Next Article in Journal
Cell Type-Specific Anti-Adhesion Properties of Peritoneal Cell Treatment with Plasma-Activated Media (PAM)
Next Article in Special Issue
Alloferon Affects the Chemosensitivity of Pancreatic Cancer by Regulating the Expression of SLC6A14
Previous Article in Journal
Tandem Repeat Diversity in Two Closely Related Hamster Species—The Chinese Hamster (Cricetulus griseus) and Striped Hamster (Cricetulus barabensis)
Previous Article in Special Issue
Triazole Modified Tetraiodothyroacetic Acid Conjugated to Polyethylene Glycol, a Thyrointegrin αvβ3 Antagonist as a Radio- and Chemo-Sensitizer in Pancreatic Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Trinity: Interplay among Cancer Cells, Fibroblasts, and Immune Cells in Pancreatic Cancer and Implication of CD8+ T Cell-Orientated Therapy

1
National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
2
Division of Hematology & Oncology, Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 804, Taiwan
3
Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
*
Authors to whom correspondence should be addressed.
Submission received: 5 March 2022 / Revised: 15 April 2022 / Accepted: 17 April 2022 / Published: 18 April 2022
(This article belongs to the Special Issue Pancreatic Cancer: From Mechanisms to Therapeutic Approaches)

Abstract

:
The microenvironment in tumors is complicated and is constituted by different cell types and stromal proteins. Among the cell types, the abundance of cancer cells, fibroblasts, and immune cells is high and these cells work as the “Trinity” in promoting tumorigenesis. Although unidirectional or bidirectional crosstalk between two independent cell types has been well characterized, the multi-directional interplays between cancer cells, fibroblasts, and immune cells in vitro and in vivo are still unclear. We summarize recent studies in addressing the interaction of the “Trinity” members in the tumor microenvironment and propose a functional network for how these members communicate with each other. In addition, we discuss the underlying mechanisms mediating the interplay. Moreover, correlations of the alterations in the distribution and functionality of cancer cells, fibroblasts, and immune cells under different circumstances are reviewed. Finally, we point out the future application of CD8+ T cell-oriented therapy in the treatment of pancreatic cancer.

Graphical Abstract

1. Introduction

Pancreatic cancer is a deadly malignancy around the world according to the latest global reports [1,2] and is expected to be the second leading cause of cancer-related death in 2030 [2]. Surgery [3], radiotherapy [4], chemotherapy [5,6], and immunotherapy [7,8] are available for pancreatic cancer treatment; however, recurrence and metastasis occur [9,10,11,12,13,14,15] frequently after treatment [16]. Therefore, the identification of additional molecular targets and development of novel therapeutic strategies are urgently needed. Pancreatic cancer cells exhibit multiple dysregulations in oncogenes [17,18,19] and tumor suppressor genes [18,20,21,22] to enhance tumor initiation, promotion, and progression. While corresponding targeted therapies work well in some cancer types [23,24,25,26,27], the fibrotic nature of pancreatic cancer makes it hard for drugs to access [28,29]. This property and the immunologically “cold” tumor microenvironment [30,31] render immunotherapy as rarely effective [7,8,32]. Overcoming these barriers may provide a new direction for the design of targeted therapy [33,34,35]. In the pancreatic tumor microenvironment, fibroblasts and immune cells call for the attention of researchers due to their abundance [35,36] and functional diversity [33,34], which we introduce below.

2. Fibroblasts and Immune Cells in Pancreatic Cancer

2.1. Fibroblast

Fibroblasts in pancreatic cancer can be divided into not only quiescent/activated statuses but also different subtypes, with the latter based on single cell RNA sequencing on patient specimens [37]. The quiescent status of fibroblasts in pancreatic cancer is transformed by external signals [38], leading to morphological change as well as factor secretion such as cytokines, chemokines, and extracellular components [38,39]. The subtype differences in fibroblasts around the pancreas exist before tumor formation [40]. When pancreatic pre-cancerous lesions develop, the fibroblasts surrounding them may receive differential clues due to spatial distribution [35,36,41,42,43,44] and give rise to different phenotypes such as myofibroblast-like, inflammatory, and antigen-presenting. The subtype classifications are further supported by single cell RNA sequencing on mouse pancreatic tumors [45,46]. In three independent single cell RNA sequencing studies [37,45,46], the myofibroblast-like subtype and inflammatory subtype existed in pancreatic bulky tumors, responded to diverse signals, and modulated disease progression differentially. As these subtypes are interchangeable [43,46] and have different prognostic impacts [47], targeting fibroblasts in pancreatic cancer should be considered carefully [35,36,41,42,48,49,50,51], especially with the previous in vivo findings that fibroblast depletion in pancreatic cancer speeded up cancer metastasis [52,53].
Myofibrotic cancer-associated fibroblasts (myCAFs) and inflammatory CAFs (iCAFs) are frequently identified in pancreatic cancer [43,45,46,54,55] and other cancer types [56,57,58,59,60]. In spontaneous pancreatic cancer mouse models, myCAFs and iCAFs exhibit different genetic profiles and oncogenic activities [49]. A myCAF functions in ECM remodeling, expresses α-SMA, localizes near cancer cells, and responds to transforming growth factor β (TGFβ) [36,49,60,61,62,63,64,65,66]. An iCAF acts in factor secretion, expresses interleukin-6 (IL6), localizes distantly from tumor, and responds to IL1 [49,60,62,63,64,65,66]. While an iCAF is tumor-promoting, the comprehensive influence of a myCAF on pancreatic cancer development is still under debate [49,62,65]. What makes CAFs in pancreatic cancer more complicated is their interchangeability [36].
As well as the main CAF subtypes, including myCAF and iCAF, CAF heterogenicity is another important issue in pancreatic cancer development. This heterogenicity starts from the origins of CAFs, which may be resident fibroblasts, pancreatic stellate cells, adipocytes, pericytes, endothelial cells, and mesenchymal stem cells. In terms of subtype heterogenicity, the antigen-presenting CAF (apCAF) [38,67,68] is also present. Common markers for a myCAF include α-SMA [38,40,67] and transgelin [38,40]. For an iCAF, these markers are IL6 [38,40,67] and CXCL12 [38,40]. For the apCAF, the markers are MHCII [40,68] and Saa3 [38,40]. The myCAF in pancreatic cancer associates with increased ECM deposition [69], decreased T cell infiltration [67,68], and altered tumor development [67,68,69]. The iCAF links to increased pancreatic cancer stemness [67], drug resistance [67], tumorigenesis [68,69], and immunosuppression [68,69]. The apCAF mostly associates with antigen presentation [68,69].

2.2. Immune Cell

The immune cell is another critical component in the pancreatic cancer microenvironment [29,33,70,71,72,73,74,75,76,77]; though, with its immunologically cold nature partially attributed to desmoplasia, pancreatic cancer patients with CD8+ lymphocyte infiltration have a better prognosis [78,79]. For other lymphoid cells, the role of CD4+ lymphocytes in pancreatic cancer heavily depends on the helper cell type. The existence of FoxP3+ regulatory T lymphocytes (Treg) are associated with a higher risk for pancreatic cancer [78]. Conversely, the amount of B lymphocytes predicts better pancreatic cancer prognosis [78,79]. The presence of nature killer (NK) cells also benefits patient survival [70] as long as their cytotoxicity is retained [70,77,79]. In myeloid cells, mast cells associate with pancreatic cancer growth and angiogenesis [80,81,82]. A higher ratio of neutrophils/lymphocytes links to a poor clinical outcome of pancreatic cancer patients in meta-analyses [83,84]. On the contrary, macrophages affect pancreatic cancer cell behaviors in a polarization-dependent manner. M2 macrophages, with antiinflammatory property, frequently block immune surveillance and promote cancer growth. In contrast, M1 macrophages with pro-inflammatory character elicit immunity to fight against tumor. However, dissection of tumor-associated macrophages (TAM) in pancreatic cancer revealed that these macrophages may display features of both polarization types. These phenomena were widely observed and reviewed in detail [34,71,73,76,79,85]. While the mechanism by which dendritic cells influence pancreatic tumors is still unclear, its potential anticancer efficiency via vaccination has been observed for a long time [86,87,88,89,90,91] and has been recently revisited [92,93,94].
In addition to the M2 macrophage, myeloid-derived suppressor cells (MDSC) play an important role in pancreatic tumorigenesis [95]. These suppressor cells include monocytic (M-MDSC) and polymorphonuclear cells (PMN-MDSC). They arise from the impaired differentiation of myeloid progenitor cells during myelopoiesis, and this impairment is commonly observed during tumorigenesis [96]. For M-MDSC, Trovato et al. found both M-MDSC and PMN-MDSC in pancreatic cancer tissues, negatively correlated with T cell population [97]. In the peripheral blood mononuclear cell (PBMC) subset, M-MDSC percentage was higher in pancreatic cancer patients than in healthy donors. Using whole blood samples in the validation cohort, the authors found CD14+ M-MDSC still displayed greater suppression on T cell proliferation than CD66b+ PMN-MDSC did. This resulted from STAT3 activation and subsequent arginase expression, and could be counteracted by arginase inhibition. For PMN-MDSC, Peng et al. reported that, in an H7 orthotopic pancreatic cancer mouse model, apolipoprotein A-I mimetic peptide L-4F decreased tumorigenesis via PMN-MDSC suppression [98]. Increments in CD8+ T cells and IFNγ in the tumor and spleen after L-4F treatment were observed, compared to a control Sc-4F treatment group. Mechanistically, this suppression was achieved by inhibitions of ROS production and STAT3 phosphorylation. Conversely, Kramer et al. showed that PMN-MDSC in pancreatic cancer was associated with the increased expression of Wnt inhibitor Dickkopf-1 (Dkk1) [99].

3. Mechanisms Underlying the Interplay among Pancreatic Cancer Cells, Fibroblasts, and Immune Cells

3.1. Cytokine and Chemokine

What makes the situation even more complicated is the interplay among pancreatic cancer cells, fibroblasts, and immune cells. A change in one component affects the whole picture in a pancreatic tumor [33,34,35,36,48,50,51,70,73,74,75,77,100]. Mechanistically, cytokines and chemokines secreted from pancreatic cancer cells, fibroblasts, or immune cells modulate the behaviors of the other two populations [35,36,49]. However, only limited targets have been identified in these multi-directional interplays [101] (Table 1 and Figure 1). Mucciolo et al. utilized knockout mice and antibody treatment to explore the role of IL17 in the pancreatic tumor microenvironment and found the loss of this cytokine modulated extracellular matrix (ECM) composition and increased CD3+ cell infiltration [101]. In pancreatic cancer cells, mutant Kras elicited IL17 expression [101,102], possibly via the STAT3 signaling pathway [103,104]. IL17, in turn, worked on fibroblasts to reshape the ECM [101,105] and on immune cells to block their antitumor function [106,107,108]. With a similar approach, Zhang et al. revealed that regulatory T cells’ (Treg) depletion in a transgenic pancreatic cancer mouse model surprisingly accelerated pancreatic tumorigenesis via inactivation of the PDGFRβ+ SMA+ myofibroblast [109]. In addition, compensation by other pro-tumor myeloid-derived suppressor cells (MDSC) was observed once Treg were depleted. The results of RNA sequencing pointed out that in this process, many C-C chemokine receptor type 1 (CCR1) ligands were upregulated, and CCR1 inhibition retarded pancreatic tumorigenesis, resulting from Treg depletion. In terms of fibroblasts, Feig et al. showed that fibroblast activation of protein-positive (FAP)+ cancer-associated fibroblasts (CAFs) participated in pancreatic cancer formation, and the depletion of FAP+ fibroblasts in a transgenic pancreatic cancer mouse model decreased tumor development [110]. This might be through C-X-C motif chemokine ligand 12 (CXCL12) expression in FAP+ fibroblasts and its effect on T cell exclusion, specifically via high mobility group box 1 (HMGB1) [111,112]. Accordingly, loss of the CD4/CD8 cells reversed the effect of FAP+ fibroblast depletion on tumor growth. In addition, the inhibition of CXCR4, the cognate receptor for CXCL12, by AMD3100 improved anti-PD-L1 efficiency.

