IMR Press / FBL / Volume 28 / Issue 12 / DOI: 10.31083/j.fbl2812361
Open Access Review
An Update on Gemcitabine-Based Chemosensitization Strategies in Pancreatic Ductal Adenocarcinoma
Show Less
1 Department of Surgery, Bengbu Medical College, 233030 Bengbu, AnHui, China
2 General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310000 Hangzhou, Zhejiang, China
*Correspondence: taohdac9@yahoo.com (Ran Tao)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(12), 361; https://doi.org/10.31083/j.fbl2812361
Submitted: 1 June 2023 | Revised: 22 July 2023 | Accepted: 10 August 2023 | Published: 29 December 2023
(This article belongs to the Special Issue New Targets in Anticancer Therapy)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Pancreatic cancer is the seventh leading cause of cancer-related deaths, and chemotherapy is one of the most important treatments for pancreatic cancer. Unfortunately, pancreatic cancer cells can block chemotherapy drugs from entering the tumor. This is owing to interactions between the tumor’s environment and the cancer cells. Here, we review the latest research on the mechanisms by which pancreatic cancer cells block the chemotherapy drug, gemcitabine. The results of our review can help identify potential therapeutic targets for the blocking of gemcitabine by pancreatic cancer cells and may provide new strategies to help chemotherapy drugs penetrate tumors.

Keywords
pancreatic cancer
chemotherapy resistance
chemosensitization
tumor microenvironment
gemcitabine
1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) accounts for approximately 90% of pancreatic cancers, which have one of the worst prognoses among solid malignancies in humans. Pancreatic cancer is the seventh leading cause of cancer death, with a 5-year survival rate of only 10% [1, 2, 3]. Surgical resection is the only possible cure for early-stage PDAC; however, only 15%–20% of patients are eligible for surgery, and the overall 5-year survival rate after surgery is only 10%–25% [4, 5]. Therefore, chemotherapy remains a crucial treatment option for PDAC.

Effective delivery of gemcitabine (GEM) to tumor foci is a critical aspect of PDAC treatment. However, the presence of highly fibrotic and densely vascularized tumor tissues poses challenges to drug delivery, leading to limited delivery and difficulty in achieving adequate drug concentrations [6]. Importantly, the interaction between PDAC cells and the tumor microenvironment (TME) is known to stimulate extensive fibrous proliferation [7]. Patients with PDAC have shown poor response rates to immune checkpoint inhibitors, such as anti-programmed cell death protein 1 (PD-1), anti-programmed death ligand 1 (PD-L1), and anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination [8]. However, Bockorny et al. [8] recently showed that the combination of CXCR4 and PD-1 may enhance the chemotherapeutic efficacy for PDAC, and this finding was later confirmed in a subsequent randomized controlled trial (RCT). The TME in PDAC is characterized by its complexity and diverse cellular components, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and immune cells. These cellular elements can generate chemical signals that are associated with GEM and can, consequently, impact the efficacy of the drug [9].

GEM has been the basis of first-line chemotherapy for patients with PDAC. Although FOLFIRINOX has shown some benefits in certain studies, a recent randomized phase II clinical trial (NCT02562716) demonstrated that, compared to the AG regimen, the FOLFIRINOX regimen did not prolong survival, instead it substantially increased drug toxicity [10]. The mechanism underlying GEM cytotoxicity involves its role as a deoxycytidine nucleoside analog that competes for DNA strand incorporation and terminates its replication, ultimately leading to cell death [11]. Our team elucidated the role of metabolic remodeling in GEM-resistant pancreatic cancer chemotherapy [12]. Recently, several key developments have been uncovered regarding the involvement of TME in GEM chemotherapy resistance mechanisms in PDAC; however, few reviews are currently available. Therefore, this review aims to provide the current research progress on the mechanism of GEM chemoresistance, with the goal of identifying potential therapeutic targets for GEM resistance and providing new strategies for chemosensitization.

2. Non-Cellular Components of TME in Chemotherapy Resistance
2.1 Hypoxia

Extensive fibrosis and reduced blood supply vessels are among the important features of PDAC, which lead to severe tissue hypoxia. Persistent hypoxia has been demonstrated in pancreatic cancer, with a 17-fold decrease in oxygen utilization in PDAC tissues compared with that of other tumors. The average oxygenation level in the healthy pancreas is 6.8%, whereas it is only 0.4% in pancreatic cancer [13]. Hypoxia-induced activation of hypoxia-inducible factor (HIF) signaling is present in almost all types of malignancies [14]. Additionally, HIF-1α, HIF-2α, and HIF-3α isoforms have been identified. In PDAC, the primary sources of extracellular H+ are HIF-1α-mediated glycolytic lactate production and carbonic anhydrase (CA)-mediated carbonic acid production [15]. HIF-1α induces CA expression by upregulating polo-like kinase 4 (PLK4), mediating increased PDAC glycolysis, changes in intracellular and extracellular pH, and increasing GEM resistance [16] (Fig. 1). Similarly, another study demonstrated that HIF-1α is an upstream regulator of retention in endoplasmic reticulum sorting receptor 1 (RER1), and knockdown of RER1 inhibited PDAC cell proliferation, migration, and invasion [17]. Deletion of HIF-2α inhibited tumor progression in PDAC mouse models by reducing macrophage chemotaxis and M2-type polarization, suggesting that HIF-2α is a potential target for chemosensitization [18]. A promising clinical trial of an HIF-2 inhibitor (belzutifan) II is underway (NCT04924075). Additionally, the expression of HIF-3α is more substantial in PDAC cells than that of HIF-1α and HIF-2α under hypoxic conditions. HIF-3α promotes invasion and metastasis of pancreatic cancer cells in vivo by activating the Ras homolog family member C (RhoC)-Rho-associated coiled-coil containing protein kinase 1 (ROCK1) signaling pathway, and targeted inhibition of the HIF-3α/RhoC-ROCK1 signaling pathway may be a novel approach to reverse chemoresistance in pancreatic cancer [19] (Fig. 1).

Fig. 1.

In the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC), an abundance of fibrous tissue is observed, leading to a persistent hypoxic condition. This fibrosis is also responsible for impeding drug penetration due to its high levels. HIF, hypoxia-inducible factor; LM, Laminin; RhoC, Ras homolog family member C; ROCK1, Rho-associated coiled-coil containing protein kinase 1; HA, Hyaluronan; FAK, focal adhesion kinase; PLK4, polo-like kinase4; NF-κB, nuclear factor kappa B.

