021;12:732822.

    Article  CAS  PubMed  Google Scholar 

  143. Göthlin Eremo A, et al. Evaluation of SPP1/osteopontin expression as predictor of recurrence in tamoxifen treated breast cancer. Sci Rep. 2020;10(1):1451.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Qian J, et al. Cancer-associated mesothelial cells promote ovarian cancer chemoresistance through paracrine osteopontin signaling. J Clin Inv. 2021;131(16):e146186.

  145. Pietras A, et al. Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell. 2014;14(3):357–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Yan B, et al. Paracrine HGF/c-MET enhances the stem cell-like potential and glycolysis of pancreatic cancer cells via activation of YAP/HIF-1α. Exp Cell Res. 2018;371(1):63–71.

    Article  CAS  PubMed  Google Scholar 

  147. Zhang X, et al. Yes-associated protein (YAP) binds to HIF-1α and sustains HIF-1α protein stability to promote hepatocellular carcinoma cell glycolysis under hypoxic stress. J Exp Clin Cancer Res. 2018;37(1):216.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Peng F, et al. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene. 2018;37(8):1062–74.

    Article  CAS  PubMed  Google Scholar 

  149. Nelson JK, et al. USP25 promotes pathological HIF-1-driven metabolic reprogramming and is a potential therapeutic target in pancreatic cancer. Nat Commun. 2022;13(1):2070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ji L, et al. Worenine reverses the Warburg effect and inhibits colon cancer cell growth by negatively regulating HIF-1α. Cell Mol Biol Lett. 2021;26(1):19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Dong L, et al. Arylsulfonamide 64B Inhibits Hypoxia/HIF-Induced Expression of c-Met and CXCR4 and Reduces Primary Tumor Growth and Metastasis of Uveal Melanoma. Clin Cancer Res. 2019;25(7):2206–18.

    Article  CAS  PubMed  Google Scholar 

  152. Hou P, et al. USP21 deubiquitinase promotes pancreas cancer cell stemness via Wnt pathway activation. Genes Dev. 2019;33(19–20):1361–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Mosa MH, et al. A Wnt-Induced Phenotypic Switch in Cancer-Associated Fibroblasts Inhibits EMT in Colorectal Cancer. Can Res. 2020;80(24):5569–82.

    Article  CAS  Google Scholar 

  154. Kesh K, et al. Stroma secreted IL6 selects for “stem-like” population and alters pancreatic tumor microenvironment by reprogramming metabolic pathways. Cell Death Dis. 2020;11(11):967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Li X, et al. SDF-1/CXCR4 signaling induces pancreatic cancer cell invasion and epithelial-mesenchymal transition in vitro through non-canonical activation of Hedgehog pathway. Cancer Lett. 2012;322(2):169–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Lonardo E, et al. Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell. 2011;9(5):433–46.

    Article  CAS  PubMed  Google Scholar 

  157. Panni RZ, et al. Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol Immunother. 2014;63(5):513–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Chang J, et al. microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol Toxicol. 2022;38(4):577–90.

    Article  CAS  PubMed  Google Scholar 

  159. Mitchem JB, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73(3):1128–41.

    Article  CAS  PubMed  Google Scholar 

  160. Wang VM, et al. CD9 identifies pancreatic cancer stem cells and modulates glutamine metabolism to fuel tumour growth. Nat Cell Biol. 2019;21(11):1425–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Mani SA, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Morel A-P, et al. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE. 2008;3(8):e2888.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Zhang J, et al. Notch-4 silencing inhibits prostate cancer growth and EMT via the NF-κB pathway. Apoptosis. 2017;22(6):877–84.