3.2. Extracellular Signal

In addition to cytokines and chemokines, molecules such as extracellular components and stress signals also participate [113,114] in the interaction among the “Trinity”. Chen et al. demonstrated that the loss of collagen I in α-SMA+ fibroblasts enhanced pancreatic cancer progression via CXCL5 upregulation in cancer cells and the subsequent activation of MDSC [113]. As CXCL5 is one of the most upregulated factors in a pancreatic cancer mouse model with collagen I knockout, the authors proposed that this ECM alteration stimulated SOX9 transcription activation [113,115] in cancer cells [113]. CXCL5 then recruited and polarized MDSC into a pro-tumor type to enhance the growth of pancreatic cancer cells. Furthermore, MDSC suppressed functions of T and B cells via arginine depletion. These effects on ECM alteration and immune suppression via cancer cells and MDSC emphasized the importance of α-SMA+ fibroblasts in pancreatic tumorigenesis by affecting collagen deposition. In terms of stress signals, Zhang et al. established primary cultures of fibroblasts from human pancreatic cancer tissues, and found they affected monocytes in a reactive oxygen species (ROS)-dependent manner [114]. With either a CAF-conditioned medium or direct CAF co-culture, monocytes displayed increased M2 polarization. This phenomenon was associated with upregulated ROS production in monocytes, and both ROS amount and M2 polarization were inhibited by antioxidants. CAFs increased ROS in monocytes via M-CSF, which is specific to cancer-associated fibroblasts as normal fibroblasts rarely produce this factor. Moreover, the CAF-stimulated monocytes are capable of inducing Stat3 and Akt phosphorylation in pancreatic cancer cells and subsequently increasing their growth. Pancreatic cancer cells also express a variety of factors including VEGF [116] to activate Stat3 and Akt [117,118], and subsequently modulate macrophage behavior. This study suggests the complex interaction among cancer cells, fibroblasts, and immune cells during pancreatic cancer development.

3.3. Pathway Modulation

Researchers also utilized additional approaches to dissect the interplay among the “Trinity” members in pancreatic tumors, such as inhibitor treatment and gene delivery. Zhao et al. applied M-CPA/PTX, a polymeric micelle-based nanoformulation of sonic hedgehog inhibitor (cyclopamine, CPA) and paclitaxel (PTX), to modulate stroma in pancreatic cancer [119]. M-CPA/PTX increased intratumoral vasculature density to benefit CD8+ T cell infiltration without altering the ECM and fibroblasts. In the bulky tumor, VEGFR2 and Shh phosphorylations were suppressed by M-CPA/PTX. Moreover, M-CPA/PTX improved anti-PD-1 efficiency via IFN-γ from CD8+ T cells. Zhou et al. directly applied gene delivery targeting relaxin to hamper fibrosis in cancers [120]. Relaxin is an insulin-like peptide hormone with diverse biological functions but is known for fibrosis resolution therapeutically [120,121]. The authors utilized this advantage to test its effect on pancreatic desmoplasia by targeting macrophages as they express the relaxin receptor (relaxin family peptide receptor type 1, RXFP1). They found that local relaxin plasmid (pRLN) delivery in the format of aminoethyl anisamide (AEAA)-conjugated lipid-protamine-DNA (LPD) nanoparticle induced macrophage recruitment [121]. These macrophages uptook relaxin and activated themselves to express cytokines and enzymes for fibrosis depletion. This phenomenon is accompanied with (1) a decrease in α-SMA+ fibroblasts; (2) a reduction in collagen I; (3) an increase in antitumor CD8+ T and NK cells; (4) attenuation in pro-tumor Treg. Furthermore, pRLN benefited anti-PD-L1 efficiency, even though pRLN itself already displayed a better treatment effect in vivo. Although the advantage of stroma targeting in pancreatic cancer is still under debate [52,53], the works from Zhao et al. [119] and Zhou et al. [120] supported the notion that the entry of CD8+ T cells has a higher benefit than the depletion of fibroblasts in this process, as CD8+ T cell infiltration is the main determinant of treatment efficiency with or without fibroblast alteration.

4. Correlation of Alterations in the “Trinity” Population in Preclinical Model and Clinical Setting

In addition to discussing the mechanisms of the multi-functional interplay between cancer cells, fibroblasts, and immune cells, we also summarized the correlation of alterations of the “Trinity” members under different circumstances (Table 2). Pancreatic tumor tissues showed increased focal adhesion kinase (FAK) activation and ECM accumulation. Jiang et al. reported that the FAK inhibitor accordingly reversed ECM alteration and suppressed tumorigenesis in animals. The authors demonstrated that MDSC infiltration was suppressed by the FAK inhibitor. In this preclinical model, co-reduction in fibroblasts and MDSCs was found after FAK inhibition, suggesting a role of FAK in controlling the population of these two cell types in pancreatic cancer [122].
Gorchs et al. isolated fibroblasts from human pancreatic tumors and found that CAFs expressed more PD-L1 and PD-L2 compared to normal skin fibroblast [123]. The co-culture of CAFs resulted in the suppression of T cell proliferation in a prostaglandin E2 (PGE2)-dependent manner. Consequently, PGE2 inhibition by the COX2 inhibitor restored this phenotypic change. For those T cells still proliferating, CAFs also hindered their function by (1) increasing co-inhibitory molecules and (2) decreasing functionality, suggesting the antagonism of cell number and biological activity between fibroblasts and T cells.
Ireland et al. showed stromal Gas6 inhibition by neutralizing antibody suppressed metastasis in a spontaneous pancreatic cancer mouse model [124]. To find the origin of Gas6 secretion, the authors used a cell sorting approach and confirmed macrophages and fibroblasts were the main Gas6 producers. Additionally, immunohistochemical (IHC) staining on serial sections further verified the activation of downstream AXL signaling [125,126]. A Gas6 blockade decreased the epithelial–mesenchymal transition (EMT) in pancreatic cancer cells, while neither myeloid cells nor T cells were affected. However, Gas6 inhibition reduced NK cell recruitment. These results suggest that fibroblasts and macrophages modulated cancer cells in a unidirectional way via Gas6.
The correlation of the ECM and immune cells in clinical samples has also been investigated. Kamionka et al. showed the collagen composition in pancreatic tumor tissues was not significantly linked with immune cell infiltration, suggesting collagen organization has a minor impact on T cell distribution [127].
To understand the associations of different cell types in the tumor microenvironment, Lawal et al. applied databases to analyze multi-omics results of multiple cancers emphasizing the STAT3/CDK2/4/6 axis [128]. As this axis is highly expressed across various cancer types, the authors correlated it to epigenetics and immunity, and found an association between CAFs and STAT3 in pancreatic cancer. With a similar high-throughput concept, MacNeil et al. surveyed the prognostic impact of CAFs and tumor-infiltrating lymphocytes (TIL) in pancreatic cancer patients [129]. Multivariate analysis showed that CD8+ T cells of TIL and FAP+ CAF are predictors of survival; the former predicted better survival, while the latter correlated with worse survival. On the other hand, Tahkola et al. directly analyzed the effect of hyaluronan accumulation on pancreatic cancer immune cell infiltration, and found the infiltration was significantly interrupted by hyaluronan as reflected by a low immune cell score across survival types (overall and disease-specific) via univariate and multivariate analyses [130]. Moreover, Nizri et al. studied pancreatic cancer lymph node (LN) metastasis and analyzed its association to desmoplasia and the ECM profile. They found collagen, TGFβ (fibroblast-related), and GATA3 ((Th2) immunity-related) are expressed mainly in desmoplasia-high LN [131]. A similar association in pancreatic cancer was also observed in the study by Sadozai et al. [132]. The authors analyzed the components in the pancreatic tumor microenvironment by IHC staining and RNA sequencing to compare the differences in long-term and short-term survivors. The results from both studies revealed that (1) a long-term survivor has less α-SMA and (2) a long-term survivor has more CD3+ T cells in the tumor and stroma parts. State-of-the-art technology such as single cell RNA sequencing accelerates target/solution identification in cancer research, and with this approach, Wang et al. identified the CAF subtype with a highly activated metabolic state (meCAF) in pancreatic cancer patients with loose stroma (low desmoplasia) [55]. Under this circumstance, CD8+ T cells are more abundant in both single cell RNA sequencing and IHC analysis. While the meCAF predicted worse survival, it displayed a better response toward anti-PD-1. This work suggested the identification and potential application of a novel fibroblast subtype for pancreatic cancer research and therapy. Collectively, the aforementioned studies conclude that the alterations in the “Trinity” members in pancreatic tumors provide useful information in predicting a patient’s clinical outcome and in providing a therapeutic strategy.

5. Treating Pancreatic Cancer with CD8+ T Cell-Orientated Approach

5.1. Preclinical Study

Treating pancreatic cancer by directly increasing CD8+ T cell infiltration and/or function is a new idea with clinical merit (Figure 2). Mace et al. found that an IL6 blockade improved anti-PD-L1 efficiency and retarded pancreatic tumorigenesis [133]. With the IL6 expression by pancreatic stellate cell (PSC) in mind, the authors tested the effect of an IL6 neutralizing antibody on subcutaneous tumors of MT5 and Panc02 (mouse pancreatic cancer cell lines), and demonstrated that while IL6 antibody itself had a minor effect, its combination benefited anti-PD-L1 efficiency. This is correlated to increased effector CD8+ T cell infiltration in the tumors. On the other hand, only CD8+ cell neutralization, but not CD4+ cell neutralization, blunted the antitumor effect that resulted from the IL6 blockade. In an orthotopic model, treatment of the IL6 antibody could not prevent CD3+ cell recruitment. Principe et al. demonstrated that TGFβ is of equal importance in this kind of treatment, as KC mouse (P48-Cre × LSL-KRASG12D) receiving wild-type CD8+ T cell still displayed disease progression, but tumor growth was ceased when the CD8+ T cells harvested from Tgfbr1+/− mice were injected [134]. Consistent with this finding, TGFβ inhibitor galunisertib also showed the same inhibitory effect in the animal model and displayed better efficiency when combined with anti-PD-1 antibody. While CD8+ T cell infiltration was not increased, its functions regrading memory subset and IFNγ/perforin expression were all boosted. In addition to IL6 and TGFβ, CXCR4-mediated signaling also affects pancreatic cancer treatment significantly. Two studies addressed the importance of CXCR4 in pancreatic tumorigenesis [135,136]. Seo et al. applied patient specimens to investigate the influence of CXCR4 inhibitor AMD3100 on apoptosis, and found it did so by redistributing the CD8+ T cells to the juxtatumoral stroma with the potential release of granzyme B and IFNγ. In the COMBAT clinical trial, Bockorny et al. applied another CXCR4 antagonist, BL-8040, to test its antitumor effect together with pembrolizumab for pancreatic cancer patients. Patients receiving combinatory therapy displayed increased CD8+ T cell mobilization, especially that of cytotoxic (granzyme B+) CD8+ T cells. When further combined with chemotherapy, most patients displayed a better response compared to their own baselines.
In addition to cytokine/chemokine, the direct activation of CD8+ T cells by membrane proteins is also implicated in pancreatic cancer treatment. Audenaerde et al. and Panni et al. investigated the role of CD40 agonist/IL15 and CD11b agonist, respectively, for this purpose [137,138]. Audenaerde et al. injected mouse cancer cells subcutaneously and tested the effect of different molecules on tumor growth. They found that CD40 agonist/IL15 is the most effective for suppressing tumor growth. This was achieved by the CD8+ T cell as its depletion reversed the therapeutic effect. Accompanying this phenomenon, antitumor factors and effector/memory subsets were all increased. Moreover, the suppressive effect of CD40 agonist/IL15 could still be observed in re-challenge experiments after a 100-day tumor-free period. Panni et al., alternatively, utilized CD11b agonist ADH-503 on an orthotopic mouse model and demonstrated this agonist promoted the recruitment of immune cells including cytotoxic and memory CD8+ T cells under rapid proliferation and activated their antitumor functions. Mechanistically, ADH-503 acted through a conventional dendritic cell (cDC) to achieve tumor suppression, as demonstrated by the lack of response to ADH-503 in a cDC-lacking Batf3−/− mouse. Impressively, the CD11b agonist itself displayed significant efficiency in pancreatic cancer treatment as a sole agent in both subcutaneous and orthotopic models. More importantly, ADH-503 combined with chemotherapy and immunotherapy to dramatically inhibit tumor growth. Modulation of the immune microenvironment is equally important. Michaelis et al. demonstrated that TLR7/8 agonist R848 prolonged survival in intraperitoneal and orthotopic mouse models of pancreatic cancer [139]. R848 itself has been shown to decrease tumor mass via modulations on the immune microenvironment and cachexia.
Finally, the questions raised are how is CD8+ T cell-oriented therapy applied to pancreatic cancer treatment and does the entry of T cells modulate other immune cells to create a favorable anticancer immunity? Ajina et al. showed that the T cell is the determinant for pancreatic cancer suppression in various mouse strains and is a critical factor affecting the recruitment of myeloid cells [140]. They applied NanoString analysis and found Stat1 in a pancreatic cancer cell was upregulated when a T cell was presented. The upregulation of Stat1, in turn, activated the myeloid cell to blunt the T cell’s antitumor effect. As a result, FDA-approved Stat inhibitor ruxolitinib further improved anti-PD-L1 efficiency in the aforementioned models, although ruxolitinib itself displayed minor efficiency. While this improvement on pancreatic cancer suppression may not solely result from myeloid cell inhibition, as it was suggested by the authors that the abundance of MDSC was not significantly decreased, this study pinpointed the importance of CD8+ T cell-orientated treatment [141].