2.2 Biological Barriers

Collagen is a major component of the TME and plays a role in forming dense lattices in cancer fibrosis. Type I collagen, which is closely related to PDAC, is a heterotrimer and includes many Gly-X-Y repeat sequences [20]. Type I collagen activates focal adhesion kinase (FAK), a multifunctional non-receptor protein tyrosine kinase, by binding to β1 integrin and associating with multiple α subunits [21]. This induces self-renewal and invasion of pancreatic cancer stem cells (CSCs) [22], in addition to fibrous proliferation that enhances the physical barrier to chemotherapeutic drug delivery [23]. Defactinib is a potent oral FAK inhibitor, and a recent phase I clinical trial (NCT02546531) study has shown that the triple drug combination of defactinib, pembrolizumab, and GEM in patients with PDAC prolonged their overall survival [24]. Additionally, collagen restricts the effect of GEM by upregulating membrane type-1 matrix metalloproteinase (MT1-MMP)-dependent ERK1/2 phosphorylation and allowing MT1-MMP-dependent overexpression of high mobility group protein A2 (HMGA2) [25]. HMGA2 has three lysine- and arginine-rich AT-hook domains that bind to AT-rich sequences in DNA minor grooves. These AT-rich sequences and AT structural domains promote cytoprotective DNA base excision repair by their cleavage enzyme activity [26], thereby reducing the killing effect of GEM. Therefore, targeting collagen-related pathways may be a new GEM sensitization strategy [27] (Fig. 1).

Laminin (LM) and fibronectin are important basement membrane components involved in blood vessel formation [28]. LM-332, composed of α3, β3, and γ2 subunits, is a major member of the LM family, which is overexpressed in PDAC. Targeted inhibition of the three subunits hinders tumor cell proliferation, migration, and invasion; thus, the three subunits of LM-332 are expected to be new chemotherapeutic sensitization targets [29]. A xenograft model showed that fibroblasts stimulated by transglutaminase 2 (TG2) secreted by PDAC cells prompted the secretion of LM-A1, which protected cancer cells from GEM-induced cytotoxicity. This process is associated with the activation of nuclear factor kappa B (NF-κB) and FAK/PI3K/AKT pro-survival signaling pathways [30, 31]. Additionally, LM promotes survival protein expression (survivin) by inducing FAK/Akt phosphorylation, which resists GEM-induced cytotoxicity and apoptosis. Therefore, selective inhibition of FAK phosphorylation may be another approach to enhance the chemosensitivity of pancreatic cancer [32] (Fig. 1).

Fibronectin is less abundant in the extracellular matrix (ECM), but can interact with various proteins for signal transduction, promote angiogenesis [33] and play an essential role in cell adhesion and mobility [34]. Fibronectin can resist GEM-induced apoptosis by inducing ERK1/2 phosphorylation, thereby mediating GEM drug resistance [35]. However, fibronectin was abundantly expressed in PDAC but did not affect patient survival [36]. Therefore, fibronectin may play a dual role in the development of PDAC.

Hyaluronan (HA) is a proteoglycan secreted by stromal cells highly expressed in the PDAC stroma. It has been associated with elevated interstitial fluid pressure, compression of intra-tumor vessels, decrease in tumor perfusion and impaired vascular permeability, thus, leading to increased physical barriers for chemotherapeutic drug delivery and resulting in GEM resistance [37]. In addition, HA should bind specific receptors to function, primarily CD44, a non-kinase transmembrane receptor with multiple isoforms located on the cell membrane. GEM can induce CD44 subtype switching and promote switching to the more resistant and aggressive PDAC phenotype [38]. Activation of epidermal growth factor receptor (EGFR) and insulin-like growth factor-1 receptor (IGF1R) can induce CD44 subtype switching and promote chemoresistance. However, activation of these growth factor receptors may not be required to maintain the resistant phenotype, and blocking IGF1R signaling has been shown to contribute to downregulation of GEM-induced CD44 isoform conversion, thereby inhibiting cell invasiveness and improving chemoresistance [39] (Fig. 1). Moreover, 4-methylumbelliferone (4-MU), an inhibitor of HA synthesis, has been shown to reduce HA production and PDAC cell migration. Therefore, 4-MU could be used to improve GEM sensitivity [40].

3. Cellular Components of the TME in Chemotherapy Resistance

The cellular components of the TME in chemoresistance include CAFs, TAMs, immune cells, and various other types of cells. These diverse cell types play a crucial role in mediating chemoresistance. Moreover, the TME also comprises additional cell types, such as mesenchymal cells and mesenchymal stem cells that have the potential to considerably influence tumor development and chemotherapy resistance.

3.1 Mesenchymal Cells

Activated pancreatic stellate cells (aPSCs) can interact with PDAC cells directly and through the paracrine (ECM protein) pathway, which is additionally involved in GEM resistance [41, 42, 43, 44]. In PDAC, CAFs are mainly derived from the activation of PSCs [41, 45, 46] and are induced to form two subtypes, inflammatory (iCAF) and myofibroblast (myCAF), by the direct action of PDAC cells and paracrine signaling [47]. myCAFs are naturally resistant to GEM and can induce acquired resistance through multiple mechanisms [45, 48]. iCAFs promote tumor chemoresistance through a series of cytokines and chemokines [49]. Mechanistically, iCAFs further activate the JAK-STAT signaling pathway through interleukin-1 (IL-1) stimulation to induce chemoresistance. Preclinical studies have shown that JAK inhibitors can reverse this effect [50] (Fig. 2). Unfortunately, ruxolitinib (a JAK1 and JAK2 inhibitor) did not improve the overall survival of patients with locally advanced or metastatic PDAC in two phase III RCTs (NCT02117479 and NCT02119663) [51]. Furthermore, genomic analysis has revealed that IL-6 is overexpressed in GEM-resistant iCAFs and may be a key inhibitor of immune checkpoint blockade therapy in PDAC [52]. A phase II trial (NCT02767557) is currently underway, which targets IL-6 inhibitors in combination with GEM, nab-paclitaxel, and golimumab [53]. Notably, GEM treatment has been found to induce CAFs to secrete an increased number of exosomes. CAFs present in tumors release exosomes that exhibit high expression of miR-3173-5p. This particular microRNA promotes drug tolerance by suppressing ferroptosis through the miR-3173-5p/ACSL4 pathway [54]. Ferroptosis is a newly identified form of iron-dependent programmed cell death that has been associated with GEM chemoresistance in PDAC [55]. ACSL4 plays a crucial role in regulating ferroptosis by activating fatty acid metabolism. Numerous exosome studies are ongoing [56], which may provide new insights into overcoming GEM resistance.

Fig. 2.

PDAC cells interact with stromal cells. Stromal cells can interact with PDAC cells through direct contact or paracrine secretion, enhancing the efficacy of gemcitabine (GEM). Additionally, stromal and immune cells can secrete various cytokines and chemokines, activating multiple signaling pathways to interact with PDAC. They can also induce GEM resistance by promoting stem cell self-renewal. Long non-coding RNA (lncRNA) can also affect PDAC cell proliferation and induce GEM resistance through multiple signaling pathways. PD-1, programmed death-1; PD-L1, programmed death ligand 1; CTLA-4, cytotoxic T lymphocyte associated antigen-4; HIF, hypoxia-inducible factor; CSCs, cancer stem cells; PSCs, pancreatic stellate cells; CAFs, cancer-associated fibroblasts; IL, interleukin; LncRNA, long non-coding RNA; TAMs, tumor-associated macrophages; MDSCs, myeloid-derived suppressor cells; CXCL, chemokine ligands; TILs, tumor-infiltrating lymphocytes; PLTs, platelets; M-MDSCs, monocytic-myeloid-derived suppressor cells; GTGF, connective tissue growth factor; Tregs, regulatory T cells; TGFβ1, transforming growth factor β1.