    Article  CAS  PubMed  Google Scholar 

  164. Krebs AM, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol. 2017;19(5):518–29.

    Article  CAS  PubMed  Google Scholar 

  165. Zhang H, et al. CD44 splice isoform switching determines breast cancer stem cell state. Genes Dev. 2019;33(3–4):166–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wu CL, et al. Dual role of CD44 isoforms in ampullary adenocarcinoma: CD44s predicts poor prognosis in early cancer and CD44ν is an indicator for recurrence in advanced cancer. BMC Cancer. 2015;15:903.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Zhou J, et al. CD44 Expression Predicts Prognosis of Ovarian Cancer Patients Through Promoting Epithelial-Mesenchymal Transition (EMT) by Regulating Snail, ZEB1, and Caveolin-1. Front Oncol. 2019;9:802.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Matsuda Y, et al. Systemic Administration of Small Interfering RNA Targeting Human Nestin Inhibits Pancreatic Cancer Cell Proliferation and Metastasis. Pancreas. 2016;45(1):93–100.

  169. Matsuda Y, et al. Nestin is a novel target for suppressing pancreatic cancer cell migration, invasion and metastasis. Cancer Biol Ther. 2011;11(5):512–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Maier HJ, et al. NF-kappaB promotes epithelial-mesenchymal transition, migration and invasion of pancreatic carcinoma cells. Cancer Lett. 2010;295(2):214–28.

    Article  CAS  PubMed  Google Scholar 

  171. Chua HL, et al. NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene. 2007;26(5):711–24.

    Article  CAS  PubMed  Google Scholar 

  172. Banerjee S, et al. CD133+ tumor initiating cells in a syngenic murine model of pancreatic cancer respond to Minnelide. Clin Cancer Res. 2014;20(9):2388–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Nomura A, et al. NFκB-Mediated Invasiveness in CD133 Pancreatic TICs Is Regulated by Autocrine and Paracrine Activation of IL1 Signaling. Mol Cancer Res. 2018;16(1):162–72.

    Article  CAS  PubMed  Google Scholar 

  174. Nomura A, et al. Microenvironment mediated alterations to metabolic pathways confer increased chemo-resistance in CD133+ tumor initiating cells. Oncotarget. 2016;7(35):56324–37.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Huang H, et al. Suppression of mitochondrial ROS by prohibitin drives glioblastoma progression and therapeutic resistance. Nat Commun. 2021;12(1):3720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Nimmakayala RK, et al. Metabolic programming of distinct cancer stem cells promotes metastasis of pancreatic ductal adenocarcinoma. Oncogene. 2021;40(1):215–31.

    Article  CAS  PubMed  Google Scholar 

  177. Wang L, et al. Gemcitabine treatment induces endoplasmic reticular (ER) stress and subsequently upregulates urokinase plasminogen activator (uPA) to block mitochondrial-dependent apoptosis in Panc-1 cancer stem-like cells (CSCs). PLoS ONE. 2017;12(8):e0184110.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Zhao S, et al. CD44 Expression Level and Isoform Contributes to Pancreatic Cancer Cell Plasticity, Invasiveness, and Response to Therapy. Clin Cancer Re. 2016;22(22):5592–604.

    Article  CAS  Google Scholar 

  179. Li L, et al. Antibody against CD44s inhibits pancreatic tumor initiation and postradiation recurrence in mice. Gastroenterology. 2014;146(4):1108–18.

    Article  CAS  PubMed  Google Scholar 

  180. Dzobo K, Sinkala M. Cancer Stem Cell Marker CD44 Plays Multiple Key Roles in Human Cancers: Immune Suppression/Evasion, Drug Resistance, Epithelial-Mesenchymal Transition, and Metastasis. OMICS. 2021;25(5):313–32.

    Article  CAS  PubMed  Google Scholar 

  181. Hong SP, et al. CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer. 2009;125(10):2323–31.

    Article  CAS  PubMed  Google Scholar 

  182. Fletcher JI, et al. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updat. 2016;26:1–9.

    Article  PubMed  Google Scholar 

  183. Bates RC, et al. A CD44 survival pathway triggers chemoresistance via lyn kinase and phosphoinositide 3-kinase/Akt in colon carcinoma cells. Can Res. 2001;61(13):5275–83.

    CAS  Google Scholar 

  184. Liu C-M, et al. Hyaluronan substratum induces multidrug resistance in human mesenchymal stem cells via CD44 signaling. Cell Tissue Res. 2009;336(3):465–75.