5.2. Clinical Trial

In validation of the results of the preclinical studies described above, we reviewed related clinical trials in the database (https://www.clinicaltrials.gov/ (accessed on 28 February 2022). For pancreatic cancer, STAT inhibitor was applied in trials and the benefits of this agent were reported (NCT02993731) [142] (Table 3). In this randomized, open-label, multi-center, phase 3 study, the effect of napabucasin (2-acetylfuro-1,4-naphthoquinone, BBI-608), a STAT3 inhibitor, on metastatic pancreatic ductal adenocarcinoma was investigated. The effects on disease progression of 1134 patients treated with napabucasin + nab-paclitaxel or nab-paclitaxel + gemcitabine were compared. In the napabucasin + nab-paclitaxel arm, patients were given napabucasin twice daily and the other two agents weekly. In arm 2 of nab-paclitaxel + gemcitabine, patients were given nab-paclitaxel and gemcitabine with the same frequency. The primary outcome was overall survival, and the secondary outcomes were progression-free survival, overall response rate, adverse events, and quality of life. No difference in overall survival was found between the two groups’ 565 patients (11.43 vs. 11.73 months in arms 1 and 2, respectively. For secondary outcomes, the progression-free survival in arm 1 and 2 was 6.70 vs. 6.08 months, respectively; disease control rate was 74 vs. 76%, respectively; overall response rate in both arms was 43%; adverse events was 99.8% vs. 99.3%, respectively; quality of life score was −1.63 versus −0.57, respectively (higher score represents a higher quality of life). Although the combination of napabucasin and nab-paclitaxel does not show a better effect than nab-paclitaxel + gemcitabine, this study helps to define how this STAT3 inhibitor influences disease progression in phospho-STAT3-positive patients and its potential application in the future. Recently, the effects of napabucasin in clinical trials for multiple cancers have been updated [142,143,144].
Although the benefits of targeting IL6, TGFβ, CXCR4, and CD40 in pancreatic cancer remain elusive, clinical trials for these factors in other cancer types have shown promise. The effects of the anti-IL6 antibody in treating multiple myeloma were tested in NCT00911859 and NCT01484275 trials. In NCT00911859, this randomized, open-label, phase 2 study investigated an IL6 antibody (CNTO 328, siltuximab) and its combination with velcade-melphalan-prednisone (VMP) to treat multiple myeloma [145]. The safety of siltuximab (11 mg/kg as a 1-h intravenous infusion every 3 weeks) in patients was first confirmed in part 1 of this trial. In part 2, treatment efficiency was compared between arm A of VMP + siltuximab and arm B of VMP. The primary outcome was complete response, and secondary outcomes were overall response, duration of response, progression-free survival, overall survival, and quality of life. In arm A (52 patients) and arm B (54 patients), complete response was 26.5 and 22.4%, respectively. For secondary outcomes, in arm A and arm B, the overall response was 87.8 versus 79.6%, respectively; duration of response was 583 versus 497 days, respectively; progression-free survival was 519 versus 518 days, respectively. Conversely, the quality of life score was 8.33 and 14.78 for arm A and B, respectively. In a related publication [145], San-Miguel et al. further showed that (1) the partial response in arm A and arm B was 71 and 51%, respectively (p = 0.0382); (2) adverse event in arm A and arm B was 92 and 81%, respectively (p = 0.09). The authors concluded that there is no significant improvement in terms of complete response and outcome using siltuximab among patients with multiple myeloma. In NCT01484275, siltuximab was applied to test its safety and efficiency for patients with high-risk smoldering multiple myeloma (SMM). The safety of siltuximab in SMM patients was confirmed (15 mg/kg as a 1-h intravenous infusion every 4 weeks). This randomized, double-blind, placebo-controlled, multi-center, phase 2 trial enrolled 85 patients. The siltuximab arm (43 patients) and placebo arm (42 patients) had one-year progression-free survival rates of 84.5 and 74.4%, respectively. The time to disease progression was 125.5 versus 118 days in the siltuximab and placebo group, respectively. Therefore, siltuximab does not improve the survival of SMM patients [146].
For CXCR4, two trials (NCT00906945 and NCT00512252) studied the impact of CXCR4 blockage on disease inhibition in acute myeloid leukemia (AML). In the NCT00906945 study, the applications of G-CSF as well as CXCR4 blockade (plerixafor, AMD3100) were performed to investigate the clinical benefits. This non-randomized, open-label, phase 1/2 trial enrolled 39 patients. The results of this study showed a complete response rate of 30% and suggested the benefit of CXCR4 inhibition in AML patients [147]. In the NCT00512252 trial, without G-CSF, AMD3100 still increased the effectiveness of mitoxantrone, etoposide, and cytarabine (MEC) in AML patients. It was concluded that the CXCR4 inhibitor mobilized the AML cell to circulation and improved treatment efficiency, and plerixafor could be safely combined with chemotherapy to treat relapsed or refractory AML [148].
For membrane protein activation and its related cancer treatment, targeting CD40 was investigated in NCT00101166 and NCT01433172 trials. These trials applied a cancer vaccine that is composed of tumor cells from patient and bystander cells expressing GM-CSF and CD40L. These cells were treated with high-dose X-rays to ensure the loss of oncogenicity. This vaccine is able to better boost immunity in patients. In the NCT00101166 trial, the investigators reported that the recruitment of the CD1a+CD86+ dendritic cell could be observed in the injection site via immunohistochemistry analysis [149]. In addition, the cancer vaccine worked well for several types of solid tumors. In the NCT01433172 study, in addition to the cancer vaccine mentioned above, cytokine CCL21 was added to assist T cell recruitment and responsiveness. The results of this study revealed that GM-CSF + CD40L + CCL21 vaccination induced abundant tumor-infiltrating lymphocytes in tumors, which suggested the benefit of CD40 blockade in cancer treatment [150]. Taken together, these clinical trials shed light on the importance of IL6, TGFβ, CXCR4, and CD40 in cancer treatment, as well as their potential therapeutic effects on pancreatic cancer. Even though these clinical trials were applied to patients with cancer types other than pancreatic cancer, and the hazard ratio as well as statistical significance may be restricted due to the enrolled patient numbers, they suggest the potential application of the above treatments for pancreatic cancer patients.

5.3. Clinical Correlation

In addition to macrophages and neutrophils, the dendritic cell (DC) adds an important portion to cancer immunity via antigen presentation. The dendritic cell in pancreatic cancer also correlated with better survival [151]. Conventional DC type 1 (cDC1), type 2 (cDC2), and plasmacytoid DC (pDC) were enriched more in the stroma than in the tumor. cDC1 in the tumor stroma further predicted better disease-free survival (DFS). Mechanistically, DC function in pancreatic cancer is affected by multiple factors and proteins. Pancreatic cancer cell-derived prostaglandin E2 (PGE2) and regenerating islet-derived protein 3A (Reg3A) decreased DC function via cellular stresses and JAK2 modulation, respectively [152,153]. This subsequently led to a decrease in T cell proliferation [153]. The stromal protein also influenced DC effectiveness during pancreatic tumorigenesis as Giri et al. reported that the loss of heat shock protein 70 (Hsp70) in DC increased its functionality [154]. In terms of pancreatic cancer treatment, DC is also a critical target. Sadeghlar et al. showed that CD40L transduction in human DC increased its function and proliferation, even in the presence of a pancreatic cancer cell in co-culture [155], and Kim et al. reported that cetuximab-conjugated maleimide-polyethylene glycol-chlorin e6 induced pancreatic cancer cell death, which was accompanied by DC activation and maturation [156]. Salah et al. demonstrated that pulsed DC increased antigen-specific cytotoxic T cells among pancreatic cancer patients [157]. Recently, Kvedaraite et al. also summarized that pancreatic cancer patients had fewer DC progenitors and cDC1s, and pDC predicted better prognosis [158] (Figure 3).
Predictive signatures are essential for improvements in pancreatic cancer diagnosis and treatment. With databases including The Cancer Genome Atlas (TCGA), 2-gene TP53-associated immune prognostic model (TIPM) [159], 15-gene immune, stromal, and proliferation-associated (ISP) signature [160], 47-gene immune-related gene pairs (IRGPs) [161], 8-gene signature [162], 59-gene TMEscore-high group [163], and 5-gene module immune cluster [164], we could predict pancreatic cancer prognosis more precisely. At protein level with tissue arrays, signatures of leukocyte subtype/stromal composition [165] also predicted differential survival among pancreatic cancer patients. Santiago et al. reviewed immunophenotypes in pancreatic cancer in great detail [166].
To sum up, with all the studies and reviews above, it is clear that the complex nature of the interplay between pancreatic cancer cells, fibroblasts, and immune cells is a key factor determining tumor formation and treatment response. In addition to therapies targeting fibroblasts [35,36] and immune cells [71,72], how these cells affect the rest of the bulky tumor to create a tumor-promoting microenvironment is a critical issue for continuous exploration. Both hypothesis-driven study [113,120] and high-throughput analysis [45,55] will strengthen our understanding of this important interplay guided by the cancer cells, stroma, and immunity in pancreatic cancer [140,167].