3.2 Tumor Stem Cells

CSCs account for approximately 0.5%–1% of pancreatic cancer cells. Furthermore, CSCs can self-renew; hence, playing a key role in the development of PDAC and chemoresistance [57, 58]. Preclinical studies have shown that methyl-CpG binding domain 3 (MBD3) is overexpressed in PDAC. The knockdown of MBD3 reduces Oct4, Nanog, and Sox2 expression; impairs the self-renewal capacity of CSCs; and inhibits the proliferation of PDAC cells [59] (Fig. 2). Similarly, inhibition of Oct4 and Nanog expression suppressed the tumorigenic phenotype of CSCs and increased the chemosensitivity of CSCs in mice [60]. Betulinic acid (BA) can downregulate the expression of Sox2, Oct4, and Nanog by activating the AMPK signaling pathway [61]. Therefore, BA is expected to be a potential GEM chemotherapy-sensitizing agent. Moreover, NR5A2 overexpressed in PDAC cells plays an important role in maintaining stemness and promoting epithelial-mesenchymal transition (EMT) in CSCs [62], suggesting that it could be a potential therapeutic target. There is growing evidence that the dysregulation of long non-coding RNAs (lncRNAs) is closely associated with the development and metastasis of PDAC. HOX transcript antisense RNA is a class of lncRNAs, which can promote proliferation and hinder apoptosis of CSCs by inhibiting miR-34a-activated JAK2/STAT3 and inducing GEM resistance [63, 64]. MACC1-AS1 was recently identified using lncRNA microarray as the most abundant lncRNA in PDAC—it promotes PDAC cell proliferation and metastasis through MACC1-AS1/PAX8/NOTCH1 axis [65, 66]. The current research on CSCs and related non-coding RNAs has received extensive attention, and it is predicted that they are highly likely to constitute another strategy for GEM sensitization.

3.3 Immune Cells

TAMs consist of two main subtypes, i.e., M1 and M2, with the M1 type activating the immune response against the tumor and increasing chemotherapy sensitivity, contrary to the M2 type [67]. Therefore, induction of TAM reprogramming to M1 or specific inhibition of M2 conversion can help increase GEM sensitivity. CCL2/CCR2 axis activation can recruit inflammatory monocytes to the TME and achieve M2-type conversion, and CCR2 inhibitors can effectively block TAM recruitment [68]. A phase Ib clinical trial (NCT01413022) confirmed that a CCR inhibitor (PF-04136309) in combination with GEM enhances chemotherapeutic efficacy, and further RCT results are expected [69]. M2 can also promote EMT in PDAC through TLR4/IL-10 signaling and induce GEM resistance [70]. Therefore, the TLR4/IL-10 pathway may also be a promising target for chemosensitization.

Myeloid-derived suppressor cells (MDSCs) are heterogeneous immune cell populations of immature myeloid cells that suppress T-cell immunity and promote angiogenesis [71]. Among them, monocyte-like MDSCs (M-MDSCs) have a high plasticity in the TME [72]. The TME can increase the number of M-MDSCs via secretion of CCL2, CCL5, and CSF1 [73]. In addition, under hypoxic conditions with upregulated HIF-1α, M-MDSCs can differentiate into TAMs, thus, promoting PDAC progression and GEM resistance [74] (Fig. 2). Inhibition of the CCL2–CCR2 signaling pathway considerably reduces MDSCs migration, decreases TAM numbers, and inhibits tumor growth [75]. Hypoxia inhibits the STAT3 pathway by upregulating CD45 tyrosine phosphatase activity in MDSCs and induces differentiation of M-MDSCs to TAM; therefore, this pathway may also be a potential target for chemotherapy sensitization [76].

Circulating neutrophils can also be recruited by tumors via chemokine action; however, the role of TME neutrophils in chemotherapy may be bidirectional, which may be related to distinct differentiation subtypes [77]. Inhibition of the CXCR2 axis blocks neutrophil recruitment and increases T-cell recruitment [78]. In vitro experiments have confirmed that CXCR2 inhibitors can enhance antitumor immunity and toxic effects of GEM [77, 79]. Notably, neutrophil extracellular trap (NET) is a newly discovered form of neutrophil activation that has been shown to play a crucial role in innate immunity and tumor progression [80, 81]. NET can promote EMT in PDAC cells through the IL-1β/EGFR/ERK pathway and is considered to be a new potential target for chemosensitization [82].

Tumor-infiltrating lymphocytes (TILs) are key cells involved in antitumor immunity and regulation of immune checkpoints. The results of several studies on PD-1/PD-L1/CTLA-4 inhibitors suggest that GEM combined with immunotherapy is a viable option, although many questions remain regarding the underlying mechanisms [83] (Fig. 2). GEM combined with anti-PD-1 antibodies can enhance the immune response of Th1 lymphocytes, resulting in benefits for patients with advanced PDAC [84]. Based on histopathological features, TME immune cells can be classified into three subtypes: immune enriched, escape, and exhaustion. Among them, the immune escape subtype is the most common and characterized by a low content of T and B cells and an enrichment of regulatory T cells (Tregs) [85]. However, Treg depletion failed to alleviate immunosuppression in PDAC tumors, but instead led to accelerated tumor progression. Thus, Tregs infiltration may have a protective role in PDAC; although the exact mechanism is unknown [86, 87]. In summary, studies on the complex mechanisms underlying the role of TILs in TME immunity are emerging and may provide new insights for the comprehensive treatment of PDAC.

The role of platelets (PLTs) in tumor development remains controversial [88]. Recently, it has been shown that PDAC cells can induce the recruitment activation of PLTs. This further promotes EMT and induces GEM resistance through direct contact or release of transforming growth factor β1 [89, 90, 91]. Therefore, PLTs are prospective chemotherapy sensitization targets.

4. Interaction between Cellular and Non-Cellular Components of TME in Chemotherapy Resistance

Cytokines and chemokines play a key role in mediating the interaction between PDAC cells and ECM, and in the induction of GEM resistance (Fig. 2).

4.1 Cytokines

Connective tissue growth factor (CTGF) can interact with other growth factors to mediate chemoresistance. CTGF and FGF2 have been shown to jointly induce ERK phosphorylation through fibroblast growth factor receptor 2 (FGFR2) signaling, leading to resistance to GEM-induced cytotoxicity and promoting chemoresistance [92]. Moreover, TGF-β promotes CTGF production in PC cell lines and further induces FGFR2 expression. Sensitization to GEM chemotherapy can be achieved with a CTGF antagonist (mAbFG-3019) [93]. A subsequent phase II study (NCT02210559) showed that pamrevlumab (FG-3019) enhanced the sensitivity to GEM in patients with locally advanced pancreatic cancer [94].