    Article  CAS  PubMed  Google Scholar 

  185. Huang W, et al. The inhibitory effect and mechanism of Yi-qi-hua-yu-jie-du decoction on the drug resistance of gastric cancer stem cells based on ABC transporters. Chinese Medicine. 2022;17(1):93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Ruivo CF, et al. Extracellular Vesicles from Pancreatic Cancer Stem Cells Lead an Intratumor Communication Network (EVNet) to fuel tumour progression. Gut. 2022;71(10):2043–68.

  187. Xiong W-C, Mei L. Agrin to YAP in Cancer and Neuromuscular Junctions. Trends In Cancer. 2017;3(4):247–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Ikenaga N, et al. Characterization of CD24 expression in intraductal papillary mucinous neoplasms and ductal carcinoma of the pancreas. Hum Pathol. 2010;41(10):1466–74.

    Article  CAS  PubMed  Google Scholar 

  189. Lee SH, et al. CD24 and S100A4 expression in resectable pancreatic cancers with earlier disease recurrence and poor survival. Pancreas. 2014;43(3):380–8.

    Article  CAS  PubMed  Google Scholar 

  190. Li X, et al. Prognostic value of cancer stem cell marker CD133 expression in pancreatic ductal adenocarcinoma (PDAC): a systematic review and meta-analysis. Int J Clin Exp Pathol. 2015;8(10):12084–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Li X-P, et al. Expression of CD44 in pancreatic cancer and its significance. Int J Clin Exp Pathol. 2015;8(6):6724–31.

    PubMed  PubMed Central  Google Scholar 

  192. He Z, et al. MicroRNA-137 reduces stemness features of pancreatic cancer cells by targeting KLF12. J Exp Clin Cancer Res. 2019;38(1):126.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Yang X-L, et al. microRNA-873 inhibits self-renewal and proliferation of pancreatic cancer stem cells through pleckstrin-2-dependent PI3K/AKT pathway. Cell Signal. 2021;84:110025.

    Article  CAS  PubMed  Google Scholar 

  194. Kan S, et al. Latexin deficiency attenuates adipocyte differentiation and protects mice against obesity and metabolic disorders induced by high-fat diet. Cell Death Dis. 2022;13(2):175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Izumiya M, et al. Chemoresistance is associated with cancer stem cell-like properties and epithelial-to-mesenchymal transition in pancreatic cancer cells. Anticancer Res. 2012;32(9):3847–53.

    CAS  PubMed  Google Scholar 

  196. Verma RK, et al. α-Mangostin-encapsulated PLGA nanoparticles inhibit pancreatic carcinogenesis by targeting cancer stem cells in human, and transgenic (Kras(G12D), and Kras(G12D)/tp53R270H) mice. Sci Rep. 2016;6:32743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Butti R, et al. Breast cancer stem cells: Biology and therapeutic implications. Int J Biochem Cell Biol. 2019;107:38–52.

    Article  CAS  PubMed  Google Scholar 

  198. Chae YC, Kim JH. Cancer stem cell metabolism: target for cancer therapy. BMB Rep. 2018;51(7):319–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Weibin Wang received the support from the National Natural Science Foundation of China (No. 82173074), the National High Level Hospital Clinical Research Funding (No. 2022-PUMCH-D-001, No. 2022-PUMCH-B-004), the CAMS Innovation Fund for Medical Sciences (CIFMS) (No. 2021-I2M-1–002), and the Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences (2018PT32014). Cheng Qin received support from the Fundamental Research Funds for the Central Universities (3332022114).

Author information

Authors and Affiliations

Authors

Contributions

WW conceived, designed the review and gave constructive guidance. YZ and CQ collected related literature and finished the manuscript. BZ, YW, ZL, TL, XY participated in the design of this review. All authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Weibin Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, Y., Qin, C., Zhao, B. et al. Pancreatic cancer stemness: dynamic status in malignant progression. J Exp Clin Cancer Res 42, 122 (2023). https://doi.org/10.1186/s13046-023-02693-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13046-023-02693-2

Keywords