Author Contributions

Conceptualization, Y.-H.H. and W.-C.H.; writing—original draft preparation, Y.-H.H.; writing—review and editing, W.-C.H.; supervision, L.-T.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grant CA-110-PP-17 from National Health Research Institutes, Ministry of Health and Welfare to Wen-Chun Hung.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Traub, B.; Link, K.H.; Kornmann, M. Curing pancreatic cancer. Semin. Cancer Biol. 2021, 76, 232–246. [Google Scholar] [CrossRef] [PubMed]
  4. Yazal, T.; Bailleul, J.; Ruan, Y.; Sung, D.; Chu, F.I.; Palomera, D.; Dao, A.; Sehgal, A.; Gurunathan, V.; Aryan, L.; et al. Radiosensitizing Pancreatic Cancer via Effective Autophagy Inhibition. Mol. Cancer Ther. 2022, 21, 79–88. [Google Scholar] [CrossRef]
  5. Macedo, F.I.; Ryon, E.; Maithel, S.K.; Lee, R.M.; Kooby, D.A.; Fields, R.C.; Hawkins, W.G.; Williams, G.; Maduekwe, U.; Kim, H.J.; et al. Survival Outcomes Associated with Clinical and Pathological Response Following Neoadjuvant FOLFIRINOX or Gemcitabine/Nab-Paclitaxel Chemotherapy in Resected Pancreatic Cancer. Ann. Surg. 2019, 270, 400–413. [Google Scholar] [CrossRef]
  6. Yu, S.; Zhang, C.; Xie, K.P. Therapeutic resistance of pancreatic cancer: Roadmap to its reversal. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188461. [Google Scholar] [CrossRef]
  7. Timmer, F.E.F.; Geboers, B.; Nieuwenhuizen, S.; Dijkstra, M.; Schouten, E.A.C.; Puijk, R.S.; de Vries, J.J.J.; van den Tol, M.P.; Bruynzeel, A.M.E.; Streppel, M.M.; et al. Pancreatic Cancer and Immunotherapy: A Clinical Overview. Cancers 2021, 13, 4138. [Google Scholar] [CrossRef]
  8. Principe, D.R.; Korc, M.; Kamath, S.D.; Munshi, H.G.; Rana, A. Trials and tribulations of pancreatic cancer immunotherapy. Cancer Lett. 2021, 504, 1–14. [Google Scholar] [CrossRef]
  9. Kang, J.S.; Choi, Y.J.; Byun, Y.; Han, Y.; Kim, J.H.; Lee, J.M.; Sohn, H.J.; Kim, H.; Kwon, W.; Jang, J.Y. Radiological tumour invasion of splenic artery or vein in patients with pancreatic body or tail adenocarcinoma and effect on recurrence and survival. Br. J. Surg. 2021, 109, 105–113. [Google Scholar] [CrossRef]
  10. Zhu, X.; Cao, Y.; Liu, W.; Ju, X.; Zhao, X.; Jiang, L.; Ye, Y.; Jin, G.; Zhang, H. Stereotactic body radiotherapy plus pembrolizumab and trametinib versus stereotactic body radiotherapy plus gemcitabine for locally recurrent pancreatic cancer after surgical resection: An open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2021, 22, 1093–1102. [Google Scholar] [CrossRef]
  11. Chen, X.; Zhang, L.; Jiang, Y.; Song, L.; Liu, Y.; Cheng, F.; Fan, X.; Cao, X.; Gong, A.; Wang, D.; et al. Radiotherapy-induced cell death activates paracrine HMGB1-TLR2 signaling and accelerates pancreatic carcinoma metastasis. J. Exp. Clin. Cancer Res. 2018, 37, 77. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, L.; Shi, H.; Chen, H.; Gong, A.; Liu, Y.; Song, L.; Xu, X.; You, T.; Fan, X.; Wang, D.; et al. Dedifferentiation process driven by radiotherapy-induced HMGB1/TLR2/YAP/HIF-1α signaling enhances pancreatic cancer stemness. Cell Death Dis. 2019, 10, 724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kawahara, K.; Takano, S.; Furukawa, K.; Takayashiki, T.; Kuboki, S.; Ohtsuka, M. The effect of the low stromal ratio induced by neoadjuvant chemotherapy on recurrence patterns in borderline resectable pancreatic ductal adenocarcinoma. Clin. Exp. Metastasis 2022, 39, 311–322. [Google Scholar] [CrossRef] [PubMed]
  14. Hu, Z.I.; Hellmann, M.D.; Wolchok, J.D.; Vyas, M.; Shia, J.; Stadler, Z.K.; Diaz, L.A., Jr.; O’Reilly, E.M. Acquired resistance to immunotherapy in MMR-D pancreatic cancer. J. Immunother. Cancer 2018, 6, 127. [Google Scholar] [CrossRef]
  15. Jiang, J.; Zhou, H.; Ni, C.; Hu, X.; Mou, Y.; Huang, D.; Yang, L. Immunotherapy in pancreatic cancer: New hope or mission impossible? Cancer Lett. 2019, 445, 57–64. [Google Scholar] [CrossRef]
  16. Dreyer, S.B.; Upstill-Goddard, R.; Legrini, A.; Biankin, A.V.; Jamieson, N.B.; Chang, D.K.; Jamieson, N.B.; Chang, D.K. Genomic and Molecular Analyses Identify Molecular Subtypes of Pancreatic Cancer Recurrence. Gastroenterology 2022, 162, 320–324.e324. [Google Scholar] [CrossRef]
  17. Asimgil, H.; Ertetik, U.; Çevik, N.C.; Ekizce, M.; Doğruöz, A.; Gökalp, M.; Arık-Sever, E.; Istvanffy, R.; Friess, H.; Ceyhan, G.O.; et al. Targeting the undruggable oncogenic KRAS: The dawn of hope. JCI Insight 2022, 7, e153688. [Google Scholar] [CrossRef]
  18. Saiki, Y.; Jiang, C.; Ohmuraya, M.; Furukawa, T. Genetic Mutations of Pancreatic Cancer and Genetically Engineered Mouse Models. Cancers 2021, 14, 71. [Google Scholar] [CrossRef]
  19. Ala, M. Target c-Myc to treat pancreatic cancer. Cancer Biol. Ther. 2022, 23, 34–50. [Google Scholar] [CrossRef]
  20. Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; Neale, R.E.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2016, 2, 16022. [Google Scholar] [CrossRef]
  21. Collisson, E.A.; Bailey, P.; Chang, D.K.; Biankin, A.V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 207–220. [Google Scholar] [CrossRef] [PubMed]
  22. Ou, A.; Zhao, X.; Lu, Z. The potential roles of p53 signaling reactivation in pancreatic cancer therapy. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188662. [Google Scholar] [CrossRef] [PubMed]
  23. Asghar, U.S.; Kanani, R.; Roylance, R.; Mittnacht, S. Systematic Review of Molecular Biomarkers Predictive of Resistance to CDK4/6 Inhibition in Metastatic Breast Cancer. JCO Precis. Oncol. 2022, 6, e2100002. [Google Scholar] [CrossRef] [PubMed]
  24. Fassl, A.; Geng, Y.; Sicinski, P. CDK4 and CDK6 kinases: From basic science to cancer therapy. Science 2022, 375, eabc1495. [Google Scholar] [CrossRef]
  25. Killock, D. Abemaciclib crowned in monarchE. Nat. Rev. Clin. Oncol. 2022, 19, 5. [Google Scholar] [CrossRef]
  26. Cheng, B.; Pan, W.; Xing, Y.; Xiao, Y.; Chen, J.; Xu, Z. Recent advances in DDR (DNA damage response) inhibitors for cancer therapy. Eur. J. Med. Chem. 2022, 230, 114109. [Google Scholar] [CrossRef]
  27. Prados-Carvajal, R.; Irving, E.; Lukashchuk, N.; Forment, J.V. Preventing and Overcoming Resistance to PARP Inhibitors: A Focus on the Clinical Landscape. Cancers 2021, 14, 44. [Google Scholar] [CrossRef]
  28. Ji, Y.; Liu, X.; Li, J.; Xie, X.; Huang, M.; Jiang, J.; Liao, Y.P.; Donahue, T.; Meng, H. Use of ratiometrically designed nanocarrier targeting CDK4/6 and autophagy pathways for effective pancreatic cancer treatment. Nat. Commun. 2020, 11, 4249. [Google Scholar] [CrossRef]
  29. Wang, S.P.; Li, Y.; Huang, S.H.; Wu, S.Q.; Gao, L.L.; Sun, Q.; Lin, Q.W.; Huang, L.; Meng, L.Q.; Zou, Y.; et al. Discovery of Potent and Novel Dual PARP/BRD4 Inhibitors for Efficient Treatment of Pancreatic Cancer. J. Med. Chem. 2021, 64, 17413–17435. [Google Scholar] [CrossRef]
  30. Majidpoor, J.; Mortezaee, K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin. Immunol. 2021, 226, 108707. [Google Scholar] [CrossRef]
  31. Torphy, R.J.; Schulick, R.D.; Zhu, Y. Understanding the immune landscape and tumor microenvironment of pancreatic cancer to improve immunotherapy. Mol. Carcinog. 2020, 59, 775–782. [Google Scholar] [CrossRef] [PubMed]
  32. Wandmacher, A.M.; Letsch, A.; Sebens, S. Challenges and Future Perspectives of Immunotherapy in Pancreatic Cancer. Cancers 2021, 13, 4235. [Google Scholar] [CrossRef] [PubMed]
  33. Saka, D.; Gökalp, M.; Piyade, B.; Cevik, N.C.; Arik Sever, E.; Unutmaz, D.; Ceyhan, G.O.; Demir, I.E.; Asimgil, H. Mechanisms of T-Cell Exhaustion in Pancreatic Cancer. Cancers 2020, 12, 2274. [Google Scholar] [CrossRef] [PubMed]
  34. Kemp, S.B.; Pasca di Magliano, M.; Crawford, H.C. Myeloid Cell Mediated Immune Suppression in Pancreatic Cancer. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 1531–1542. [Google Scholar] [CrossRef]
  35. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef] [Green Version]
  36. Liu, H.; Shi, Y.; Qian, F. Opportunities and delusions regarding drug delivery targeting pancreatic cancer-associated fibroblasts. Adv. Drug Deliv. Rev. 2021, 172, 37–51. [Google Scholar] [CrossRef]
  37. Elyada, E.; Bolisetty, M.; Laise, P.; Flynn, W.F.; Courtois, E.T.; Burkhart, R.A.; Teinor, J.A.; Belleau, P.; Biffi, G.; Lucito, M.S.; et al. Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov. 2019, 9, 1102–1123. [Google Scholar] [CrossRef] [Green Version]
  38. Sunami, Y.; Häußler, J.; Kleeff, J. Cellular Heterogeneity of Pancreatic Stellate Cells, Mesenchymal Stem Cells, and Cancer-Associated Fibroblasts in Pancreatic Cancer. Cancers 2020, 12, 3770. [Google Scholar] [CrossRef]
  39. Sun, Q.; Zhang, B.; Hu, Q.; Qin, Y.; Xu, W.; Liu, W.; Yu, X.; Xu, J. The impact of cancer-associated fibroblasts on major hallmarks of pancreatic cancer. Theranostics 2018, 8, 5072–5087. [Google Scholar] [CrossRef]
  40. Helms, E.; Onate, M.K.; Sherman, M.H. Fibroblast Heterogeneity in the Pancreatic Tumor Microenvironment. Cancer Discov. 2020, 10, 648–656. [Google Scholar] [CrossRef] [Green Version]
  41. Ping, Q.; Yan, R.; Cheng, X.; Wang, W.; Zhong, Y.; Hou, Z.; Shi, Y.; Wang, C.; Li, R. Cancer-associated fibroblasts: Overview, progress, challenges, and directions. Cancer Gene Ther. 2021, 28, 984–999. [Google Scholar] [CrossRef] [PubMed]
  42. Neesse, A.; Bauer, C.A.; Öhlund, D.; Lauth, M.; Buchholz, M.; Michl, P.; Tuveson, D.A.; Gress, T.M. Stromal biology and therapy in pancreatic cancer: Ready for clinical translation? Gut 2019, 68, 159–171. [Google Scholar] [CrossRef] [PubMed]
  43. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef] [PubMed]