Targeted anti-EGFR molecular strategies have been used to reverse the resistance of PDAC to GEM [95]. Among them, erlotinib is one of the most studied EGFR inhibitors. An earlier phase III clinical study showed that erlotinib combined with GEM treatment prolonged the overall survival of patients with pancreatic cancer [96]. Mechanistically, targeted inhibition of EGFR blocks STAT3 pathway activation, remodels the tumor stroma, increases microvascular density, and enhances GEM efficacy [97]. Unfortunately, a recent multicenter phase III clinical study (NCT02694536) found no notable difference in disease-free and overall survival for erlotinib combined with GEM compared with GEM monotherapy for advanced PDAC [98]. Additionally, c-Kit, an important member of the tyrosine kinase receptor protein family, is overexpressed in PDAC, and masitinib is a novel specific c-Kit inhibitor. Masitinib reverses the efficacy of PDAC against GEM and improves patient prognosis [99]. The IGF-1/IGF-1R pathway is upregulated in approximately 72% of patients with PDAC [100], promoting PDAC proliferation and inducing GEM resistance by activating the downstream PI3K/Akt/mTOR and MEK/ERK pathways [101]. Moreover, ganitumab specifically inhibits IGF-1R, and an earlier phase II clinical study showed that ganitumab combined with GEM prolonged the overall survival of PDAC with a favorable safety profile [102]. Unfortunately, subsequent phase III studies showed no positive results [103]. Notably, a recent phase I/II clinical trial in PDAC (NCT00769483) has demonstrated that the (MK-0646) IGF-1R inhibitor in combination with GEM + erlotinib considerably improved survival compared to GEM + erlotinib [104]. Reassuringly, a recent phase 1b study showed better results with dovitinib in combination with GEM in treating patients with advanced pancreatic cancer [105]. Dovitinib (TKI258) is a novel multi-targeted tyrosine kinase inhibitor (TKI) targeting FGFRs, PLT-derived growth factor receptor β, and vascular endothelial growth factor receptor-2 by modulating the Akt/Mcl-1 axis to promote apoptosis in tumor cells [106]. Similarly, tanshinone IIA (TanIIA; C19H18O3) enhanced GEM sensitivity by blocking Ras/Raf/MERK/ERK and PI3K/AKT/mTOR signaling pathways by downregulating EGFR, IGF1R, and VEGFR expression [107]. Therefore, multi-targeted TKI inhibitors combined with GEM and/or immunosuppressants are promising future strategies for a comprehensive treatment of PDAC.

4.2 Chemokines

The CXC chemokine family (CXCL1-17) are small secreted proteins that induce GEM resistance by binding to their receptors (CXC chemokine receptor, CXCR) to regulate paracrine and autocrine pathways in PDAC mesenchymal cells that lead to GEM resistance [108, 109]. A typical example is the CXCL12/CXCR4 axis that activates FAK, ERK, and Akt pathways to promote survivin expression, thus, inducing GEM resistance [110, 111]. A phase IIa clinical study (NCT02826486) showed that the CXCR4 antagonist BL-8040 (motixafortide) in combination with pembrolizumab prolonged the overall survival of patients [8]. Notably, in PDAC cells, artemin can induce CXCR4 overexpression by activating ERK1/2 and Akt pathways and further through NF-κB. Therefore, targeted inhibition of artemin may provide new therapeutic ideas for chemotherapy sensitization [112].

5. Conclusions and Prospects

In the past decade, there has been little improvement in the overall survival rate of PDAC, and the primary treatment regimen is still GEM-based chemotherapy. Overcoming chemoresistance is a key aspect of improving prognosis. Although current clinical trials targeting TME stromal components have yielded unsatisfactory results, research on the TME continues to advance, and clinical trials combining GEM with multiple immunotherapy targets have shown encouraging results, although previous immune monotherapies have mostly been ineffective [113] (Table 1, Ref. [16, 18, 19, 24, 37, 49, 50, 51, 52, 53, 59, 60, 61, 65, 66, 67, 68, 69, 74, 80, 81, 82, 83, 84, 92, 93, 94, 97, 98, 100, 101, 102, 103, 104, 108, 109, 110, 111, 112]). In addition to immunotherapy, targeted therapies, gene editing, microbial therapies, and combination therapies have exhibited promising results in the clinical management of PDAC. It is crucial to highlight that while these cutting-edge treatments hold immense potential for PDAC patients, many of them are still undergoing early-stage clinical trials, and need further research and validation to establish broader clinical applicability.

Table 1.Potential targets and mechanisms of multiple cell and cytokine induction of gemcitabine (GEM) resistance in pancreatic ductal adenocarcinoma (PDAC) cells in the tumor microenvironment (TME).
Potential targets Drug resistance mechanism Drug/premedication Clinical trials Phases Experimental status Experimental result References
HIF-1 Upregulation of PLK4 and induction of differentiation of M-MDSCs into TAM NA NA NA NA NA [16, 74]
HiF-2 Reduces macrophage chemotaxis and M2-type polarization Belzutifan NCT04924075 Phase II Recruiting NA [18]
HIF-3 RhoC-ROCK1 signaling pathway NA NA NA NA NA [19]
FAK Substrate physical barrier Defactinib NCT02546531 Phase I completed Positive [24]
HA Adding IFP NA NA NA NA NA [37]
iCAFs Activation of JAK-STAT pathway Ruxolitinib NCT02117479, NCT02119663, NCT02767557 Phase III, Phase II completed Negative [49, 50, 51, 52, 53]
CSCs Increased Oct4, Nanog, Sox2 expression, induction of EMT NA NA NA NA NA [59, 60, 61]
lncRNA MACC1-AS1/PAX8/NOTCH1 NA NA NA NA NA [65, 66]
TAMs Activation of TLR4/IL-10 pathway, M2-type polarization PF-04136309 NCT01413022 Phase I/II completed Positive [67, 68, 69]
NETs Activation of IL-1β/EGFR/ERK pathway NA NA NA NA NA [80, 81, 82]
PD-1/PD-L1/CTLA-4 Immune response of Th1 lymphocytes NA NA NA NA NA [83, 84]
CTGF Phosphorylation of ERK mAbFG-3019 NCT02210559 Phase II completed Positive [92, 93, 94]
EGFR Remodeling of tumor stroma, Activation of STAT3 pathway Erlotinib NCT02694536 Phase III completed Negative [97, 98]
IGF-1R Activation of PI3K/Akt, mTOR, MEK/ERK pathway MK-0646 NCT00769483 Phase I/II completed Positive [100, 101, 102, 103, 104]
CXCL12/CXCR4 Activation of FAK, ERK and Akt pathways, Promotion of β-catenin, NF-κB pathway motixafortide NCT02826486 Phase II completed Positive [108, 109, 110, 111, 112]

CSC, cancer stem cells; CTGF, connective tissue growth factor; EGFR, epidermal growth factor receptor; EMT, epithelial-mesenchymal transition; FAK, focal adhesion kinase; HA, hyaluronan; iCAF, inflammatory cancer-associated fibroblast; IGF-1R, insulin-like growth factor 1 receptor; lncRNA, long non-coding RNA; MDSC, myeloid-derived suppressor cells; NET, neutrophil extracellular trap; TAM, tumor-associated macrophages; NA, not available.

Abbreviations

PDAC, pancreatic ductal adenocarcinoma; GEM, gemcitabine; TME, tumor microenvironment; PD-1, programmed death-1; PD-L1, programmed death ligand 1; CTLA-4, cytotoxic T lymphocyte associated antigen-4; HIF, hypoxia-inducible factor; CA, carbonic anhydrase; PLK4, polo-like kinase4; CSCs, cancer stem cells; RhoC, Ras homolog family member C; ROCK1, Rho-associated coiled-coil containing protein kinase 1; FAK, focal adhesion kinase; MT1-MMP, membrane type-1 matrix metalloproteinase; HMGA2, high mobility group protein A2; LM, Laminin; TG2, transglutaminase 2; NF-κB, nuclear factor kappa B; aPSCs, Activated pancreatic stellatecells; CAFs, cancer-associated fibroblasts; IL-1, interleukin-1; EMT, epithelial–mesenchymal transition; LncRNA, long non-coding RNA; TAMs, tumor-associated macrophages; MDSCs, Myeloid-derived suppressor cells; CXCL, chemokine ligands; CXCR, chemokine receptors; TILs, Tumor-infiltrating lymphocytes; PLTs, platelets; CTGF, Connective tissue growth factor; EGFR, epidermal growth factor receptor; STAT3, signal transducer and activator of transcription 3; IGF-1R, insulin-like growth factor 1 receptor; M-MDSCs, monocytic-myeloid-derived suppressor cells; MSCs, mesenchymal stem cells.