  44. Xiao, Q.; Zhou, D.; Rucki, A.A.; Williams, J.; Zhou, J.; Mo, G.; Murphy, A.; Fujiwara, K.; Kleponis, J.; Salman, B.; et al. Cancer-Associated Fibroblasts in Pancreatic Cancer Are Reprogrammed by Tumor-Induced Alterations in Genomic DNA Methylation. Cancer Res. 2016, 76, 5395–5404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Dominguez, C.X.; Müller, S.; Keerthivasan, S.; Koeppen, H.; Hung, J.; Gierke, S.; Breart, B.; Foreman, O.; Bainbridge, T.W.; Castiglioni, A.; et al. Single-Cell RNA Sequencing Reveals Stromal Evolution into LRRC15(+) Myofibroblasts as a Determinant of Patient Response to Cancer Immunotherapy. Cancer Discov. 2020, 10, 232–253. [Google Scholar] [CrossRef] [Green Version]
  46. Biffi, G.; Oni, T.E.; Spielman, B.; Hao, Y.; Elyada, E.; Park, Y.; Preall, J.; Tuveson, D.A. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFβ to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov. 2019, 9, 282–301. [Google Scholar] [CrossRef] [Green Version]
  47. Neuzillet, C.; Tijeras-Raballand, A.; Ragulan, C.; Cros, J.; Patil, Y.; Martinet, M.; Erkan, M.; Kleeff, J.; Wilson, J.; Apte, M.; et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J. Pathol. 2019, 248, 51–65. [Google Scholar] [CrossRef] [Green Version]
  48. Baker, A.T.; Abuwarwar, M.H.; Poly, L.; Wilkins, S.; Fletcher, A.L. Cancer-Associated Fibroblasts and T Cells: From Mechanisms to Outcomes. J. Immunol. 2021, 206, 310–320. [Google Scholar] [CrossRef]
  49. Huang, H.; Brekken, R.A. Recent advances in understanding cancer-associated fibroblasts in pancreatic cancer. Am. J. Physiol. Cell Physiol. 2020, 319, C233–C243. [Google Scholar] [CrossRef]
  50. Norton, J.; Foster, D.; Chinta, M.; Titan, A.; Longaker, M. Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers 2020, 12, 1347. [Google Scholar] [CrossRef]
  51. Polani, F.; Grierson, P.M.; Lim, K.H. Stroma-targeting strategies in pancreatic cancer: Past lessons, challenges and prospects. World J. Gastroenterol. 2021, 27, 2105–2121. [Google Scholar] [CrossRef] [PubMed]
  52. Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Rhim, A.D.; Oberstein, P.E.; Thomas, D.H.; Mirek, E.T.; Palermo, C.F.; Sastra, S.A.; Dekleva, E.N.; Saunders, T.; Becerra, C.P.; Tattersall, I.W.; et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014, 25, 735–747. [Google Scholar] [CrossRef] [Green Version]
  54. Steele, N.G.; Biffi, G.; Kemp, S.B.; Zhang, Y.; Drouillard, D.; Syu, L.; Hao, Y.; Oni, T.E.; Brosnan, E.; Elyada, E.; et al. Inhibition of Hedgehog Signaling Alters Fibroblast Composition in Pancreatic Cancer. Clin. Cancer Res. 2021, 27, 2023–2037. [Google Scholar] [CrossRef]
  55. Wang, Y.; Liang, Y.; Xu, H.; Zhang, X.; Mao, T.; Cui, J.; Yao, J.; Wang, Y.; Jiao, F.; Xiao, X.; et al. Single-cell analysis of pancreatic ductal adenocarcinoma identifies a novel fibroblast subtype associated with poor prognosis but better immunotherapy response. Cell Discov. 2021, 7, 36. [Google Scholar] [CrossRef] [PubMed]
  56. Affo, S.; Nair, A.; Brundu, F.; Ravichandra, A.; Bhattacharjee, S.; Matsuda, M.; Chin, L.; Filliol, A.; Wen, W.; Song, X.; et al. Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations. Cancer Cell 2021, 39, 883. [Google Scholar] [CrossRef] [PubMed]
  57. Strating, E.; Wassenaar, E.; Verhagen, M.; Rauwerdink, P.; van Schelven, S.; de Hingh, I.; Rinkes, I.B.; Boerma, D.; Witkamp, A.; Lacle, M.; et al. Fibroblast activation protein identifies Consensus Molecular Subtype 4 in colorectal cancer and allows its detection by (68)Ga-FAPI-PET imaging. Br. J. Cancer 2022. [Google Scholar] [CrossRef]
  58. Chen, Z.; Zhou, L.; Liu, L.; Hou, Y.; Xiong, M.; Yang, Y.; Hu, J.; Chen, K. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat. Commun. 2020, 11, 5077. [Google Scholar] [CrossRef]
  59. Qi, J.; Sun, H.; Zhang, Y.; Wang, Z.; Xun, Z.; Li, Z.; Ding, X.; Bao, R.; Hong, L.; Jia, W.; et al. Single-cell and spatial analysis reveal interaction of FAP(+) fibroblasts and SPP1(+) macrophages in colorectal cancer. Nat. Commun. 2022, 13, 1742. [Google Scholar] [CrossRef]
  60. Mhaidly, R.; Mechta-Grigoriou, F. Role of cancer-associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer. Immunol. Rev. 2021, 302, 259–272. [Google Scholar] [CrossRef]
  61. Ganguly, K.; Shah, A.; Atri, P.; Rauth, S.; Ponnusamy, M.P.; Kumar, S.; Batra, S.K. Chemokine-mucinome interplay in shaping the heterogeneous tumor microenvironment of pancreatic cancer. Semin. Cancer Biol. 2022. [Google Scholar] [CrossRef] [PubMed]
  62. Hosein, A.N.; Brekken, R.A.; Maitra, A. Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 487–505. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, J.J.; Bernard, V.; Semaan, A.; Monberg, M.E.; Huang, J.; Stephens, B.M.; Lin, D.; Rajapakshe, K.I.; Weston, B.R.; Bhutani, M.S.; et al. Elucidation of Tumor-Stromal Heterogeneity and the Ligand-Receptor Interactome by Single-Cell Transcriptomics in Real-world Pancreatic Cancer Biopsies. Clin. Cancer Res. 2021, 27, 5912–5921. [Google Scholar] [CrossRef] [PubMed]
  64. Domen, A.; Quatannens, D.; Zanivan, S.; Deben, C.; Van Audenaerde, J.; Smits, E.; Wouters, A.; Lardon, F.; Roeyen, G.; Verhoeven, Y.; et al. Cancer-Associated Fibroblasts as a Common Orchestrator of Therapy Resistance in Lung and Pancreatic Cancer. Cancers 2021, 13, 987. [Google Scholar] [CrossRef]
  65. Gorchs, L.; Kaipe, H. Interactions between Cancer-Associated Fibroblasts and T Cells in the Pancreatic Tumor Microenvironment and the Role of Chemokines. Cancers 2021, 13, 2995. [Google Scholar] [CrossRef] [PubMed]
  66. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
  67. Boyd, L.N.C.; Andini, K.D.; Peters, G.J.; Kazemier, G.; Giovannetti, E. Heterogeneity and plasticity of cancer-associated fibroblasts in the pancreatic tumor microenvironment. Semin. Cancer Biol. 2021. [Google Scholar] [CrossRef]
  68. Geng, X.; Chen, H.; Zhao, L.; Hu, J.; Yang, W.; Li, G.; Cheng, C.; Zhao, Z.; Zhang, T.; Li, L.; et al. Cancer-Associated Fibroblast (CAF) Heterogeneity and Targeting Therapy of CAFs in Pancreatic Cancer. Front. Cell Dev. Biol. 2021, 9, 655152. [Google Scholar] [CrossRef]
  69. Vaish, U.; Jain, T.; Are, A.C.; Dudeja, V. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma: An Update on Heterogeneity and Therapeutic Targeting. Int. J. Mol. Sci. 2021, 22, 13408. [Google Scholar] [CrossRef]
  70. Van Audenaerde, J.R.M.; Roeyen, G.; Darcy, P.K.; Kershaw, M.H.; Peeters, M.; Smits, E.L.J. Natural killer cells and their therapeutic role in pancreatic cancer: A systematic review. Pharmacol. Ther. 2018, 189, 31–44. [Google Scholar] [CrossRef]
  71. Yang, J.; Li, Y.; Sun, Z.; Zhan, H. Macrophages in pancreatic cancer: An immunometabolic perspective. Cancer Lett. 2021, 498, 188–200. [Google Scholar] [CrossRef] [PubMed]
  72. Gan, L.L.; Hii, L.W.; Wong, S.F.; Leong, C.O.; Mai, C.W. Molecular Mechanisms and Potential Therapeutic Reversal of Pancreatic Cancer-Induced Immune Evasion. Cancers 2020, 12, 1872. [Google Scholar] [CrossRef] [PubMed]
  73. Pergamo, M.; Miller, G. Myeloid-derived suppressor cells and their role in pancreatic cancer. Cancer Gene Ther. 2017, 24, 100–105. [Google Scholar] [CrossRef]
  74. Vonderheide, R.H.; Bear, A.S. Tumor-Derived Myeloid Cell Chemoattractants and T Cell Exclusion in Pancreatic Cancer. Front. Immunol. 2020, 11, 605619. [Google Scholar] [CrossRef] [PubMed]
  75. Ware, M.B.; El-Rayes, B.F.; Lesinski, G.B. Mirage or long-awaited oasis: Reinvigorating T-cell responses in pancreatic cancer. J. Immunother. Cancer 2020, 8, e001100. [Google Scholar] [CrossRef] [PubMed]
  76. Lankadasari, M.B.; Mukhopadhyay, P.; Mohammed, S.; Harikumar, K.B. TAMing pancreatic cancer: Combat with a double edged sword. Mol. Cancer 2019, 18, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Fincham, R.E.A.; Delvecchio, F.R.; Goulart, M.R.; Yeong, J.P.S.; Kocher, H.M. Natural killer cells in pancreatic cancer stroma. World J. Gastroenterol. 2021, 27, 3483–3501. [Google Scholar] [CrossRef] [PubMed]
  78. Orhan, A.; Vogelsang, R.P.; Andersen, M.B.; Madsen, M.T.; Hölmich, E.R.; Raskov, H.; Gögenur, I. The prognostic value of tumour-infiltrating lymphocytes in pancreatic cancer: A systematic review and meta-analysis. Eur. J. Cancer 2020, 132, 71–84. [Google Scholar] [CrossRef]
  79. Xu, C.; Sui, S.; Shang, Y.; Yu, Z.; Han, J.; Zhang, G.; Ntim, M.; Hu, M.; Gong, P.; Chen, H.; et al. The landscape of immune cell infiltration and its clinical implications of pancreatic ductal adenocarcinoma. J. Adv. Res. 2020, 24, 139–148. [Google Scholar] [CrossRef]
  80. Theoharides, T.C. Mast cells and pancreatic cancer. N. Engl. J. Med. 2008, 358, 1860–1861. [Google Scholar] [CrossRef] [Green Version]
  81. Ma, Y.; Ullrich, S.E. Intratumoral mast cells promote the growth of pancreatic cancer. Oncoimmunology 2013, 2, e25964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Longo, V.; Tamma, R.; Brunetti, O.; Pisconti, S.; Argentiero, A.; Silvestris, N.; Ribatti, D. Mast cells and angiogenesis in pancreatic ductal adenocarcinoma. Clin. Exp. Med. 2018, 18, 319–323. [Google Scholar] [CrossRef] [PubMed]
  83. Mowbray, N.G.; Griffith, D.; Hammoda, M.; Shingler, G.; Kambal, A.; Al-Sarireh, B. A meta-analysis of the utility of the neutrophil-to-lymphocyte ratio in predicting survival after pancreatic cancer resection. HPB 2018, 20, 379–384. [Google Scholar] [CrossRef] [Green Version]
  84. Zhou, Y.; Wei, Q.; Fan, J.; Cheng, S.; Ding, W.; Hua, Z. Prognostic role of the neutrophil-to-lymphocyte ratio in pancreatic cancer: A meta-analysis containing 8252 patients. Clin. Chim. Acta 2018, 479, 181–189. [Google Scholar] [CrossRef]
  85. Yu, M.; Guan, R.; Hong, W.; Zhou, Y.; Lin, Y.; Jin, H.; Hou, B.; Jian, Z. Prognostic value of tumor-associated macrophages in pancreatic cancer: A meta-analysis. Cancer Manag. Res. 2019, 11, 4041–4058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Dallal, R.M.; Christakos, P.; Lee, K.; Egawa, S.; Son, Y.I.; Lotze, M.T. Paucity of dendritic cells in pancreatic cancer. Surgery 2002, 131, 135–138. [Google Scholar] [CrossRef]