Author Contributions

ZTG and RT proposed the research topic of the article, TPZ and RN collected the literature and wrote the paper. XW and QTJ performed the image design and revision. All authors participated in the editorial revision of the manuscript. All authors read and approved the final manuscript. All authors participated fully in the work, assumed public responsibility for appropriate portions of the content, and agreed to be accountable for all aspects of the work for issues related to the accuracy or completeness of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work was funded by This work was funded by RT’s Provincial Health Ministry Major 2020, grant number A-2019-W153.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA: A Cancer Journal for Clinicians. 2021; 71: 7–33.
[2]
Long D, Chen K, Yang Y, Tian X. Unfolded protein response activated by endoplasmic reticulum stress in pancreatic cancer: potential therapeutical target. Frontiers in Bioscience-Landmark. 2021; 26: 1689–1696.
[3]
Muggilli M, Russell D, Zhou Z. Comparison of programmed death ligand 1 immunostaining for pancreatic ductal adenocarcinoma between paired cytological and surgical samples. Cytojournal. 2021; 18: 28.
[4]
Sun D, Cao M, Li H, He S, Chen W. Cancer burden and trends in China: a review and comparison with Japan and South Korea. Chinese Journal of Cancer Research. 2020; 32: 129–139.
[5]
Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. The Lancet. 2020; 395: 2008–2020.
[6]
von Ahrens D, Bhagat TD, Nagrath D, Maitra A, Verma A. The role of stromal cancer-associated fibroblasts in pancreatic cancer. Journal of Hematology & Oncology. 2017; 10: 76.
[7]
Neesse A, Algül H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015; 64: 1476–1484.
[8]
Bockorny B, Semenisty V, Macarulla T, Borazanci E, Wolpin BM, Stemmer SM, et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nature Medicine. 2020; 26: 878–885.
[9]
Neesse A, Bauer CA, Öhlund D, Lauth M, Buchholz M, Michl P, et al. Stromal biology and therapy in pancreatic cancer: ready for clinical translation? Gut. 2019; 68: 159–171.
[10]
Sohal DPS, Duong M, Ahmad SA, Gandhi NS, Beg MS, Wang-Gillam A, et al. Efficacy of Perioperative Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncology. 2021; 7: 421.
[11]
Saif MW, Lee Y, Kim R. Harnessing gemcitabine metabolism: a step towards personalized medicine for pancreatic cancer. Therapeutic Advances in Medical Oncology. 2012; 4: 341–346.
[12]
Gu Z, Du Y, Zhao X, Wang C. Tumor microenvironment and metabolic remodeling in gemcitabine‐based chemoresistance of pancreatic cancer. Cancer Letters. 2021; 521: 98–108.
[13]
McKeown SR. Defining normoxia, physoxia and hypoxia in tumours—implications for treatment response. The British Journal of Radiology. 2014; 87: 20130676.
[14]
Yuen A, Díaz B. The impact of hypoxia in pancreatic cancer invasion and metastasis. Hypoxia. 2014; 2: 91–106.
[15]
Schwartz DL, Bankson JA, Lemos R, Lai SY, Thittai AK, He Y, et al. Radiosensitization and Stromal Imaging Response Correlates for the HIF-1 Inhibitor PX-478 Given with or without Chemotherapy in Pancreatic Cancer. Molecular Cancer Therapeutics. 2010; 9: 2057–2067.
[16]
Mittal K, Kaur J, Sharma S, Sharma N, Wei G, Choudhary I, et al. Hypoxia Drives Centrosome Amplification in Cancer Cells via HIF1α-dependent Induction of Polo-Like Kinase 4. Molecular Cancer Research. 2022; 20: 596–606.
[17]
Chen S, Zhang J, Chen J, Wang Y, Zhou S, Huang L, et al. RER1 enhances carcinogenesis and stemness of pancreatic cancer under hypoxic environment. Journal of Experimental & Clinical Cancer Research. 2019; 38: 15.
[18]
Garcia Garcia CJ, Huang Y, Fuentes NR, Turner MC, Monberg ME, Lin D, et al. Stromal HIF2 Regulates Immune Suppression in the Pancreatic Cancer Microenvironment. Gastroenterology. 2022; 162: 2018–2031.
[19]
Zhou X, Guo X, Chen M, Xie C, Jiang J. HIF-3α Promotes Metastatic Phenotypes in Pancreatic Cancer by Transcriptional Regulation of the RhoC-ROCK1 Signaling Pathway. Molecular cancer research. 2018; 16: 124–134.
[20]
Gu L, Shan T, Ma Y, Tay FR, Niu L. Novel Biomedical Applications of Crosslinked Collagen. Trends in Biotechnology. 2019; 37: 464–491.
[21]
Seguin L, Desgrosellier JS, Weis SM, Cheresh DA. Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends in Cell Biology. 2015; 25: 234–240.
[22]
Lu J, Zhou S, Siech M, Habisch H, Seufferlein T, Bachem MG. Pancreatic stellate cells promote hapto-migration of cancer cells through collagen i-mediated signalling pathway. British Journal of Cancer. 2014; 110: 409–420.
[23]
Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nature Medicine. 2016; 22: 851–860.
[24]
Wang-Gillam A, Lim K, McWilliams R, Suresh R, Lockhart AC, Brown A, et al. Defactinib, Pembrolizumab, and Gemcitabine in Patients with Advanced Treatment Refractory Pancreatic Cancer: a Phase i Dose Escalation and Expansion Study. Clinical Cancer Research. 2022; 28: 5254–5262.
[25]
Dangi-Garimella S, Krantz SB, Barron MR, Shields MA, Heiferman MJ, Grippo PJ, et al. Three-Dimensional Collagen i Promotes Gemcitabine Resistance in Pancreatic Cancer through MT1-MMP–Mediated Expression of HMGA2. Cancer Research. 2011; 71: 1019–1028.
[26]
Summer H, Li O, Bao Q, Zhan L, Peter S, Sathiyanathan P, et al. HMGA2 exhibits dRP/AP site cleavage activity and protects cancer cells from DNA-damage-induced cytotoxicity during chemotherapy. Nucleic Acids Research. 2009; 37: 4371–4384.
[27]
Dangi-Garimella S, Sahai V, Ebine K, Kumar K, Munshi HG. Three-dimensional collagen I promotes gemcitabine resistance in vitro in pancreatic cancer cells through HMGA2-dependent histone acetyltransferase expression. PLoS ONE. 2013; 8: e64566.
[28]
Hallmann R, Hannocks M, Song J, Zhang X, Di Russo J, Luik A, et al. The role of basement membrane laminins in vascular function. the International Journal of Biochemistry & Cell Biology. 2020; 127: 105823.
[29]
Huang C, Chen J. Laminin-332 mediates proliferation, apoptosis, invasion, migration and epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma. Molecular Medicine Reports. 2021; 23: 11.
[30]
Lee J, Yakubov B, Ivan C, Jones DR, Caperell-Grant A, Fishel M, et al. Tissue Transglutaminase Activates Cancer-Associated Fibroblasts and Contributes to Gemcitabine Resistance in Pancreatic Cancer. Neoplasia. 2016; 18: 689–698.
[31]
Verma A, Mehta K. Tissue transglutaminase-mediated chemoresistance in cancer cells. Drug Resistance Updates. 2007; 10: 144–151.
[32]
Huanwen W, Zhiyong L, Xiaohua S, Xinyu R, Kai W, Tonghua L. Intrinsic chemoresistance to gemcitabine is associated with constitutive and laminin-induced phosphorylation of FAK in pancreatic cancer cell lines. Molecular Cancer. 2009; 8: 125.
[33]
Topalovski M, Brekken RA. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Letters. 2016; 381: 252–258.
[34]
Mukai M, Togawa A, Imamura F, Iwasaki T, Ayaki M, Mammoto T, et al. Sustained tyrosine-phosphorylation of FAK through Rho-dependent adhesion to fibronectin is essential for cancer cell migration. Anticancer Research. 2002; 22: 3175–3184.
[35]
Amrutkar M, Aasrum M, Verbeke CS, Gladhaug IP. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer. 2019; 19: 596.
[36]
Leppänen J, Lindholm V, Isohookana J, Haapasaari K, Karihtala P, Lehenkari PP, et al. Tenascin C, Fibronectin, and Tumor-Stroma Ratio in Pancreatic Ductal Adenocarcinoma. Pancreas. 2019; 48: 43–48.
[37]
DuFort C, DelGiorno K, Carlson M, Osgood R, Zhao C, Huang Z, et al. Interstitial Pressure in Pancreatic Ductal Adenocarcinoma is Dominated by a Gel-Fluid Phase. Biophysical Journal. 2016; 110: 2106–2119.
[38]
Zhao S, Chen C, Chang K, Karnad A, Jagirdar J, Kumar AP, et al. CD44 Expression Level and Isoform Contributes to Pancreatic Cancer Cell Plasticity, Invasiveness, and Response to Therapy. Clinical Cancer Research. 2016; 22: 5592–5604.
[39]
Koltai T, Reshkin SJ, Carvalho TMA, Cardone RA. Targeting the Stromal Pro-Tumoral Hyaluronan-CD44 Pathway in Pancreatic Cancer. International Journal of Molecular Sciences. 2021; 22: 3953.
[40]
Cheng XB, Sato N, Kohi S, Koga A, Hirata K. 4-Methylumbelliferone inhibits enhanced hyaluronan synthesis and cell migration in pancreatic cancer cells in response to tumor-stromal interactions. Oncology Letters. 2018; 15: 6297–6301.
[41]
Apte MV, Wilson JS, Lugea A, Pandol SJ. A Starring Role for Stellate Cells in the Pancreatic Cancer Microenvironment. Gastroenterology. 2013; 144: 1210–1219.
[42]
Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan GW, Korsten MA, et al. Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture. Gut. 1998; 43: 128–133.
[43]
Erkan M, Adler G, Apte MV, Bachem MG, Buchholz M, Detlefsen S, et al. StellaTUM: current consensus and discussion on pancreatic stellate cell research. Gut. 2012; 61: 172–178.
[44]
Liu SL, Cao SG, Li Y, Sun B, Chen D, Wang DS, et al. Pancreatic stellate cells facilitate pancreatic cancer cell viability and invasion. Oncology Letters. 2019; 17: 2057–2062.
[45]
Zhao X, Li Z, Gu Z. A New Era: Tumor Microenvironment in Chemoresistance of Pancreatic Cancer. Journal of Cancer Science and Clinical Therapeutics. 2022; 6: 61–86.
[46]
Haniffa MA, Collin MP, Buckley CD, Dazzi F. Mesenchymal stem cells: the fibroblasts’ new clothes? Haematologica. 2009; 94: 258–263.
[47]
Hosein AN, Brekken RA, Maitra A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nature Reviews Gastroenterology & Hepatology. 2020; 17: 487–505.
[48]
Huang H, Brekken RA. Recent advances in understanding cancer-associated fibroblasts in pancreatic cancer. American Journal of Physiology-Cell Physiology. 2020; 319: C233–C243.
[49]
Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. Journal of Experimental Medicine. 2017; 214: 579–596.
[50]
Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFβ to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discovery. 2019; 9: 282–301.
[51]
Hurwitz H, Van Cutsem E, Bendell J, Hidalgo M, Li C, Salvo MG, et al. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Investigational New Drugs. 2018; 36: 683–695.
[52]
Neumann CCM, von Hörschelmann E, Reutzel-Selke A, Seidel E, Sauer IM, Pratschke J, et al. Tumor–stromal cross-talk modulating the therapeutic response in pancreatic cancer. Hepatobiliary & Pancreatic Diseases International. 2018; 17: 461–472.
[53]
McAndrews KM, Chen Y, Darpolor JK, Zheng X, Yang S, Carstens JL, et al. Identification of Functional Heterogeneity of Carcinoma-Associated Fibroblasts with Distinct IL6-Mediated Therapy Resistance in Pancreatic Cancer. Cancer Discovery. 2022; 12: 1580–1597.
[54]
Qi R, Bai Y, Li K, Liu N, Xu Y, Dal E, et al. Cancer-associated fibroblasts suppress ferroptosis and induce gemcitabine resistance in pancreatic cancer cells by secreting exosome-derived ACSL4-targeting miRNAs. Drug Resistance Updates. 2023; 68: 100960.
[55]
Ye Z, Zhuo Q, Hu Q, Xu X, Mengqi L, Zhang Z, et al. FBW7-NRA41-SCD1 axis synchronously regulates apoptosis and ferroptosis in pancreatic cancer cells. Redox Biology. 2021; 38: 101807.
[56]
Sheta M, Taha EA, Lu Y, Eguchi T. Extracellular Vesicles: New Classification and Tumor Immunosuppression. Biology (Basel). 2023; 12: 110.
[57]
Ishiwata T, Matsuda Y, Yoshimura H, Sasaki N, Ishiwata S, Ishikawa N, et al. Pancreatic cancer stem cells: features and detection methods. Pathology & Oncology Research. 2018; 24: 797–805.
[58]
Lee CJ, Dosch J, Simeone DM. Pancreatic Cancer Stem Cells. Journal of Clinical Oncology. 2008; 26: 2806–2812.
[59]
Wang H, Feng W, Chen W, He J, Min J, Liu Y, et al. Methyl-CpG-binding domain 3 inhibits stemness of pancreatic cancer cells via Hippo signaling. Experimental Cell Research. 2020; 393: 112091.
[60]
Lu Y, Zhu H, Shan H, Lu J, Chang X, Li X, et al. Knockdown of Oct4 and Nanog expression inhibits the stemness of pancreatic cancer cells. Cancer Letters. 2013; 340: 113–123.
[61]
Sun L, Cao J, Chen K, Cheng L, Zhou C, Yan B, et al. Betulinic acid inhibits stemness and EMT of pancreatic cancer cells via activation of AMPK signaling. International Journal of Oncology. 2019; 54: 98–110.
[62]
Luo Z, Li Y, Zuo M, Liu C, Tian W, Yan D, et al. Effect of NR5A2 inhibition on pancreatic cancer stem cell (CSC) properties and epithelial-mesenchymal transition (EMT) markers. Molecular Carcinogenesis. 2017; 56: 1438–1448.
[63]
Wang L, Dong P, Wang W, Huang M, Tian B. Gemcitabine treatment causes resistance and malignancy of pancreatic cancer stem-like cells via induction of lncRNA HOTAIR. Experimental and Therapeutic Medicine. 2017; 14: 4773–4780.
[64]
Deng S, Wang J, Zhang L, Li J, Jin Y. LncRNA HOTAIR Promotes Cancer Stem-Like Cells Properties by Sponging miR-34a to Activate the JAK2/STAT3 Pathway in Pancreatic Ductal Adenocarcinoma. OncoTargets and Therapy. 2021; 14: 1883–1893.
[65]
Zhang Z, Han H, Rong Y, Zhu K, Zhu Z, Tang Z, et al. Hypoxia potentiates gemcitabine-induced stemness in pancreatic cancer cells through AKT/Notch1 signaling. Journal of Experimental & Clinical Cancer Research. 2018; 37: 291.
[66]
Yang MH, Zhao L, Wang L, Ou-Yang W, Hu SS, Li WL, et al. Nuclear lncRNA HOXD-AS1 suppresses colorectal carcinoma growth and metastasis via inhibiting HOXD3-induced integrin β3 transcriptional activating and MAPK/AKT signalling. Molecular Cancer. 2019; 18: 31.
[67]
Noy R, Pollard J. Tumor-Associated Macrophages: from Mechanisms to Therapy. Immunity. 2014; 41: 49–61.
[68]
Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, et al. Inflammatory Monocyte Mobilization Decreases Patient Survival in Pancreatic Cancer: A Role for Targeting the CCL2/CCR2 Axis. Clinical Cancer Research. 2013; 19: 3404–3415.
[69]
Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. The Lancet Oncology. 2016; 17: 651–662.
[70]
Liu C, Xu J, Shi X, Huang W, Ruan T, Xie P, et al. M2-polarized tumor-associated macrophages promoted epithelial–mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Laboratory Investigation. 2013; 93: 844–854.
[71]
Bergenfelz C, Leandersson K. The Generation and Identity of Human Myeloid-Derived Suppressor Cells. Frontiers in Oncology. 2020; 10: 109.
[72]
Tcyganov E, Mastio J, Chen E, Gabrilovich DI. Plasticity of myeloid-derived suppressor cells in cancer. Current Opinion in Immunology. 2018; 51: 76–82.
[73]
Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends in Immunology. 2016; 37: 208–220.
[74]
Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. The Journal of Experimental Medicine. 2010; 207: 2439–2453.
[75]
Hartwig T, Montinaro A, von Karstedt S, Sevko A, Surinova S, Chakravarthy A, et al. The TRAIL-Induced Cancer Secretome Promotes a Tumor-Supportive Immune Microenvironment via CCR2. Molecular Cell. 2017; 65: 730–742.e5.
[76]
Kumar V, Cheng P, Condamine T, Mony S, Languino LR, McCaffrey JC, et al. CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation. Immunity. 2016; 44: 303–315.
[77]
Nywening TM, Belt BA, Cullinan DR, Panni RZ, Han BJ, Sanford DE, et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut. 2018; 67: 1112–1123.
[78]
Chao T, Furth EE, Vonderheide RH. CXCR2-Dependent Accumulation of Tumor-Associated Neutrophils Regulates T-cell Immunity in Pancreatic Ductal Adenocarcinoma. Cancer Immunology Research. 2016; 4: 968–982.