  87. Bauer, C.; Dauer, M.; Saraj, S.; Schnurr, M.; Bauernfeind, F.; Sterzik, A.; Junkmann, J.; Jakl, V.; Kiefl, R.; Oduncu, F.; et al. Dendritic cell-based vaccination of patients with advanced pancreatic carcinoma: Results of a pilot study. Cancer Immunol. Immunother. 2011, 60, 1097–1107. [Google Scholar] [CrossRef]
  88. Ochi, A.; Nguyen, A.H.; Bedrosian, A.S.; Mushlin, H.M.; Zarbakhsh, S.; Barilla, R.; Zambirinis, C.P.; Fallon, N.C.; Rehman, A.; Pylayeva-Gupta, Y.; et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J. Exp. Med. 2012, 209, 1671–1687. [Google Scholar] [CrossRef]
  89. Kobayashi, M.; Shimodaira, S.; Nagai, K.; Ogasawara, M.; Takahashi, H.; Abe, H.; Tanii, M.; Okamoto, M.; Tsujitani, S.; Yusa, S.; et al. Prognostic factors related to add-on dendritic cell vaccines on patients with inoperable pancreatic cancer receiving chemotherapy: A multicenter analysis. Cancer Immunol. Immunother. 2014, 63, 797–806. [Google Scholar] [CrossRef]
  90. Mayanagi, S.; Kitago, M.; Sakurai, T.; Matsuda, T.; Fujita, T.; Higuchi, H.; Taguchi, J.; Takeuchi, H.; Itano, O.; Aiura, K.; et al. Phase I pilot study of Wilms tumor gene 1 peptide-pulsed dendritic cell vaccination combined with gemcitabine in pancreatic cancer. Cancer Sci. 2015, 106, 397–406. [Google Scholar] [CrossRef] [Green Version]
  91. Okamoto, M.; Kobayashi, M.; Yonemitsu, Y.; Koido, S.; Homma, S. Dendritic cell-based vaccine for pancreatic cancer in Japan. World J. Gastrointest. Pharmacol. Ther. 2016, 7, 133–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hegde, S.; Krisnawan, V.E.; Herzog, B.H.; Zuo, C.; Breden, M.A.; Knolhoff, B.L.; Hogg, G.D.; Tang, J.P.; Baer, J.M.; Mpoy, C.; et al. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020, 37, 289–307.e289. [Google Scholar] [CrossRef]
  93. Shangguan, A.; Shang, N.; Figini, M.; Pan, L.; Yang, J.; Ma, Q.; Hu, S.; Eresen, A.; Sun, C.; Wang, B.; et al. Prophylactic dendritic cell vaccination controls pancreatic cancer growth in a mouse model. Cytotherapy 2020, 22, 6–15. [Google Scholar] [CrossRef]
  94. Saxena, M.; Bhardwaj, N. Re-Emergence of Dendritic Cell Vaccines for Cancer Treatment. Trends Cancer 2018, 4, 119–137. [Google Scholar] [CrossRef]
  95. Cui, C.; Lan, P.; Fu, L. The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Commun. 2021, 41, 442–471. [Google Scholar] [CrossRef]
  96. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef] [Green Version]
  97. Trovato, R.; Fiore, A.; Sartori, S.; Canè, S.; Giugno, R.; Cascione, L.; Paiella, S.; Salvia, R.; De Sanctis, F.; Poffe, O.; et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J. Immunother. Cancer 2019, 7, 255. [Google Scholar] [CrossRef] [PubMed]
  98. Peng, M.; Zhang, Q.; Liu, Y.; Guo, X.; Ju, J.; Xu, L.; Gao, Y.; Chen, D.; Mu, D.; Zhang, R. Apolipoprotein A-I Mimetic Peptide L-4F Suppresses Granulocytic-Myeloid-Derived Suppressor Cells in Mouse Pancreatic Cancer. Front. Pharmacol. 2020, 11, 576. [Google Scholar] [CrossRef] [PubMed]
  99. Kramer, E.D.; Abrams, S.I. Granulocytic Myeloid-Derived Suppressor Cells as Negative Regulators of Anticancer Immunity. Front. Immunol. 2020, 11, 1963. [Google Scholar] [CrossRef] [PubMed]
  100. Krishnamoorthy, M.; Lenehan, J.G.; Burton, J.P.; Maleki Vareki, S. Immunomodulation in Pancreatic Cancer. Cancers 2020, 12, 3340. [Google Scholar] [CrossRef]
  101. Mucciolo, G.; Curcio, C.; Roux, C.; Li, W.Y.; Capello, M.; Curto, R.; Chiarle, R.; Giordano, D.; Satolli, M.A.; Lawlor, R.; et al. IL17A critically shapes the transcriptional program of fibroblasts in pancreatic cancer and switches on their protumorigenic functions. Proc. Natl. Acad. Sci. USA 2021, 118, e2020395118. [Google Scholar] [CrossRef]
  102. McAllister, F.; Bailey, J.M.; Alsina, J.; Nirschl, C.J.; Sharma, R.; Fan, H.; Rattigan, Y.; Roeser, J.C.; Lankapalli, R.H.; Zhang, H.; et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 2014, 25, 621–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hamarsheh, S.; Groß, O.; Brummer, T.; Zeiser, R. Immune modulatory effects of oncogenic KRAS in cancer. Nat. Commun. 2020, 11, 5439. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, X.P.; Ghoreschi, K.; Steward-Tharp, S.M.; Rodriguez-Canales, J.; Zhu, J.; Grainger, J.R.; Hirahara, K.; Sun, H.W.; Wei, L.; Vahedi, G.; et al. Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat. Immunol. 2011, 12, 247–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Park, M.J.; Moon, S.J.; Lee, E.J.; Jung, K.A.; Kim, E.K.; Kim, D.S.; Lee, J.H.; Kwok, S.K.; Min, J.K.; Park, S.H.; et al. IL-1-IL-17 Signaling Axis Contributes to Fibrosis and Inflammation in Two Different Murine Models of Systemic Sclerosis. Front. Immunol. 2018, 9, 1611. [Google Scholar] [CrossRef] [Green Version]
  106. Zhang, Y.; Chandra, V.; Riquelme Sanchez, E.; Dutta, P.; Quesada, P.R.; Rakoski, A.; Zoltan, M.; Arora, N.; Baydogan, S.; Horne, W.; et al. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J. Exp. Med. 2020, 217, e20190354. [Google Scholar] [CrossRef]
  107. Chen, J.; Ye, X.; Pitmon, E.; Lu, M.; Wan, J.; Jellison, E.R.; Adler, A.J.; Vella, A.T.; Wang, K. IL-17 inhibits CXCL9/10-mediated recruitment of CD8(+) cytotoxic T cells and regulatory T cells to colorectal tumors. J. Immunother. Cancer 2019, 7, 324. [Google Scholar] [CrossRef]
  108. Dadaglio, G.; Fayolle, C.; Oberkampf, M.; Tang, A.; Rudilla, F.; Couillin, I.; Torheim, E.A.; Rosenbaum, P.; Leclerc, C. IL-17 suppresses the therapeutic activity of cancer vaccines through the inhibition of CD8(+) T-cell responses. Oncoimmunology 2020, 9, 1758606. [Google Scholar] [CrossRef]
  109. Zhang, Y.; Lazarus, J.; Steele, N.G.; Yan, W.; Lee, H.J.; Nwosu, Z.C.; Halbrook, C.J.; Menjivar, R.E.; Kemp, S.B.; Sirihorachai, V.R.; et al. Regulatory T-cell Depletion Alters the Tumor Microenvironment and Accelerates Pancreatic Carcinogenesis. Cancer Discov. 2020, 10, 422–439. [Google Scholar] [CrossRef] [Green Version]
  110. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [Green Version]
  111. Tang, D.; Kang, R.; Cheh, C.W.; Livesey, K.M.; Liang, X.; Schapiro, N.E.; Benschop, R.; Sparvero, L.J.; Amoscato, A.A.; Tracey, K.J.; et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 2010, 29, 5299–5310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Schiraldi, M.; Raucci, A.; Muñoz, L.M.; Livoti, E.; Celona, B.; Venereau, E.; Apuzzo, T.; De Marchis, F.; Pedotti, M.; Bachi, A.; et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. J. Exp. Med. 2012, 209, 551–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Chen, Y.; Kim, J.; Yang, S.; Wang, H.; Wu, C.J.; Sugimoto, H.; LeBleu, V.S.; Kalluri, R. Type I collagen deletion in αSMA(+) myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021, 39, 548–565. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, A.; Qian, Y.; Ye, Z.; Chen, H.; Xie, H.; Zhou, L.; Shen, Y.; Zheng, S. Cancer-associated fibroblasts promote M2 polarization of macrophages in pancreatic ductal adenocarcinoma. Cancer Med. 2017, 6, 463–470. [Google Scholar] [CrossRef] [PubMed]
  115. Allen, J.L.; Cooke, M.E.; Alliston, T. ECM stiffness primes the TGFβ pathway to promote chondrocyte differentiation. Mol. Biol. Cell 2012, 23, 3731–3742. [Google Scholar] [CrossRef]
  116. Huber, M.; Brehm, C.U.; Gress, T.M.; Buchholz, M.; Alashkar Alhamwe, B.; von Strandmann, E.P.; Slater, E.P.; Bartsch, J.W.; Bauer, C.; Lauth, M. The Immune Microenvironment in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7307. [Google Scholar] [CrossRef] [PubMed]
  117. Zhu, X.; Zhou, W. The Emerging Regulation of VEGFR-2 in Triple-Negative Breast Cancer. Front. Endocrinol. 2015, 6, 159. [Google Scholar] [CrossRef] [Green Version]
  118. Tirpe, A.; Gulei, D.; Tirpe, G.R.; Nutu, A.; Irimie, A.; Campomenosi, P.; Pop, L.A.; Berindan-Neagoe, I. Beyond Conventional: The New Horizon of Anti-Angiogenic microRNAs in Non-Small Cell Lung Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 8002. [Google Scholar] [CrossRef]
  119. Zhao, J.; Xiao, Z.; Li, T.; Chen, H.; Yuan, Y.; Wang, Y.A.; Hsiao, C.H.; Chow, D.S.; Overwijk, W.W.; Li, C. Stromal Modulation Reverses Primary Resistance to Immune Checkpoint Blockade in Pancreatic Cancer. ACS Nano 2018, 12, 9881–9893. [Google Scholar] [CrossRef]
  120. Zhou, X.; Liu, Y.; Hu, M.; Wang, M.; Liu, X.; Huang, L. Relaxin gene delivery modulates macrophages to resolve cancer fibrosis and synergizes with immune checkpoint blockade therapy. Sci. Adv. 2021, 7, eabb6596. [Google Scholar] [CrossRef]
  121. Kanai, A.J.; Konieczko, E.M.; Bennett, R.G.; Samuel, C.S.; Royce, S.G. Relaxin and fibrosis: Emerging targets, challenges, and future directions. Mol. Cell. Endocrinol. 2019, 487, 66–74. [Google Scholar] [CrossRef] [PubMed]
  122. Jiang, H.; Hegde, S.; Knolhoff, B.L.; Zhu, Y.; Herndon, J.M.; Meyer, M.A.; Nywening, T.M.; Hawkins, W.G.; Shapiro, I.M.; Weaver, D.T.; et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016, 22, 851–860. [Google Scholar] [CrossRef] [PubMed]
  123. Gorchs, L.; Fernández Moro, C.; Bankhead, P.; Kern, K.P.; Sadeak, I.; Meng, Q.; Rangelova, E.; Kaipe, H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4(+) and CD8(+) T-Cells. Front. Immunol. 2019, 10, 847. [Google Scholar] [CrossRef] [PubMed]
  124. Ireland, L.; Luckett, T.; Schmid, M.C.; Mielgo, A. Blockade of Stromal Gas6 Alters Cancer Cell Plasticity, Activates NK Cells, and Inhibits Pancreatic Cancer Metastasis. Front. Immunol. 2020, 11, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Tanaka, M.; Siemann, D.W. Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers 2020, 12, 1850. [Google Scholar] [CrossRef]
  126. Zdżalik-Bielecka, D.; Poświata, A.; Kozik, K.; Jastrzębski, K.; Schink, K.O.; Brewińska-Olchowik, M.; Piwocka, K.; Stenmark, H.; Miączyńska, M. The GAS6-AXL signaling pathway triggers actin remodeling that drives membrane ruffling, macropinocytosis, and cancer-cell invasion. Proc. Natl. Acad. Sci. USA 2021, 118, e2024596118. [Google Scholar] [CrossRef]