[79]
Steele CW, Karim SA, Leach JDG, Bailey P, Upstill-Goddard R, Rishi L, et al. CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma. Cancer Cell. 2016; 29: 832–845.
[80]
Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011; 117: 953–959.
[81]
Park J, Wysocki RW, Amoozgar Z, Maiorino L, Fein MR, Jorns J, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Science Translational Medicine. 2016; 8: 361ra138.
[82]
Jin W, Yin H, Li H, Yu X, Xu H, Liu L. Neutrophil extracellular DNA traps promote pancreatic cancer cells migration and invasion by activating EGFR/ERK pathway. Journal of Cellular and Molecular Medicine. 2021; 25: 5443–5456.
[83]
Gong J, Hendifar A, Tuli R, Chuang J, Cho M, Chung V, et al. Combination systemic therapies with immune checkpoint inhibitors in pancreatic cancer: overcoming resistance to single-agent checkpoint blockade. Clinical and Translational Medicine. 2018; 7: 32.
[84]
Ho TTB, Nasti A, Seki A, Komura T, Inui H, Kozaka T, et al. Combination of gemcitabine and anti-PD-1 antibody enhances the anticancer effect of M1 macrophages and the Th1 response in a murine model of pancreatic cancer liver metastasis. Journal for ImmunoTherapy of Cancer. 2020; 8: e001367.
[85]
Wartenberg M, Cibin S, Zlobec I, Vassella E, Eppenberger-Castori S, Terracciano L, et al. Integrated Genomic and Immunophenotypic Classification of Pancreatic Cancer Reveals Three Distinct Subtypes with Prognostic/Predictive Significance. Clinical Cancer Research. 2018; 24: 4444–4454.
[86]
Brouwer T, Ijsselsteijn M, Oosting J, Ruano D, van der Ploeg M, Dijk F, et al. A Paradoxical Role for Regulatory T Cells in the Tumor Microenvironment of Pancreatic Cancer. Cancers. 2022; 14: 3862.
[87]
Zhang Y, Lazarus J, Steele NG, Yan W, Lee H, Nwosu ZC, et al. Regulatory T-cell Depletion Alters the Tumor Microenvironment and Accelerates Pancreatic Carcinogenesis. Cancer Discovery. 2020; 10: 422–439.
[88]
Bian X, Yin S, Yang S, Jiang X, Wang J, Zhang M, et al. Roles of platelets in tumor invasion and metastasis: a review. Heliyon. 2022; 8: e12072.
[89]
Geddings JE, Hisada Y, Boulaftali Y, Getz TM, Whelihan M, Fuentes R, et al. Tissue factor–positive tumor microvesicles activate platelets and enhance thrombosis in mice. Journal of Thrombosis and Haemostasis. 2016; 14: 153–166.
[90]
Labelle M, Begum S, Hynes R. Direct Signaling between Platelets and Cancer Cells Induces an Epithelial-Mesenchymal-Like Transition and Promotes Metastasis. Cancer Cell. 2011; 20: 576–590.
[91]
Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, et al. Epithelial to Mesenchymal Transition Contributes to Drug Resistance in Pancreatic Cancer. Cancer Research. 2009; 69: 5820–5828.
[92]
Aoyama E, Kubota S, Takigawa M. CCN2/CTGF binds to fibroblast growth factor receptor 2 and modulates its signaling. FEBS Letters. 2012; 586: 4270–4275.
[93]
Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proceedings of the National Academy of Sciences. 2013; 110: 12325–12330.
[94]
Picozzi V, Alseidi A, Winter J, Pishvaian M, Mody K, Glaspy J, et al. Gemcitabine/nab-paclitaxel with pamrevlumab: a novel drug combination and trial design for the treatment of locally advanced pancreatic cancer. ESMO Open. 2020; 5: e000668.
[95]
Li J, Yuan S, Norgard RJ, Yan F, Sun YH, Kim I, et al. Epigenetic and Transcriptional Control of the Epidermal Growth Factor Receptor Regulates the Tumor Immune Microenvironment in Pancreatic Cancer. Cancer Discovery. 2021; 11: 736–753.
[96]
Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. Journal of Clinical Oncology. 2007; 25: 1960–1966.
[97]
Dosch AR, Dai X, Reyzer ML, Mehra S, Srinivasan S, Willobee BA, et al. Combined Src/EGFR Inhibition Targets STAT3 Signaling and Induces Stromal Remodeling to Improve Survival in Pancreatic Cancer. Molecular Cancer Research. 2020; 18: 623–631.
[98]
Sinn M, Bahra M, Liersch T, Gellert K, Messmann H, Bechstein W, et al. CONKO-005: Adjuvant Chemotherapy with Gemcitabine Plus Erlotinib Versus Gemcitabine alone in Patients after R0 Resection of Pancreatic Cancer: a Multicenter Randomized Phase III Trial. Journal of Clinical Oncology. 2017; 35: 3330–3337.
[99]
Humbert M, Casteran N, Letard S, Hanssens K, Iovanna J, Finetti P, et al. Masitinib combined with standard gemcitabine chemotherapy: in vitro and in vivo studies in human pancreatic tumour cell lines and ectopic mouse model. PLoS ONE. 2010; 5: e9430.
[100]
Ireland L, Santos A, Ahmed MS, Rainer C, Nielsen SR, Quaranta V, et al. Chemoresistance in Pancreatic Cancer is Driven by Stroma-Derived Insulin-Like Growth Factors. Cancer Research. 2016; 76: 6851–6863.
[101]
Steelman LS, Chappell WH, Abrams SL, Kempf CR, Long J, Laidler P, et al. Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging. 2011; 3: 192–222.
[102]
Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ, Jr, Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Annals of Oncology. 2012; 23: 2834–2842.
[103]
Fuchs CS, Azevedo S, Okusaka T, Van Laethem JL, Lipton LR, Riess H, et al. A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: the GAMMA trial. Annals of Oncology. 2015; 26: 921–927.
[104]
Abdel-Wahab R, Varadhachary GR, Bhosale PR, Wang X, Fogelman DR, Shroff RT, et al. Randomized, phase i/II study of gemcitabine plus IGF-1R antagonist (MK-0646) versus gemcitabine plus erlotinib with and without MK-0646 for advanced pancreatic adenocarcinoma. Journal of Hematology and Oncology. 2018; 11: 71.
[105]
Ma WW, Xie H, Fetterly G, Pitzonka L, Whitworth A, LeVea C, et al. A Phase Ib Study of the FGFR/VEGFR Inhibitor Dovitinib with Gemcitabine and Capecitabine in Advanced Solid Tumor and Pancreatic Cancer Patients. American Journal of Clinical Oncology. 2019; 42: 184–189.
[106]
Zhang H, Hylander BL, LeVea C, Repasky EA, Straubinger RM, Adjei AA, et al. Enhanced FGFR signalling predisposes pancreatic cancer to the effect of a potent FGFR inhibitor in preclinical models. British Journal of Cancer. 2014; 110: 320–329.
[107]
Su CC. Tanshinone IIA can inhibit MiaPaCa‑2 human pancreatic cancer cells by dual blockade of the Ras/Raf/MEK/ERK and PI3K/AKT/mTOR pathways. Oncology Reports. 2018; 40: 3102–3111.
[108]
Lee N, Nikfarjam M, He H. Functions of the CXC ligand family in the pancreatic tumor microenvironment. Pancreatology. 2018; 18: 705–716.
[109]
Sleightholm RL, Neilsen BK, Li J, Steele MM, Singh RK, Hollingsworth MA, et al. Emerging roles of the CXCL12/CXCR4 axis in pancreatic cancer progression and therapy. Pharmacology & Therapeutics. 2017; 179: 158–170.
[110]
Singh S, Srivastava SK, Bhardwaj A, Owen LB, Singh AP. CXCL12–CXCR4 signalling axis confers gemcitabine resistance to pancreatic cancer cells: a novel target for therapy. British Journal of Cancer. 2010; 103: 1671–1679.
[111]
Singh AP, Arora S, Bhardwaj A, Srivastava SK, Kadakia MP, Wang B, et al. CXCL12/CXCR4 protein signaling axis induces sonic hedgehog expression in pancreatic cancer cells via extracellular regulated kinase- and Akt kinase-mediated activation of nuclear factor κB: implications for bidirectional tumor-stromal interactions. Journal of Biological Chemistry. 2012; 287: 39115–39124.
[112]
Wang J, Wang H, Cai J, Du S, Xin B, Wei W, et al. Artemin regulates CXCR4 expression to induce migration and invasion in pancreatic cancer cells through activation of NF-κB signaling. Experimental Cell Research. 2018; 365: 12–23.
[113]
Hilmi M, Bartholin L, Neuzillet C. Immune therapies in pancreatic ductal adenocarcinoma: Where are we now? World Journal of Gastroenterology. 2018; 24: 2137–2151.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top