  127. Kamionka, E.M.; Qian, B.; Gross, W.; Bergmann, F.; Hackert, T.; Beretta, C.A.; Dross, N.; Ryschich, E. Collagen Organization Does Not Influence T-Cell Distribution in Stroma of Human Pancreatic Cancer. Cancers 2021, 13, 3648. [Google Scholar] [CrossRef]
  128. Lawal, B.; Lin, L.C.; Lee, J.C.; Chen, J.H.; Bekaii-Saab, T.S.; Wu, A.T.H.; Ho, C.L. Multi-Omics Data Analysis of Gene Expressions and Alterations, Cancer-Associated Fibroblast and Immune Infiltrations, Reveals the Onco-Immune Prognostic Relevance of STAT3/CDK2/4/6 in Human Malignancies. Cancers 2021, 13, 954. [Google Scholar] [CrossRef]
  129. MacNeil, T.; Vathiotis, I.A.; Shafi, S.; Aung, T.N.; Zugazagoitia, J.; Gruver, A.M.; Driscoll, K.; Rimm, D.L. Multiplex Quantitative Analysis of Tumor-Infiltrating Lymphocytes, Cancer-Associated Fibroblasts, and CD200 in Pancreatic Cancer. Cancers 2021, 13, 5501. [Google Scholar] [CrossRef]
  130. Tahkola, K.; Ahtiainen, M.; Mecklin, J.P.; Kellokumpu, I.; Laukkarinen, J.; Tammi, M.; Tammi, R.; Väyrynen, J.P.; Böhm, J. Stromal hyaluronan accumulation is associated with low immune response and poor prognosis in pancreatic cancer. Sci. Rep. 2021, 11, 12216. [Google Scholar] [CrossRef]
  131. Nizri, E.; Bar-David, S.; Aizic, A.; Sternbach, N.; Lahat, G.; Wolf, I.; Klausner, J. Desmoplasia in Lymph Node Metastasis of Pancreatic Adenocarcinoma Reveals Activation of Cancer-Associated Fibroblasts Pattern and T-helper 2 Immune Cell Infiltration. Pancreas 2019, 48, 367–373. [Google Scholar] [CrossRef] [PubMed]
  132. Sadozai, H.; Acharjee, A.; Eppenberger-Castori, S.; Gloor, B.; Gruber, T.; Schenk, M.; Karamitopoulou, E. Distinct Stromal and Immune Features Collectively Contribute to Long-Term Survival in Pancreatic Cancer. Front. Immunol. 2021, 12, 643529. [Google Scholar] [CrossRef] [PubMed]
  133. Mace, T.A.; Shakya, R.; Pitarresi, J.R.; Swanson, B.; McQuinn, C.W.; Loftus, S.; Nordquist, E.; Cruz-Monserrate, Z.; Yu, L.; Young, G.; et al. IL-6 and PD-L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer. Gut 2018, 67, 320–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Principe, D.R.; Park, A.; Dorman, M.J.; Kumar, S.; Viswakarma, N.; Rubin, J.; Torres, C.; McKinney, R.; Munshi, H.G.; Grippo, P.J.; et al. TGFβ Blockade Augments PD-1 Inhibition to Promote T-Cell-Mediated Regression of Pancreatic Cancer. Mol. Cancer Ther. 2019, 18, 613–620. [Google Scholar] [CrossRef] [Green Version]
  135. Seo, Y.D.; Jiang, X.; Sullivan, K.M.; Jalikis, F.G.; Smythe, K.S.; Abbasi, A.; Vignali, M.; Park, J.O.; Daniel, S.K.; Pollack, S.M.; et al. Mobilization of CD8(+) T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 3934–3945. [Google Scholar] [CrossRef] [Green Version]
  136. Bockorny, B.; Semenisty, V.; Macarulla, T.; Borazanci, E.; Wolpin, B.M.; Stemmer, S.M.; Golan, T.; Geva, R.; Borad, M.J.; Pedersen, K.S.; et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: The COMBAT trial. Nat. Med. 2020, 26, 878–885. [Google Scholar] [CrossRef]
  137. Van Audenaerde, J.R.; Marcq, E.; von Scheidt, B.; Davey, A.S.; Oliver, A.J.; De Waele, J.; Quatannens, D.; Van Loenhout, J.; Pauwels, P.; Roeyen, G.; et al. Novel combination immunotherapy for pancreatic cancer: Potent anti-tumor effects with CD40 agonist and interleukin-15 treatment. Clin. Transl. Immunol. 2020, 9, e1165. [Google Scholar] [CrossRef]
  138. Panni, R.Z.; Herndon, J.M.; Zuo, C.; Hegde, S.; Hogg, G.D.; Knolhoff, B.L.; Breden, M.A.; Li, X.; Krisnawan, V.E.; Khan, S.Q.; et al. Agonism of CD11b reprograms innate immunity to sensitize pancreatic cancer to immunotherapies. Sci. Transl. Med. 2019, 11, eaau9240. [Google Scholar] [CrossRef]
  139. Michaelis, K.A.; Norgard, M.A.; Zhu, X.; Levasseur, P.R.; Sivagnanam, S.; Liudahl, S.M.; Burfeind, K.G.; Olson, B.; Pelz, K.R.; Angeles Ramos, D.M.; et al. The TLR7/8 agonist R848 remodels tumor and host responses to promote survival in pancreatic cancer. Nat. Commun. 2019, 10, 4682. [Google Scholar] [CrossRef] [Green Version]
  140. Ajina, R.; Malchiodi, Z.X.; Fitzgerald, A.A.; Zuo, A.; Wang, S.; Moussa, M.; Cooper, C.J.; Shen, Y.; Johnson, Q.R.; Parks, J.M.; et al. Antitumor T-cell Immunity Contributes to Pancreatic Cancer Immune Resistance. Cancer Immunol. Res. 2021, 9, 386–400. [Google Scholar] [CrossRef]
  141. Herting, C.J.; Karpovsky, I.; Lesinski, G.B. The tumor microenvironment in pancreatic ductal adenocarcinoma: Current perspectives and future directions. Cancer Metastasis Rev. 2021, 40, 675–689. [Google Scholar] [CrossRef] [PubMed]
  142. Sonbol, M.B.; Ahn, D.H.; Goldstein, D.; Okusaka, T.; Tabernero, J.; Macarulla, T.; Reni, M.; Li, C.P.; O’Neil, B.; Van Cutsem, E.; et al. CanStem111P trial: A Phase III study of napabucasin plus nab-paclitaxel with gemcitabine. Future Oncol. 2019, 15, 1295–1302. [Google Scholar] [CrossRef] [PubMed]
  143. Jonker, D.J.; Nott, L.; Yoshino, T.; Gill, S.; Shapiro, J.; Ohtsu, A.; Zalcberg, J.; Vickers, M.M.; Wei, A.C.; Gao, Y.; et al. Napabucasin versus placebo in refractory advanced colorectal cancer: A randomised phase 3 trial. Lancet Gastroenterol. Hepatol. 2018, 3, 263–270. [Google Scholar] [CrossRef]
  144. Taniguchi, H.; Masuishi, T.; Kawazoe, A.; Muro, K.; Kadowaki, S.; Bando, H.; Iino, S.; Kageyama, R.; Yoshino, T. Phase I study of napabucasin in combination with FOLFIRI + bevacizumab in Japanese patients with metastatic colorectal cancer. Int. J. Clin. Oncol. 2021, 26, 2017–2024. [Google Scholar] [CrossRef] [PubMed]
  145. San-Miguel, J.; Bladé, J.; Shpilberg, O.; Grosicki, S.; Maloisel, F.; Min, C.K.; Polo Zarzuela, M.; Robak, T.; Prasad, S.V.; Tee Goh, Y.; et al. Phase 2 randomized study of bortezomib-melphalan-prednisone with or without siltuximab (anti–IL-6) in multiple myeloma. Blood 2014, 123, 4136–4142. [Google Scholar] [CrossRef] [Green Version]
  146. Brighton, T.A.; Khot, A.; Harrison, S.J.; Ghez, D.; Weiss, B.M.; Kirsch, A.; Magen, H.; Gironella, M.; Oriol, A.; Streetly, M.; et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Study of Siltuximab in High-Risk Smoldering Multiple Myeloma. Clin. Cancer Res. 2019, 25, 3772–3775. [Google Scholar] [CrossRef] [Green Version]
  147. Uy, G.L.; Rettig, M.P.; Stone, R.M.; Konopleva, M.Y.; Andreeff, M.; McFarland, K.; Shannon, W.; Fletcher, T.R.; Reineck, T.; Eades, W.; et al. A phase 1/2 study of chemosensitization with plerixafor plus G-CSF in relapsed or refractory acute myeloid leukemia. Blood Cancer J. 2017, 7, e542. [Google Scholar] [CrossRef]
  148. Uy, G.L.; Rettig, M.P.; Motabi, I.H.; McFarland, K.; Trinkaus, K.M.; Hladnik, L.M.; Kulkarni, S.; Abboud, C.N.; Cashen, A.F.; Stockerl-Goldstein, K.E.; et al. A phase 1/2 study of chemosensitization with the CXCR4 antagonist plerixafor in relapsed or refractory acute myeloid leukemia. Blood 2012, 119, 3917–3924. [Google Scholar] [CrossRef]
  149. Dessureault, S.; Noyes, D.; Lee, D.; Dunn, M.; Janssen, W.; Cantor, A.; Sotomayor, E.; Messina, J.; Antonia, S.J. A phase-I trial using a universal GM-CSF-producing and CD40L-expressing bystander cell line (GM.CD40L) in the formulation of autologous tumor cell-based vaccines for cancer patients with stage IV disease. Ann. Surg. Oncol. 2007, 14, 869–884. [Google Scholar] [CrossRef]
  150. Gray, J.E.; Chiappori, A.; Williams, C.C.; Tanvetyanon, T.; Haura, E.B.; Creelan, B.C.; Kim, J.; Boyle, T.A.; Pinder-Schenck, M.; Khalil, F.; et al. A phase I/randomized phase II study of GM.CD40L vaccine in combination with CCL21 in patients with advanced lung adenocarcinoma. Cancer Immunol. Immunother. 2018, 67, 1853–1862. [Google Scholar] [CrossRef] [Green Version]
  151. Plesca, I.; Benešová, I.; Beer, C.; Sommer, U.; Müller, L.; Wehner, R.; Heiduk, M.; Aust, D.; Baretton, G.; Bachmann, M.P.; et al. Clinical Significance of Tumor-Infiltrating Conventional and Plasmacytoid Dendritic Cells in Pancreatic Ductal Adenocarcinoma. Cancers 2022, 14, 1216. [Google Scholar] [CrossRef] [PubMed]
  152. Gilardini Montani, M.S.; Benedetti, R.; Piconese, S.; Pulcinelli, F.M.; Timperio, A.M.; Romeo, M.A.; Masuelli, L.; Mattei, M.; Bei, R.; D’Orazi, G.; et al. PGE2 Released by Pancreatic Cancer Cells Undergoing ER Stress Transfers the Stress to DCs Impairing Their Immune Function. Mol. Cancer Ther. 2021, 20, 934–945. [Google Scholar] [CrossRef] [PubMed]
  153. Guo, J.; Liao, M.; Hu, X.; Wang, J. Tumour-Derived Reg3A Educates Dendritic Cells to Promote Pancreatic Cancer Progression. Mol. Cells 2021, 44, 647–657. [Google Scholar] [CrossRef]
  154. Giri, B.; Sharma, P.; Jain, T.; Ferrantella, A.; Vaish, U.; Mehra, S.; Garg, B.; Iyer, S.; Sethi, V.; Malchiodi, Z.; et al. Hsp70 modulates immune response in pancreatic cancer through dendritic cells. Oncoimmunology 2021, 10, 1976952. [Google Scholar] [CrossRef] [PubMed]
  155. Sadeghlar, F.; Vogt, A.; Mohr, R.U.; Mahn, R.; van Beekum, K.; Kornek, M.; Weismüller, T.J.; Branchi, V.; Matthaei, H.; Toma, M.; et al. Induction of cytotoxic effector cells towards cholangiocellular, pancreatic, and colorectal tumor cells by activation of the immune checkpoint CD40/CD40L on dendritic cells. Cancer Immunol. Immunother. 2021, 70, 1451–1464. [Google Scholar] [CrossRef]
  156. Kim, D.; Lee, S.; Na, K. Immune Stimulating Antibody-Photosensitizer Conjugates via Fc-Mediated Dendritic Cell Phagocytosis and Phototriggered Immunogenic Cell Death for KRAS-Mutated Pancreatic Cancer Treatment. Small 2021, 17, 2006650. [Google Scholar] [CrossRef]
  157. Salah, A.; Wang, H.; Li, Y.; Ji, M.; Ou, W.B.; Qi, N.; Wu, Y. Insights into Dendritic Cells in Cancer Immunotherapy: From Bench to Clinical Applications. Front. Cell Dev. Biol. 2021, 9, 686544. [Google Scholar] [CrossRef]
  158. Kvedaraite, E.; Ginhoux, F. Human dendritic cells in cancer. Sci. Immunol. 2022, 7, eabm9409. [Google Scholar] [CrossRef]
  159. Liu, Y.; Cheng, L.; Song, X.; Li, C.; Zhang, J.; Wang, L. A TP53-associated immune prognostic signature for the prediction of the overall survival and therapeutic responses in pancreatic cancer. Math. Biosci. Eng. 2022, 19, 191–208. [Google Scholar] [CrossRef]
  160. Kandimalla, R.; Tomihara, H.; Banwait, J.K.; Yamamura, K.; Singh, G.; Baba, H.; Goel, A. A 15-Gene Immune, Stromal, and Proliferation Gene Signature that Significantly Associates with Poor Survival in Patients with Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2020, 26, 3641–3648. [Google Scholar] [CrossRef] [Green Version]
  161. Li, Y.; Yao, P.; Zhao, K.; Ye, Z.; Zhang, H.; Cao, J.; Zhang, S.; Xing, C. Individualized prognostic signature for pancreatic carcinoma validated by integrating immune-related gene pairs (IRGPs). Bioengineered 2021, 12, 88–95. [Google Scholar] [CrossRef] [PubMed]
  162. Meng, Z.; Ren, D.; Zhang, K.; Zhao, J.; Jin, X.; Wu, H. Using ESTIMATE algorithm to establish an 8-mRNA signature prognosis prediction system and identify immunocyte infiltration-related genes in Pancreatic adenocarcinoma. Aging 2020, 12, 5048–5070. [Google Scholar] [CrossRef]
  163. Zhang, C.; Ding, J.; Xu, X.; Liu, Y.; Huang, W.; Da, L.; Ma, Q.; Chen, S. Tumor Microenvironment Characteristics of Pancreatic Cancer to Determine Prognosis and Immune-Related Gene Signatures. Front. Mol. Biosci. 2021, 8, 645024. [Google Scholar] [CrossRef] [PubMed]
  164. Liu, J.; Liu, Q.; Zhang, X.; Cui, M.; Li, T.; Zhang, Y.; Liao, Q. Immune subtyping for pancreatic cancer with implication in clinical outcomes and improving immunotherapy. Cancer Cell Int. 2021, 21, 137. [Google Scholar] [CrossRef] [PubMed]
  165. Mahajan, U.M.; Langhoff, E.; Goni, E.; Costello, E.; Greenhalf, W.; Halloran, C.; Ormanns, S.; Kruger, S.; Boeck, S.; Ribback, S.; et al. Immune Cell and Stromal Signature Associated with Progression-Free Survival of Patients with Resected Pancreatic Ductal Adenocarcinoma. Gastroenterology 2018, 155, 1625–1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. De Santiago, I.; Yau, C.; Heij, L.; Middleton, M.R.; Markowetz, F.; Grabsch, H.I.; Dustin, M.L.; Sivakumar, S. Immunophenotypes of pancreatic ductal adenocarcinoma: Meta-analysis of transcriptional subtypes. Int. J. Cancer 2019, 145, 1125–1137. [Google Scholar] [CrossRef]
  167. Akce, M.; Zaidi, M.Y.; Waller, E.K.; El-Rayes, B.F.; Lesinski, G.B. The Potential of CAR T Cell Therapy in Pancreatic Cancer. Front. Immunol. 2018, 9, 2166. [Google Scholar] [CrossRef]
Figure 1. Mechanisms underlying the interplay among pancreatic cancer cells, fibroblasts, and immune cells. (A) Collagen I around fibroblasts (brown) stimulates pancreatic cancer cells (red) and increases the Rho/SOX9 activation, which leads to CXCL5 expression to activate myeloid cells (purple). Myeloid cells, in turn, express arginase 1 to suppress CD8+ T cell (green). (B) Fibroblasts (brown) modulate macrophage (purple) polarization and function via ROS, and M2 polarized macrophages assist the proliferation of pancreatic cancer cell (red) via macrophage colony-stimulating factor (M-CSF). (C) Treg (gray) express TGFβ to stimulate the expression of CCR1 ligands in fibroblasts (brown), and the CCR1 ligands recruit myeloid cells (purple) to promote the growth of pancreatic cancer cells (red). (D) Fibroblasts (brown) express CXCL12 to suppress CD8+ T cells (green) and block immune surveillance against pancreatic cancer cells (red).
Figure 1. Mechanisms underlying the interplay among pancreatic cancer cells, fibroblasts, and immune cells. (A) Collagen I around fibroblasts (brown) stimulates pancreatic cancer cells (red) and increases the Rho/SOX9 activation, which leads to CXCL5 expression to activate myeloid cells (purple). Myeloid cells, in turn, express arginase 1 to suppress CD8+ T cell (green). (B) Fibroblasts (brown) modulate macrophage (purple) polarization and function via ROS, and M2 polarized macrophages assist the proliferation of pancreatic cancer cell (red) via macrophage colony-stimulating factor (M-CSF). (C) Treg (gray) express TGFβ to stimulate the expression of CCR1 ligands in fibroblasts (brown), and the CCR1 ligands recruit myeloid cells (purple) to promote the growth of pancreatic cancer cells (red). (D) Fibroblasts (brown) express CXCL12 to suppress CD8+ T cells (green) and block immune surveillance against pancreatic cancer cells (red).
Biomedicines 10 00926 g001
Figure 2. Treating pancreatic cancer with CD8+ T cell-orientated approaches. Potential CD8+ T cell-orientated treatments for pancreatic cancer were reviewed and blockade of IL6/TGFβ/CXCR4/STAT and activation of CD40/IL15/CD11b were proposed to be effective in pancreatic cancer therapy. Solid arrow represents direct effect, while dotted arrow represents indirect effect.
Figure 2. Treating pancreatic cancer with CD8+ T cell-orientated approaches. Potential CD8+ T cell-orientated treatments for pancreatic cancer were reviewed and blockade of IL6/TGFβ/CXCR4/STAT and activation of CD40/IL15/CD11b were proposed to be effective in pancreatic cancer therapy. Solid arrow represents direct effect, while dotted arrow represents indirect effect.
Biomedicines 10 00926 g002
Figure 3. Scheme shows the potential effects of fibroblast on immune cell recruitment/function in pancreatic cancer. Based on the findings of recent studies, the potential effect of fibroblast on recruitment and function of CD8+ T cell, Treg, macrophage, MDSC, and DC in pancreatic cancer is proposed.
Figure 3. Scheme shows the potential effects of fibroblast on immune cell recruitment/function in pancreatic cancer. Based on the findings of recent studies, the potential effect of fibroblast on recruitment and function of CD8+ T cell, Treg, macrophage, MDSC, and DC in pancreatic cancer is proposed.
Biomedicines 10 00926 g003
Table 1. Mechanisms underlying the interplay among pancreatic cancer cells, fibroblasts, and immune cells.
Table 1. Mechanisms underlying the interplay among pancreatic cancer cells, fibroblasts, and immune cells.
MoleculeCell TypeMechanismReference
TGFβTregTGFβ from Treg activates SMA+ myofibroblasts and their expression of CCR1 ligands to recruit MDSC [109]
CXCL12CAFCXCL12 from FAP+ CAF increases T cell exclusion and cancer growth[110]
Type I collagenMyofibroblastType I collagen from myofibroblast decreases SOX9 expression and subsequently increases CXCL5 in PDAC to recruit MDSC[113]
ROSCAFROS from CAF increases monocyte differentiation into M2 macrophages and their production of M-CSF to increase the invasiveness of PDAC[114]
Table 2. Correlation of the alterations in cancer cells, fibroblasts, and immune cells in co-culture, animal models, and clinical samples.
Table 2. Correlation of the alterations in cancer cells, fibroblasts, and immune cells in co-culture, animal models, and clinical samples.
MoleculeCell TypeAssociationReference
NAPDACCollagen/fibroblast↓ post FAKi treatment[122]
Macrophage↓ post FAKi treatment
G-MDSC↓ post FAKi treatment
NAPDACCAFPD-L1/PD-L2↑[123]
CD4/CD8 proliferation ↓
CD4/CD8 co-inhibitory marker↑
CD8 function ↓
NAPDACVimentin↓ post α-Gas6 treatment[124]
NK↓ post α-Gas6 treatment
NAPDACCollagenDensity not altered[127]
T cellInfiltration not altered
CDK2/4/6PDACCAFCo-occurrence ↑[128]
STAT3CAFCo-occurrence ↑
ImmunityOnco-immune signature ↑
NAPDACCD4Disease progression ↓[129]
CD8Disease progression ↓
Thy-1+ CAFDisease progression ↓
FAP+ CAFDisease progression ↑
Stromal hyaluronan accumulationNACD8/CD3-based immune cell score[130]
NAPDACDesmoplasiaCOL11A1/COL11A2/COL1A1/TGF-β mRNA ↑[131]
Th2 immunityGATA3 ↑
NAPDACα-SMA/fibrosis↑ in STS[132]
NAPDACCD68/CD163↑ in STS
CD4↓ in STS
iNOS↓ in STS
Foxp3↑ in STS
B cell/DC↓ in STS
NAPDACMetabolic active CAF (meCAF)↑ in dense (high desmoplasia) group[55]
CD8↑ in loose (low desmoplasia) group
Response to α-PD-1↑ in loose group
FAK, focal adhesion kinase; STS, short-term survivors; ↑, increment; ↓, decrement.
Table 3. Summary of clinical trials targeting IL6, TGFβ, CXCR4, CD40, and STAT for cancer treatment.
Table 3. Summary of clinical trials targeting IL6, TGFβ, CXCR4, CD40, and STAT for cancer treatment.
Clinical Trial IDTreatmentCancer TypeReference
NCT02993731Napabucasin + nab-paclitaxel(+gemcitabine)Pancreas[142]
NCT00911859Siltuximab(+velcade-melphalan-prednisone)Multiple myeloma[145]
NCT01484275SiltuximabSmoldering multiple myeloma[146]
NCT00906945 G-CSF + plerixafor + mitoxantrone + etoposide + cytarabineAcute myeloid leukemia[147]
NCT00512252plerixafor + mitoxantrone + etoposide + cytarabineAcute myeloid leukemia[148]
NCT00101166GM.CD40L vaccinationMelanoma[149]
NCT01433172 GM.CD40L vaccination (+CCL21)Lung[150]
Napabucasin, STAT3 inhibitor; siltuximab, IL6 antibody; plerixafor, CXCR4 inhibitor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hung, Y.-H.; Chen, L.-T.; Hung, W.-C. The Trinity: Interplay among Cancer Cells, Fibroblasts, and Immune Cells in Pancreatic Cancer and Implication of CD8+ T Cell-Orientated Therapy. Biomedicines 2022, 10, 926. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040926

AMA Style

Hung Y-H, Chen L-T, Hung W-C. The Trinity: Interplay among Cancer Cells, Fibroblasts, and Immune Cells in Pancreatic Cancer and Implication of CD8+ T Cell-Orientated Therapy. Biomedicines. 2022; 10(4):926. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040926

Chicago/Turabian Style

Hung, Yu-Hsuan, Li-Tzong Chen, and Wen-Chun Hung. 2022. "The Trinity: Interplay among Cancer Cells, Fibroblasts, and Immune Cells in Pancreatic Cancer and Implication of CD8+ T Cell-Orientated Therapy" Biomedicines 10, no. 4: 926. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040926

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop