Next Article in Journal
Real-World Management of High-Risk Prostate Cancer Post-Radical Prostatectomy: Insights from a Regional Quality Collaborative
Previous Article in Journal
Deep Learning for Automated Ventricle and Periventricular Space Segmentation on CT and T1CE MRI in Neuro-Oncology Patients
Previous Article in Special Issue
Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies

by
Franciszek Glapiński
1,
Weronika Zając
1,
Marta Fudalej
2,3,
Andrzej Deptała
2,
Aleksandra Czerw
4,5,
Katarzyna Sygit
6,
Remigiusz Kozłowski
7 and
Anna Badowska-Kozakiewicz
2,*
1
Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
2
Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
3
Department of Oncology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
4
Department of Health Economics and Medical Law, Medical University of Warsaw, 02-091 Warsaw, Poland
5
Department of Economic and System Analyses, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland
6
Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
7
Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(10), 1599; https://doi.org/10.3390/cancers17101599
Submission received: 7 April 2025 / Revised: 30 April 2025 / Accepted: 6 May 2025 / Published: 8 May 2025

Simple Summary

Pancreatic cancer (PC) remains one of the most significant challenges of oncology to this day due to its inadequate response to conventional treatment, gradual rise in incidence, and poor five-year survival rates. Increasing attention is being paid to the tumor microenvironment (TME), which comprises a diverse array of cell types. The TME potentially brings a new turn in PC survival. This study aims to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PC. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.

Abstract

Pancreatic cancer (PC), with pancreatic ductal adenocarcinoma (PDAC) comprising about 90% of all cases, is one of the most aggressive and lethal solid tumors. PDAC remains one of the most significant challenges of oncology to this day due to its inadequate response to conventional treatment, gradual rise in incidence since 2004, and poor five-year survival rates. As cancer cells are the primary adversary in this uneven fight, they remain the primary research target. Nevertheless, increasing attention is being paid to the tumor microenvironment (TME). The most crucial TME constellation components are immune cells, especially macrophages, stellate cells and lymphocytes, fibroblasts, bacterial and fungal microflora, and neuronal cells. Depending on the particular phenotype of these cells, the composition of the microenvironment, and the cell ratio, patients can experience different disease outcomes and varying vulnerability to treatment approaches. This study aims to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PC. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.

1. Introduction

Pancreatic cancer (PC), with pancreatic ductal adenocarcinoma (PDAC) comprising about 90% of all cases, is one of the most aggressive and lethal solid tumors [1]. PDAC derives from ductal-like cells, which may originate from mutated ductal epithelial cells or acinar cells that undergo metaplasia to adopt a ductal phenotype. These processes are often triggered by stressors such as alcohol and nicotine consumption, chronic inflammation, diabetes, and poor nutrition [2]. Driver mutations include activating mutations of protooncogene KRAS and the inactivation of tumor suppressor genes such as SMAD4, CDKN2A, and TP53 [3]. Pancreatic cancer typically develops from well-recognized precursor lesions, such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), and mucinous cystic neoplasms (MCN). Pancreatic cancer remains one of the greatest challenges in oncology, driven by its increasing incidence, lack of reliable biomarkers, and late detection, which together contribute to poor treatment responses and low five-year survival rates [4]. Moreover, PDAC features a dense stromal microenvironment characterized by notable cellular and spatial variation, which plays a crucial role in shaping disease behavior and resistance to treatment [5]. According to the Surveillance, Epidemiology, and End Results Programme (SEER) Database, more than 67,000 new cases are expected in the United States alone. It will constitute 8.4% of all cancer deaths (almost 50,000 deaths per year), and the 5-year survival rate will be equal to only 13.5% [6]. According to the Polish National Cancer Registry, the incidence of PDAC in Poland increased by 18%, and mortality rose by 45% between 1996 and 2018. It is predicted that, by 2030, pancreatic cancer may become the second leading cause of cancer-related deaths in Poland [7]. Even though surgical resection with negative margins and adjuvant chemotherapy that target cancer cells is proven to be quite successful, in most cases, either the tumor is diagnosed at an unresectable stage, or the chance of recurrence is high, affecting the 5-year survival rate [3]. The first-line treatment chemotherapy scheme used in PDAC is a combination of fluorouracil, leucovorin, irinotecan, and oxaliplatin, known as FOLFIRNOX. The other scheme is gemcitabine associated with nab-paclitaxel. [8]. The roles of radiotherapy and immunotherapy are still being discussed, as they have not been validated by a substantial number of fully randomized trials [9]. There was a study that attempted to divide PDAC patients into subgroups based on CT images for the adjustment of treatment decisions for each patient, but the results are inconclusive, with more research required to reach a consensus. PC, even when resectable (R-PDAC) or borderline-resectable (BR-PDAC), should be presumed to be a systemic disease, and all patients should receive multimodal treatment [10]. Nevertheless, less than half of the patients respond to therapy, and they could suffer from the side effects of chemotherapeutics [11]. Genetics might explain the difference between respondents and non-respondents, but more evidence is required to use it in clinical settings [12]. Given its dramatic influence on cancer development and progression, it has prompted the exploration of novel strategies to target the tumor microenvironment rather than the parenchyma [13].
The aim of this work is to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PCs. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.

2. The Structure of the Microenvironment

PC’s tumor microenvironment (TME), also known as its stroma, is responsible for maintaining and stabilizing the parenchyma, which is composed of cancer cells [14]. It is characterized by desmoplasia, a process in which dense fibrous connective tissue replaces the normal parenchyma of the organ. It is the primary pathophysiological reason for the pancreas’s malfunction during the progression of PDAC [15]. The stroma itself can represent up to 90% of a tumor mass. It is formed by cellular components such as stellate cells (PSCs), which can constitute more than 50% of the mass of the stroma or cancer-associated fibroblasts, as well as non-cellular components, which is mainly composed of the extracellular matrix (ECM), made up of collagens, glycosaminoglycans, and proteoglycans [13,16]. Additionally, immunological components could be present, mainly in the form of regulatory T lymphocytes (Treg lymphocytes), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), together with cytokines, growth factors, and ECM-metabolizing enzymes secreted by those cells; the cells mentioned above present immunosuppressive functions [17,18]. The components of the TME maintain a connection with cancer cells indirectly, influencing tumor progression, metastasis development, and treatment resistance [19].

2.1. Pancreatic Stellate Cells (PSCs)

PSCs are star-shaped cells presented physiologically in the exocrine part of the pancreas in periacinar, perivascular, and periductal spaces in a quiescent state. Their proliferation rate is low, and they produce minute amounts of ECM. They are characterized by nestin expression and are involved in storing vitamin-A-rich lipid droplets, antigen phagocytosis, and the induction of amylase secretion [20]. They become activated during inflammation or the development of pancreatic tumors due to exposure to cytokines and growth factors produced by evolving cancer cells, inflammatory cells, acinar cells, platelets, and later by newly activated stellate cells, which means that their active state is maintained by paracrine and autocrine mechanisms [17]. Previously mentioned cytokines and chemokines include platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), transforming growth factor β (TGF–β), tumor necrosis factor-alpha (TNF-α), connective tissue growth factor (CTGF) and interleukin 1 (IL-1), IL-6, IL-8, and IL-10 [16,21,22]. The cells secreting individual factors responsible for activating stellate cells are presented in Table 1.
Other factors contributing to the activation of PSCs include hyperglycemia, hypoxia, hypoperfusion, and oxidative stress, which often result from chronic inflammation [16,23]. Additionally, ethanol, its metabolites, and cigarette smoking are also considered risk factors for PDAC development [16,23]. After activation, PSCs begin to express a phenotype closely resembling that of myofibroblasts, as supported by the expression of α-smooth muscle actin (αSMA), desmin, and the loss of lipid droplets [24]. PSCs’ primary role is to deposit ECM components such as collagens, fibronectin, laminin, or hyaluronic acid. Their other functions include promoting cancer cell proliferation, migration, and apoptosis inhibition [25]. Those processes are promoted by specific cytokines and growth factors such as iIL-1 or IL-6, and IL-8, insulin-like growth factor 1 (IGF1), vascular endothelial growth factor (VEGF), PDGF, FGF, CTGF, and C-X-C motif chemokine 12 (CXCL12) [11]. Tumor necrosis factor α (TNFα), also secreted by the MDSCs and M1 macrophages described later, is theorized to play a role in the cachectic effects associated with tumor progression [26]. Those molecules lead to the intensification of angiogenesis processes and the proliferation and migration of parenchymal cells, ultimately resulting in metastasis formation. It has also been postulated that PSCs are responsible for chemotherapy and radiotherapy resistance [27,28]. The importance of investigating these mechanisms is high, as patients with PDAC who develop irreversible cachexia have a life expectancy below 3 months [29].

2.2. Cancer-Associated Fibroblasts (CAFs)

CAFs are a heterogeneous group of spindle-shaped cells of mesenchymal origin (classified due to mesenchymal marker vimentin) in the connective tissue of nearly all body organs, including the pancreas [30]. They are another class of cells responsible for ECM deposition and the formation of PDAC’s stroma [31]. There are various theories regarding the origin of CAFs in the pancreatic stroma. They may be the population of cells in the stroma that arise from local fibroblasts, mesenchymal stem cells, vascular smooth muscle cells, adipocytes, or even hematopoietic stem cells. Still, they may also be derived from bone marrow fibrocytes that differentiate into fibroblasts and migrate to the organ thanks to the cytokines and chemokines released by PSCs and cancer cells [15,30,32,33]. Cytokines responsible for the attraction, activation, and proliferation of CAFs include TGF-β, FGF, CTGF, and PDGF [32]. Another theory suggests that cells differentiate from active forms of PSCs, and if activated forms of these cells are present in cancer tissue, they should be referred to as CAFs [34]. This is because they not only express fibroblastic markers such as fibroblast activation protein (FAP) and fibroblast-specific protein 1 (FSP1), but PSC’s markers as well, like desmin and, most abundantly, αSMA [32,34]. The third theory proposed by Iwano et al. (2002) suggests that CAFs originate from endothelial cells through an endothelial–to–mesenchymal transition (EMT). This mechanism may explain the presence of these cells near the tumor, as the abundance of growth factors such as TGF-β, EGF, and FGF-2 drives this change, while only a few endothelial cells are present [35]. Due to various subtypes of CAFs, the theories mentioned above may not be exclusive. Öhlund et al. (2017) described the presence of two distinctive subtypes. The first subtype is a myofibroblastic subtype (myCAF), classified by a high concentration of α-SMA. In contrast, the second subtype, characterized by low αSMA expression, is referred to as the inflammatory subtype (iCAF) and exhibits a distinct pattern of IL–6 expression which is entirely absent in myCAFs [34]. MyCAFs are cells located in the proximity of the tumor, similar to PSCs, with the same functions of ECM protein synthesis, deposition of connective tissue, and promotion of neovascularization mainly through TGF-β, stromal-derived factor-1 (SDF-1), PDGF, and VEGF. In contrast, iCASs are identified at distant locations from the tumor and are responsible for secreting inflammatory cytokines, the most abundant being IL-6 [30]. Other cytokines and chemokines include IL-8, CXCL12, and C–C motif chemokine 2 (CCL2) [34,36]. All of them can induce tumor growth, cell invasion, and proliferation, with IL–6 also being responsible for angiogenesis [30]. The third population, identified by Elyada et al. (2019), is referred to as antigen-presenting (apCAFs) due to the presence of MHC II and CD74. ApCAFs present antigens to T cells, but also have other functions and roles in tumor development [37].

How Cancer-Associated Fibroblasts Impact Tumor Behavior

The strong desmoplastic reaction was formerly appreciated as a defensive mechanism for restraining tumor growth, similar to the one in chronic pancreatitis. Several years ago, fibroblasts were shown to suppress the growth of several cell lines, a phenomenon known as neighbor suppression, suggesting their protective role. Different types of fibroblasts develop a spectrum of suppressive activity. For example, skin fibroblasts appear to have stronger inhibitory effects than those obtained from prostate, nasal polyps, or inguinal hernias [38]. Nevertheless, the optics have shifted over the last 15 years, and fibroblasts’ impact now appears to be more ambiguous [39].
CAFs are significant contributors to PDAC progression due to their ability to promote fibrosis, which puts up a physical barrier preventing drug penetration. Inhibiting or depleting CAFs is a promising method undergoing numerous investigations [40].
One of these methods is the inhibition of the SLC7A11 protein. High stromal expression of SLC7A11 is correlated with poor prognosis, unlike the high tumor expression of SLC7A11, which does not predict patient survival [41]. SLC7A11 is a membrane transporter that transfers cysteine into cells in exchange for glutamate. It is essential in maintaining adequate fibroblast cell growth because it sits at the crux of many metabolic pathways, especially glutathione synthesis [42].
Due to KRAS mutation and metabolic ratio changes, both PDAC cells and CAFs are exposed to oxidative stress. Cancer cells require hypermetabolism to sustain growth, which results in the production of reactive oxygen species (ROS) in mitochondria, NADPH oxidases (NOXs), peroxisomes, and the endoplasmic reticulum (ER) [43,44,45]. Cancer cells are equipped with numerous antioxidant systems to counteract elevated levels of reactive oxygen species (ROS). Glutathione is the most abundant nonenzymatic ROS scavenger in living cells, and its production can be regulated by metabolic shifts [46].
Inhibition of SLC7A11 with sulfasalazine, an immunosuppressive medication commonly used to treat Crohn’s disease, significantly reduces PDAC growth. Interestingly, it does not work by inhibiting NF-κB, the main pathway interrupted when using sulfasalazine to treat inflammatory bowel disease [47], but rather by inhibiting the SLC7A11 transporter [41,48]. Additionally, direct inhibition of SLC7A11 using specific small interfering RNAs (siRNAs) has been shown to reduce PDAC tumor growth, metastasis incidence, CAF activation, and fibrosis in orthotopic mouse models of PDAC. It also decreases CAF proliferation and viability by inducing cysteine starvation [41].
Cancer cells could lose the Col1a2 gene and accelerate PDAC development by promoting CD4+ cells, which aggravate immunosuppression and deplete CD8+ cells. Typically, fibroblasts produce heterotrimeric α1/α2/α1 collagen I, but, in contrast, cancer cells often lose the Col1a2 gene and start producing homotrimeric α1/α1/α1 degenerated collagen. Although the copy number of the Col1a2 gene in PDAC cells is similar to human pancreatic nestin-expressing cells, there are distinct differences in methylation patterns, which are inversely proportional to protein expression. Deleting the Collagen 1 gene delays the development of acinar-to-ductal metaplasia and intraepithelial neoplasia, especially in the early stages of PC development [49].
A new mechanism that facilitates PDAC metastasis has been recently discovered, highlighting the complex interactions between the stroma and cancer cells. PDAC cells, by secreting TGF-b, force endothelial cells and fibroblasts to produce IL-33. IL-33 works via the Il-33-ST2-p38/ERK-MYC-CXCL3 pathway and stimulates TAMs to produce CXCL3, which influences back fibroblasts by C–X–C–R motif chemokine 2 (CXCR2) on CAFs and turns them into myofibroblasts. In this manner, myofibroblasts can bind to collagen type 3 and form cell clusters, which can then migrate through the bloodstream and lymph vessels and metastasize (Figure 1 and Figure 2) [50].
The TME has a profound impact on the delivery of chemotherapeutic agents. The cellular component of PDAC is mainly made of alpha-smooth muscle actin (SMA+) expressing myofibroblasts [51]. SMA+ myofibroblasts associated with PDAC are thought to enhance collagen I production, thereby hindering drug penetration and T-cell infiltration [52]. The Sonic Hedgehog Homolog (SHH) pathway is a developmental morphogen contributing to stromal desmoplasia in various solid tumors. Absent in the adult pancreas, it reappears during inflammation and neoplasia. Interestingly, the SHH pathway has not been shown to contribute to normal differentiation of the pancreas. Ectopic administration of the SHH increases the number of mesenchymal cells [53,54]. Both the SHH ligand and its downstream signaling, including the transmembrane proteins Patched (Ptch) and Smoothened (SMO) [55], are induced in neoplastic lesions and increase during PDAC progression [53]. An elevated level of SHH induces a robust desmoplastic response in PDAC tumors by stimulating (αSMA+) fibroblast proliferation and chemotaxis, possibly through direct influence or by interacting with TGFβ signaling [56,57].

2.3. Extracellular Matrix

The most abundant protein in the body is collagen, represented mainly by types I and III. Still, other proteins that form the ECM are fibronectin and laminin. Other components include hyaluronic acid and a variety of proteoglycans, glycoproteins, and polysaccharides [15]. Olivares et al. (2017) hypothesize that collagen serves as the primary nutritional source for cancer cells. ECM proteins are visibly degraded, and the released amino acids, such as proline, may be utilized in the tricarboxylic acid cycle [58]. Enzymes involved in the degradation process are synthesized by PSCs and CAFs, which are matrix metalloproteinases 2, 3, 9, and 13 [59,60]. Their activation primarily relies on the concentration of TGF-β, which is secreted in significant quantities by CAFs [61]. ECM degradation (mainly collagenolytic) and remodeling are also responsible for tumor progression and invasion [60]. It has been suggested by Whatcott et al. (2017) that the predominance of specific ECM components correlates with the median survival of patients with PDAC. The median overall survival (OS) for patients with primary PDAC exhibiting strong stromal hyaluronic acid expression was 9.3 months, whereas it was 24.3 months for those with low hyaluronic acid staining [62]. Similarly, the same study revealed that low ECM type I collagen levels were associated with a median overall survival (OS) of 14.6 months, while high levels were associated with a median OS of 6.4 months [63].

2.4. Regulatory Lymphocytes (Treg Lymphocytes)

Treg lymphocytes are a subpopulation of T lymphocytes characteristic of the expression of CD4, CD25, and FOXP3 biomarkers. Their physiological role is to suppress the immune response. During tumor progression, they can actively suppress the anti-tumor immune response, indicating that they are essential for maintaining immunological self-tolerance [18,64]. It was reported that the percentages of CD4+CD25+FOXP3+ Tregs were significantly increased in PDAC tumor tissues compared to control pancreatic tissues. An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma]. According to Jang et al., a large proportion of these cells correlates with a poorer prognosis [18]. Their primary function is to release TGF-β and IL–10, which predominantly silence the immune response in the tumor’s proximity [65]. Several reports also state that Tregs hold a protective role, improving overall survival [66]. Further studies are needed to clarify the role of Tregs in PDAC and to determine how these findings might be translated into clinical management strategies for patients.

2.5. Myeloid-Derived Suppressor Cells (MDSCs)

MDSCs are defined as a heterogeneous group of immature myeloid cells originating in the bone marrow whose presence in other organs in the body almost always indicates chronic inflammation [67]. They are divided into two subtypes: the monocyte-resembling subtype (M–MDSC) and the polymorphonuclear-resembling subtype (PMN–MDSC) [68]. They are the most prominent group of immune cells in the TME of PDAC [65]. They are activated by cytokines present during inflammation, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL–13.
Both populations of MDSCs are responsible for the evasion of anti-tumor immune response by contributing to forming an immune-privileged niche within the tumor microenvironment [69]. They activate Treg lymphocytes and suppress CD8+ T lymphocytes and NK cells [65,67]. This is performed by the EGFR-MAPK-dependent regulation of tumoral PD-L1 expression. They also secrete immunosuppressive molecules such as arginase-1, nitric oxide, and reactive oxygen species [70]. They are also one of the cells secreting cytokines responsible for the M2 differentiation of TAMs, which further amplify the pro-tumor effects of the TME [71]. Several studies in preclinical models have shown that depleting MDSCs or inhibiting their function enhances the anti-tumor response [72].

2.6. Tumor-Associated Macrophages (TAMs)

TAMs are cells derived from monocytes and are present in the pancreas under physiological conditions. They are responsible for the phagocytosis of various pathogens and act as antigen-presenting cells for T and B lymphocytes. There are two phenotypes of TAMs. M1, present in early tumor development, promotes inflammation and is capable of releasing TNFα, which is speculated to contribute to cachexia. The interaction of interferon-gamma receptor (IFN-γR), granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR), and interleukin-12 receptor (IL-12R) on macrophages, with their respective ligands, induce M1-like polarization via activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway [73]. It is generally considered to have anti-tumor properties, and their activity is suppressed by CAFs and tumor cells by secreting cytokines such as IL-8, IL-10, and TGF-β [74]. The microenvironment favors the M2 phenotype. The differentiation process into the M2 phenotype is promoted by IL-4, IL-10, IL-13, and TGF-β, which are produced by multiple components of TME [75]. These cytokines promote differentiation by activating STAT3/NF-κB pathway [76]. They exhibit pro-tumor effects, promote anti-inflammatory responses, and correlate with a poor prognosis of the disease [68,77]. It may be because they secrete proteins such as TGF-β that promote PDAC progression and metastasis [78].

2.7. Angiogenesis

The stroma in PDAC is considered to be hypovascular. Cancer cells and specific microenvironment components such as PSCs develop primarily in highly hypoxic conditions [79]. Moreover, the tumoral stroma has a distorted distribution, in contrast to the homogeneous evenly distributed spectrum of vessels in healthy organs, which is rich in microvessels and capillaries. This phenomenon is not only characterized by the increased prevalence of arteries and arterioles further from the parenchyma, but also by the phenomenon of vascular mimicry (VM) [80]. Cancer cells in some tumors can form vessel-like structures, further complicating the typical vascular architecture [81]. It becomes an independent tumor blood supply system that occurs without the involvement of angiogenesis or endothelial cells, and it is linked to poor survival in PDAC patients. EGF and VEGF, cytokines involved in the process of vascular mimicry, are thought to trigger enhanced Notch signaling. This activation subsequently leads to upregulating mesenchymal markers, such as TWIST1 and SNAI1, and facilitates the formation of tube-like structures characteristic of VM [82]. However, it must be highlighted that vasculature in the tumor’s proximity constantly changes during its progression. Changes in the ratio of proangiogenic to anti-angiogenic factors are hypothesized to trigger the angiogenic switch during pancreatic tumor formation [83]. Initially, the tumor progresses through its avascular phase, but as the tumor increases in size, it enters a vascular phase characterized by rapid angiogenesis, which is facilitated by cytokines secreted by PSCs and CAFs, such as VEGF and FGF, that promote SHH signaling and the proliferation of endothelial cells [84,85].

3. Stromal Cell Relations and the Influence of Connective Tissue on PDAC

Unlike any other malignancies, PDAC consists mainly of stromal cells (up to 90%) and only a minority of proper epithelial cancer cells. PDAC-associated stroma cells are stellate cells, macrophages, fibroblasts, and lymphocytes. Two of the most critical cells influencing PDAC survival and treatment outcomes are CAFs and TAMs, which alter the disease course [50]. The intense effort put into exploring stromal components fueled great optimism and resulted in the testing of numerous potential therapeutic targets [86]. Nevertheless, recent years provided data that the depletion of TME components may even exacerbate PDAC and contribute to the acceleration of metastasis and tumor growth by enhancing cancer cell proliferation [87,88,89].

Epithelium–Stroma Relations: How Independent Is Stroma from Epithelium?

Using microdissection, primary reports studying concomitant tumor and stromal subtypes in PDAC revealed a partial dependence between epithelial and stromal molecular subtypes [51]. Up-to-the-minute convergent transcriptional tumor/stroma classification conducted in patient-derived xenografts (PDX) revealed two subtypes of PDAC with distinct outcomes. These subtypes uncovered specific alterations in DNA methylation, transcription, and signaling pathways involved in tumor-stromal cross-talk, e.g., hepatocyte growth factor (HGF), IGF, and SHH. Overall, the stroma, originating from surrounding host cells, is closely associated with the tumor phenotype, indicating that the composition and function of the TME may be influenced by tumor cells [90]. Mouse models of PDAC deficient in SHH demonstrated that Hedgehog inhibition leads to the absence of tumor stroma and the abundance of intratumoral blood vessels, yet increases tumor aggressiveness and metastasis [88].
Another essential factor in PDAC stroma development is retinoic acid (RA), which induces PSC deactivation and reduces extracellular matrix production by inhibiting the production of TβRII, PDGFRβ, β-catenin, and Wnt [91]. Further studies have demonstrated that RA-induced inhibition of EMT in tumor cells is mediated through the downregulation of IL-6 and CAFs [92]. CAFs–cancer-cell interactions in the PC organoid model could promote the EMT of cancer cells, which was reversed by the all-trans retinoic acid (ATRA)- mediated inhibition of CAFs [93].
High-mobility group A1 (HMGA1) chromatin regulators are epigenetic agents that could upregulate FGF19 expression, promote fibrotic tissue expansion, and induce desmoplasia [94].
Epithelium-stroma relations are mutual, as epithelium influences stroma, so stroma influences epithelium. CAFs could be responsible for acinar-to-ductal metaplasia (ADM) via the newly discovered laminin5/integrinα4/stat3 axis [95].
The stroma relies on cancer cells and other components of the PDAC constellation, including lymphocytes. Non-canonical CD8+ T-cell subpopulation producing IL-17A (Tc17) accelerates tumor growth via IL-17RA-dependent stroma modification, as IL-17A and TNF synergistically induce differentiation of inflammatory CAFs (iCAFs) by IL-17RA [96].

4. Neuronal PDAC Infiltration and Contribution to Metastasis

Pancreas innervation is both extrinsic and intrinsic. Massive innervation is necessary for regulating endocrine function and controlling the secretion of enzymes in response to nutrient intake. It also controls smooth muscle contractility, splanchnic microcirculation, and epithelial cell absorption [97]. Remarkably, pancreatic nerves undergo severe alterations during carcinogenesis. Intrapancreatic nerves grow in size and number. The proportion of autonomic and sensory fibers is switched, and perineural inflammatory cells, or PC cells, begin to infiltrate neurons [98]. The mechanism by which PDAC induces these changes in the neuronal environment remains unknown. Nerve hypertrophy is significantly distinct in areas with the strongest desmoplasia. It seems that PDAC produces factors necessary for nerve regeneration [99].
In general, the nerve–cancer relationship suggests a tumor-promoting relationship in PDAC. The first studies in this field revealed that neurons can increase the invasiveness and migration of cancer cells toward neuronal targets. Perineural invasion is a critical cause of local recurrence [100,101].
Although neuronal invasion occurs in most solid gastrointestinal tumors, the severity of this process is most pronounced in PDAC [102]. The formation and function of perineural invasion are regulated by molecular (e.g., involving neurotrophins, cytokines, chemokines, and neurotransmitters), metabolic (e.g., serine metabolism), and cellular mechanisms (e.g., Schwann cells, stromal cells, T cells, and macrophages) [103].
Chemokines such as CCL2 [92], or endocrine hormones and neurotrophic factors, including nerve growth factor (NGF) and glial-derived neurotrophic factor (GDNF), are known to promote both in vitro and ex vivo culture models, suggesting that cancer cells surrounding nerves exhibit increased survival [100,101,104].
NGF, BDNF, neurotrophin 3, and neurotrophin 4, along with their receptors, are expressed in different PDAC cell lines, indicating a mutual influence between neurons and PDAC cells [103]. Notably, NTRK1 and NGFR are used as opposing prognostic markers for PDAC patients; NTRK1 expression is associated with a poor prognosis, whereas NGFR is linked to a longer survival rate [105].
PDAC cells rely on serine uptake, which is essential for tumor proliferation. Axons and dorsal root ganglia can secrete serine into the PDAC environment, providing energy support. After serine deprivation, PDAC cells release more NGF, enhancing the movement of axons toward the tumor [106]. Reducing the supply of serine or inhibiting NGF may be an attractive strategy for slowing the progress of neuroinvasion (NI) and tumor growth [107].
The GDNF family comprises GDNF itself, neurturin, artemin, and persephin, which can bind to multiple receptors and perform distinct functions [108]. PDAC patients with NI express higher levels of GDN, indicating the crucial role of GDNF in the neural invasion process [109]. GDNF induces polarization and invadopodia formation in cancer cells, enabling matrix degradation through the production of matrix metalloproteinases, thereby accelerating cell invasion [110]. GDNF targets the RET receptor and GDNF family receptor alpha 1 (GFRA1). The GDNF-RET-GFRA1 axis relies on the activation of KRAS, suggesting that classical oncogenes are essential for neuronal invasion [103].
As mentioned earlier, specific cytokines appear to play a role in neural invasion (NI). C-X3-C motif chemokine ligand 1, produced by neural cells, binds to C-X3-C motif chemokine receptor 1, located on the surface of PDAC cells, to activate integrins and G protein-coupled receptors [111]. Another example of the impact of cytokines on NI is the release of CXCL12 by dorsal root ganglia, which attracts PDAC cells by binding to CXCR4 [112].
Not only do complex pathways of the nervous and immune systems influence NI, but even the seemingly uninvolved factors in tumor progression, such as catecholamines, can also exacerbate it. In preclinical models, norepinephrine induces NI by activating the β2 adrenoreceptor, thereby stimulating nerve growth factor (NGF) secretion, which suggests a correlation between neurophysiological stress and the progression of PDAC [113,114].
Various cells involved in crosstalk in perineural invasion (PNI) infiltrate the pancreatic TME. Schwann cells are typical cells in peripheral nerves. Relations between Schwann cells and PDAC cells are inconsistent and ambiguous. Schwann cells interact with PDAC cells through membrane proteins, such as NCAM1 and myelin-associated glycoprotein. They could also promote the adhesion of PDAC cells to Schwann cells with Mucin 1. Lastly, Schwann cells induce structural alterations in PDAC cells [115,116,117]. Schwann cells communicate with PDAC cells through proteins such as L1 cell adhesion molecule (L1CAM), TGF-β, MMP2, and MMP9 [118,119]. Schwann cells communicate with PDAC cells and recruit macrophages from the bloodstream to the PNI site. Macrophages under the influence of CCL2 secreted by Schwann cells express cathepsin B, which degrades collagen IV, a significant component of perineurium, increasing PNI [120].
PSCs, mentioned above as the most prominent cell type in PDAC stroma, also contribute to PNI [121,122]. PSCs release tenascin C, enhancing the interaction between PDAC and dorsal root ganglia axons. Mutual relations between PDAC cells, neurons, and PSCs seem to be entangled with the SHH pathway [123,124].
TAMs also vaguely affect PDAC–neural cross-talk. Increased TAM invasion in PNI was associated with poor prognosis in PDAC patients [125]. Colony-stimulating factor 1 (CSF1), secreted by PDAC, recruits and activates macrophages, which in turn produce GDNF, thereby increasing PNI [126].
T cells—the main component of the adaptive immune system, which are essential for killing cancer cells—could be severely impaired by the parasympathetic nervous system. The acetylcholine (ACh) secreted by the vagus nerve was elevated in PNI-detected samples from PDAC patients. ACh suppresses CCL5 expression in PDAC cells, leading to insufficient T-cell infiltration. ACh also inhibits interferon-γ expression in CD8+ T cells, favoring Th-cell type 2 over Th-type 1 differentiation. Unfortunately, vagotomy’s impact on PDAC progression is still very concerning, as different studies show opposite results. Vagotomy can lead to prolonged survival in mouse models by increasing CD8+ T cells and elevating the Th1/Th2 ratio [127]. Simultaneously, it can also accelerate tumorigenesis, possibly due to the upregulation of TNF-α [128].
By combining all of these findings, some practical applications can be identified. Firstly, several studies have identified the proteins associated with PNI and their correlation with prognosis. These include membrane and intracellular proteins [129,130,131] (Table 2).
Neuronal invasion occurs in over 80% of PCs, and it can be observed at every stage of PDAC, including the early course. It is detected without lymphatic or hematic metastasis, and even in tumors smaller than 2 cm [98,132]. However, a full perineural invasion where cancer cells infiltrate the outer layer of nerves (epineurium) is not as common as in mouse models [98]. Recent studies have not shown perineural invasion, but have identified coexisting cancer cells surrounding the nerves in KRAS-based PDAC models [133,134].
One of the most important effects of NI and pancreatic hyperinnervation is severe pain linked to extrapancreatic nerve plexus invasion by the tumors growing toward the retroperitoneal space [135]. Severe pain is considered to be an adverse prognostic factor for survival [136,137]. PNI impacts OS prediction. Retrospective studies conducted over 20 years have shown that the postoperative survival of PDAC patients was negatively correlated with nerve infiltration and could be used as a prognostic factor [138,139,140,141,142,143].
NI-free patients have better long-term survival [144]. Not only the presence of NI is a predictive factor, but the depth of nerve infiltration is also. Patients with tumor cells infiltrating beyond the axon of nerves have shorter DFS and OS than those whose cancer cells do not reach the axon [145]. Therefore, NI is one of the conditions that require finding a personalized therapeutic protocol [146].
The most critical role in causing pain is linked to protease-activated receptor 2 (PAR-2)-mediated signaling [147]. A variety of proteases secreted by TME activates PAR-2. Despite the pain, PAR-2 regulates tumor proliferation, differentiation, and invasion [148]. Interestingly, recent studies uncovered an ominous mechanism responsible for suppressing pain response in PDAC and delaying diagnosis. Glial cells, specifically Schwann cells, can be activated in the earliest precursor stages of PC, and they are responsible for anti-inflammatory regulation, which helps alleviate pain [149,150]. IL-6 and CXCL12, secreted by cancer cells, are responsible for glia-mediated pain suppression [149]. Another study indicated a significant impact of the autonomic nervous system, with emphasis on adrenergic pathways, on NGF secretion [114].
As mentioned before, the rich expression of GDNF contributed to the NI in PDAC patients [98]. The level of GDNF and the accumulation of mast cells around intrapancreatic nerves are closely associated with neuropathic pain [135].
Severe pain could simultaneously worsen a patient’s life in numerous ways. It is an adverse prognostic factor for survival, as mentioned above. In addition, it increases the risk of drug abuse and influences the quality of life.
The influence of PNI occurrence on treatment choice is a valid question that many research groups seek to answer. Patients who received neoadjuvant chemotherapy had lower PNI rates, unlike those who did not [151]. Radiotherapy also decreases the PNI rate, as appropriate irradiation reduces GDNF release in the dorsal root ganglia [152]. Also, low-dose irradiation using iodine-125 inhibited PNI and tumor growth, decreasing pain occurrence [153]. The latest studies have shown that telomerase-specific oncolytic viruses can repress PNI, indicating a potential role in PDAC treatment [154].

5. Bacterial and Fungal Influence on PDAC Behavior and Treatment Response

Despite the significant impact of stromal cells on PDAC, it is reported that pancreatic neoplasms harbor a markedly higher number of fungi and bacteria compared to the healthy pancreas [155,156]. Microbiome ablation protects against pre-invasive and invasive PDAC [157]. Meanwhile, the transfer of bacteria from PDAC-hosting mice, but not controls, reverses tumor protection [157].
Microbiota influence the response to checkpoint inhibitor therapy in certain cancers, such as melanoma and lung cancer [158]. Bacterial ablation in PDAC-carrying mice, using antibiotics, reshapes the TME, inducing immune system activation and increasing sensitivity to immunotherapy [157]. It has been demonstrated that long-term survivors (LTS) exhibit higher microbiota diversity compared to short-term survivors (STS). Moreover, 25% of tumor-associated microbiota can be found in the gut, suggesting that changes in the intestinal microbiome can alter the tumor microbiome [155]. However, the specific mechanisms by which bacteria, such as Bifidobacterium, impact PDAC remain unknown. Fecal microbiota transplantation with stool from LTS in healthy control mice mirrors the recruitment of immune cells to the TME. It is not observed when stool is transplanted from STS recruitment to the TME [155]. Gut microbiome composition can enhance the effectiveness of cancer immunotherapy by modulating the immune system. For example, immunosuppressive populations treated with stool transplants from LTS patients were modulated by decreasing tumor infiltration by Tregs [159]. The elevated presence of three bacterial taxa, Saccharopolyspora, Pseudoxanthomonas, and Streptomyces, is noted in LTS patients [155]. Saccharopolyspora rectivirgula has been shown to play a role in inflammatory lung diseases, such as hypersensitivity pneumonitis [160]. The presence of Saccharopolyspora spp. could be responsible for generating a pro-inflammatory environment. However, its role in PDAC still needs to be explored [155].
One of the investigated mechanisms was the activation of aryl hydrocarbon receptor (AhR) by tryptophan metabolites produced by bacteria. TAMs, which express high levels of the AhR, develop inflammatory types of cancer with worse survival outcomes. AhR activation promotes the expression of IL-10 in TAMs and inhibits IFNγ expression in CD8+ T cells. Loss of the gut microbiome or exclusion of tryptophan from the diet mimics the deletion of AhR [161].
Tryptophan metabolite-indole-3-acetic acid (3-IAA) is a crucial amplifier of the response to chemotherapy in PDAC. Data from observing a small cohort of patients reveal a robust correlation between 3-IAA serum concentrations during chemotherapy and OS. Clinical trials aiming to raise 3-IAA serum concentration during chemotherapy ultimately increase survival in PDAC (NCT02077881) [162]. The easiest way to achieve this goal is to increase tryptophan dietary intake; however, it was effective only in the group of patients responding to chemotherapy, which is correlated with their microbiota constellation. Many taxonomically distinct bacterial species can produce 3-IAA, and further studies are needed to assess a narrow selection of the appropriate microbiota [163].
Recently, Gammaproteobacterium (GP), in addition to the rest of the TME-residing bacteria, has been found in human PDAC. GP can metabolize gemcitabine into its inactive form, suggesting a role for GP in gemcitabine resistance [164].

6. Repressing Immune Response via Microenvironmental Components

PDAC is known to be unresponsive to checkpoint inhibitors (ICIs) like atezolizumab or durvalumab, which bind to PD-L1, or pembrolizumab and nivolumab, which bind to PD-1, despite their high efficacy in other malignancies, such as non-small cell lung cancer (NSCLC), melanoma, or renal cell cancer (RCC) [165]. Although the molecular mechanism responsible for immunity to ICPis is unknown, it is probably related to the TME. This challenge drives researchers to identify the novel agents that can enhance the immune system’s ability to recognize and eliminate cancer cells [166].
A critical factor necessary to elicit efficacy in PDAC immunotherapy is high microsatellite instability (MSI-high) [167]. Unfortunately, only circa 1% of patients present an MSI-high phenotype [168], even though the FDA approved pembrolizumab to treat MSI-high and mismatch repair deficient patients [121]. Another experimental therapy proposed for tumor-bearing mice combines radiotherapy and PD-L1 immunotherapy. Radiotherapy upregulates PD-L1 in tumor cells, enhancing the response to PD-L1 blockade. A recent study revealed an innovative therapy strategy: combining radiotherapy with the administration of pembrolizumab and trametinib, an inhibitor of MEK1/2 [122,169].
A remarkably simple yet crucial obstacle that renders PDAC resistant to immunotherapy is the poor infiltration of immune cells into the PDAC TME [1,170]. Nevertheless, the indigent immunogenicity of PDAC may not be as indigent as previously observed. Recent studies have shown that the PDAC immune constellation may be much more sophisticated than previously thought. It seems that immune cells could comprise up to 50% of the PDAC cellular component, but only a few are anti-tumor effector cells [168,171].
CD3+ T-cells are a significant immune cell type in PDAC, predominantly occurring in the stroma of the cancer rather than within the cancer cell nest. They coexist with dendritic cells, Treg cells, B cells, and high endothelial venules within tertiary lymphoid structures. Their primary source is local proliferation instead of migration [172]. Additionally, CD4+ T cells are frequently present in the PDAC environment, whereas CD8+ T cells are less common, with accumulation varying between 7% and 30%. Moreover, these cells are functionally deficient, as they express various co-inhibitory molecules [172,173].
Among CD4+ T-cells, only the Th1 subset promotes anti-tumor response, as Th2 promotes tumor development. Notably, Th2 cells constitute a significant population of CD4+ T cells within PDAC tumors, and their concentration is higher than that of Th1 cells and FoxP3+ Treg cells [174]. These findings indicate one of the mechanisms responsible for immunotolerance and identify a potential target for future therapeutic strategies. For example, chemotherapy, especially gemcitabine, can deplete Treg cells, thereby enhancing cytotoxic and helper T cell responses and restoring the anti-tumor effects of Cd40 agonists and ICIs [175,176,177,178].
Bringing lymphocytes to the table, we cannot overlook Chimer Antigen Receptor T-cells (CAR-T) cells, one of the most extensively studied technologies in modern medicine. Various artificial gene design strategies targeting PDAC stroma are being explored, despite some difficulties. PDAC is a solid tumor, unlike hematological malignancies, where CAR-T therapy was primarily introduced, and it demands extra research to work correctly. The FDA’s approval of Amtagvi, dedicated to treating unresectable melanoma, brings hope for the future of tumor-infiltrating cell therapy [168,171]. Over fifty clinical trials involving CAR-T therapy in PDAC are currently being conducted, primarily targeting CEA, PSCA, and the endogenous MSLN epitope. Only one trial made it to phase 3, yet it was withdrawn (NCT04037241).
Another approach in PDAC treatment is the use of tumor vaccines. They may contain one or several tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs). TAAs are overexpressed in proteins of cancer cells, which may also be present in some non-cancerous tissues, making them non-specific. TSAs, on the other hand, are expressed exclusively on tumor cells [179]. TSAs’ vaccines are highly specific, immunogenic, and avoid self-antigen-induced T cell tolerance [180]. One of the most promising approaches is the pancreatic cancer cell line-based vaccine, known as GVAX. GVAX contains irradiated allogeneic pancreatic cells that cannot grow and exhibit a wide range of PDAC antigens. So far, results have demonstrated that GVAX is safe, well-tolerated, and induces antigen-specific T-cell responses; nevertheless, it still does not show superiority to standard chemotherapy [179].
Intense TAM infiltration, especially secreting IL-10 M2 macrophages, is correlated with poor survival; thus, TAMs may be a promising therapeutic target for immunotherapy [136]. In PDAC, macrophages can be recruited from monocyte and tissue-resident sources [137]. The proliferation of TAMs could be driven by colony-stimulating factor-1 (CSF-1) produced by CAFs. CSF1 influences TAMs to express higher levels of p21, which is responsible for more inflammatory and immunosuppressive phenotypes [181]. Macrophages can mediate tumor immunosuppression by directly interacting with CD8+ T cells and, in a more indirect manner, by secreting immunomodulatory factors that alter the TME [182,183]. As such, preclinical and clinical studies have focused on TAM targeting, such as CD40 agonist treatment. Despite promising results in preclinical models, clinical success has yet to be demonstrated, suggesting more complex relationships in vivo [144,182,184].
CD47—known as the “don’t eat me” signal, which binds to a signal regulatory protein α (SIRPα)—is located on phagocytes and suppresses phagocytosis [144]. Preclinical studies have demonstrated that inhibiting CD47-SIRPα in vivo enhances the efficiency of PDAC clearance by macrophages, a phenomenon that correlates with the surface density of the CD47 receptor [145].
In PDAC, CD68+ tumor-infiltrating macrophages play a crucial role in expressing CD47, and their number correlates with the clinical features of PDAC patients and OS. CD163+ M2 macrophages are second to CD68+ macrophages investigated in estimating PDAC survival. They do not correlate with CD47 expression; however, high CD163+ infiltration is associated with tumor diameter and a high pT-stage. Anti-CD47 treatment resulted in changes in the TME, characterized by an increase in pro-inflammatory macrophages, a decrease in anti-inflammatory macrophages, and an increase in the number of intratumoral lymphoid cells [166].
The combined targeting of both CD47 and PD-L1 results in a synergistic inhibitory effect in MPC-83 cells, but not in Panc02 transgenic mice, likely due to the activation of different immune-activating genes [166].
The primary difference between mouse models and human PDAC is the T-lymphocyte to B-lymphocyte ratio, which is higher in human tumors. Despite finding that PDA-bearing mice had cancer cell-specific CD8+ cells, the mice, like human patients, do not respond to ICPIs. The control of PDAC growth via ICPis was achieved by depleting CAFs expressing the fibroblast activation protein (FAP). Three findings suggested that CXCL12 was responsible for the overexpression of FAP+ CAFs. Inhibition of CXCL12 resulted in T-cell accumulation and acted synergistically with PD-L1, thereby diminishing the growth of cancer cells [185].
To date, research on the PDAC immune milieu has primarily focused on TAMs, CAFs, and lymphocytes. Interestingly, currently, investigations increasingly indicate the role of neutrophils. Tumor-associated neutrophils (TANs) were found to have an unexpected positive correlation with CD8+ T-cells, which is surprising because it was previously thought that TANs hinder lymphocyte infiltration into the PDAC environment [173,186,187]. In Table 3, we summarize the TME components.

7. Clinical Trials

Regarding all various mechanisms influencing the relations between cancer cells and TME cells in PDAC, many clinical trials are taking place to find a partial solution to the poor outcomes of standard treatment regimens. We collect some of them in Table 4.

8. Conclusions

In conclusion, this paper summarizes the impact of the PDAC microenvironment on disease etiology, prognosis, and possible therapeutic options. The most crucial TME constellation components are immune cells, especially macrophages and lymphocytes, fibroblasts, bacterial, and fungal microflora, and neuronal cells. Depending on the particular phenotype of these cells, the composition of the microenvironment, and the cell ratio, patients can experience different disease outcomes and varying vulnerability to treatment approaches. Studies aiming at TME appear to be a significant future research path, as the microenvironment comprises approximately 90% [209,210] of the tumor mass. Unfortunately, many ongoing studies fail when it comes to applying in vitro or animal-based trials in human patients. Nevertheless, we hope that eventually we will cross the Rubicon, surpassing the infamous 11% five-year survival rate.

Author Contributions

Conceptualization, M.F., A.B.-K. and A.D.; methodology, F.G. and W.Z.; investigation, F.G. and W.Z.; resources, M.F.; data curation, F.G., W.Z. and M.F.; writing—original draft preparation, F.G., W.Z. and M.F.; writing—review and editing, M.F., A.B.-K. and A.D.; supervision, A.C., K.S., R.K., A.B.-K. and A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
TregRegulatory T lymphocytes
CD4Cluster of Differentiation 4
CD25Cluster of Differentiation 25
FOXP3Forkhead box P3
TGFβTransforming Growth Factor Beta
IL-10Interleukin 10
IL-1Interleukin 1
MDSCMyeloid-Derived Suppressor Cells
M-MDSCMonocyte-like Myeloid-Derived Suppressor Cells
PMN-MDSCPolymorphonuclear Myeloid-Derived Suppressor Cells
CD8+Cytotoxic T lymphocyte
NKNatural Killer cells
PDACPancreatic Ductal Adenocarcinoma
GM-CSFGranulocyte-Macrophage Colony-Stimulating Factor
IL-13Interleukin 13
HIFsHypoxia-Inducible Factors
TAMsTumor-Associated Macrophages
TNFαTumor Necrosis Factor Alpha
HIF2Hypoxia-Inducible Factor 2
CAFsCancer-Associated Fibroblasts
myCAFMyofibroblastic Cancer-Associated Fibroblasts
iCAFinflammatory Cancer-Associated Fibroblasts
apCAFAntigen-presenting Cancer-Associated Fibroblasts
ECMExtracellular Matrix
CK-19Cytokeratin-19
SOX9SRY-Box Transcription Factor 9
FOLFIRINOXFluorouracil, Leucovorin, Irinotecan, Oxaliplatin
GnPGemcitabine and Nab-Paclitaxel
R-PDACResectable Pancreatic Ductal Adenocarcinoma
BR-PDACBorderline Resectable Pancreatic Ductal Adenocarcinoma
MSI-highMicrosatellite Instability-high
PD-L1Programmed Death-Ligand 1
MEK1/2Mitogen-Activated Protein Kinase ½
CAR-TChimeric Antigen Receptor T-cells
CEACarcinoembryonic Antigen
PSCAProstate Stem Cell Antigen
MSLNMesothelin
ICIsImmune Checkpoint Inhibitors
CXCL12C-X-C Motif Chemokine Ligand 12
CCL2C-C motif chemokine 2
NGFNerve Growth Factor
GDNFGlial Cell-Derived Neurotrophic Factor
HGFHepatocyte Growth Factor
STAT3Signal Transducer and Activator of Transcription 3
β2Beta-2 Adrenergic Receptor
TMETumor Microenvironment
OSOverall Survival
DFSDisease-Free Survival
PAR-2Protease-Activated Receptor 2
ICPsImmune Checkpoints
TANsTumor-Associated Neutrophils
FAPFibroblast Activation Protein
ERKExtracellular Signal-Regulated Kinase
TILsTumor-Infiltrating Lymphocytes
PCPancreatic Cancer
CK-19Cytokeratin-19
SEERSurveillance, Epidemiology, and End Results Programme
PSCPancreatic Stellate Cells
PDGFPlatelet-derived growth factor
FGFfibroblast growth factor
CTGFConnective tissue growth factor
αSMAα-smooth muscle actin
IGF1Insulin-like growth factor 1
VEGFVascular endothelial growth factor
FSP1Fibroblasts-specific protein 1
ETMEndothelial-to-mesenchymal transition
SDF-1Stromal-derived factor-1
ROSReactive Oxygen Species
NOXsNADPH oxidases
EREndoplasmatic Reticulum
siRNAsSmall interfering RNAs
SMA+Alpha-smooth muscle actin
SHH, Sonic Hedgehog Homolog
PtchPatched transmembrane protein
SMOSmoothened transmembrane protein
GM-CSFGranulocyte-macrophage colony-stimulating factor
TIMPMatrix-metalloproteinases inhibitors
PDXPatient-dervied xenografts
RARetinoid acid
ATRAAll-trans-retinoid-acid
HMGA1High mobility group A1
ADMAcinar-to-ductal metaplasia
GFRA1GDNF family receptor alpha 1
PNIPerineural invasion
L1CAML1 cell adhesion molecule
CSF1Colony stimulating factor 1
AChAcetylcholine
AChRAcetylcholine receptor
PAR-2Protease-activated receptor 2
LTSLong-time survivors
STSShort-time survivors
3-IAAIndole-3-acetic acid
GPGammaproteobacterium
NSCLCNon-small cell lung cancer
RCCRenal cell cancer
TSAsTumor-specific antigens
GVAXPancreatic cancer cell line-based vaccine
BIRC5Baculoviral IAP Repeat Containing 5
CCND1Cyclin D1
CDH1Cadherin 1
CD74Cluster of Differentiation 74
COL6A3Collagen type VI alpha three chain
CTHRC1Collagen triple helix repeat containing 1
IL-13RA2Interleukin 13 receptor subunit alpha 2
LRP1Low-density lipoprotein receptor-related protein 1
MAP1LC3A/BMicrotubule-associated protein one light chain three alpha/beta
MDKMidkine
MYBL2MYB proto-oncogene like 2
MYCMYC proto-oncogene
PODXLPodocalyxin-like protein
PTNPleiotrophin
SDC2Syndecan 2
SDC3Syndecan 3
SNCGSynuclein gamma
TIMP2Tissue inhibitor of metalloproteinases 2
VGFVGF nerve growth factor
WASLWASP-like actin nucleation-promoting factor
FASFAS cell surface death receptor
POP1Processing of precursor 1, ribonuclease P/MRP subunit
CADM4Cell adhesion molecule 4
HPGD15-hydroxyprostaglandin dehydrogenase
CD11bCluster of differentiation 11b
VEGFR1Vascular endothelial growth factor receptor 1
VEGFR2Vascular endothelial growth factor receptor 2
VEGFR3Vascular endothelial growth factor receptor 3
CSF-1RColony-stimulating-factor receptor 1
TLR3Toll-like receptor 3
PARP1Poly(ADP-ribose) polymerase 1
PARP2Poly(ADP-ribose) polymerase 2
LRRC15Leucine-rich repeat containing 15 protein
IGF1RInsulin-like growth factor 1 receptor
HAHyaluronic acid
LIFLeukemia inhibitory factor
FOLFIRINOX5-Fluorouracil, Irinotecan, Oxaliplatin, Leucovorin

References

  1. Ho, W.J.; Jaffee, E.M.; Zheng, L. The tumour microenvironment in pancreatic cancer—Clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 2020, 17, 527–540. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, L.; Xie, D.; Wei, D. Pancreatic Acinar-to-Ductal Metaplasia and Pancreatic Cancer. Methods Mol. Biol. 2019, 1882, 299–308. [Google Scholar] [CrossRef]
  3. Grant, T.J.; Hua, K.; Singh, A. Molecular Pathogenesis of Pancreatic Cancer. Prog. Mol. Biol. Transl. Sci. 2016, 144, 241–275. [Google Scholar] [CrossRef]
  4. McGuigan, A.; Kelly, P.; Turkington, R.C.; Jones, C.; Coleman, H.G.; McCain, R.S. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J. Gastroenterol. 2018, 24, 4846–4861. [Google Scholar] [CrossRef] [PubMed]
  5. Sherman, M.H.; Beatty, G.L. Tumor Microenvironment in Pancreatic Cancer Pathogenesis and Therapeutic Resistance. Annu. Rev. Pathol. 2023, 18, 123–148. [Google Scholar] [CrossRef]
  6. National Cancer Institute. SEER Cancer Statistics; National Cancer Institute: Bethesda, MD, USA, 2025.
  7. Polish National Cancer Registry. Polish National Cancer Registry Statistics; Polish National Cancer Registry: Warszawa, Poland, 2025. [Google Scholar]
  8. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  9. Qin, Q.; Yu, R.; Eriksson, J.E.; Tsai, H.-I.; Zhu, H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett. 2024, 591, 216859. [Google Scholar] [CrossRef] [PubMed]
  10. Kwaśniewska, D.; Fudalej, M.; Nurzyński, P.; Badowska-Kozakiewicz, A.; Czerw, A.; Cipora, E.; Sygit, K.; Bandurska, E.; Deptała, A. How A Patient with Resectable or Borderline Resectable Pancreatic Cancer should Be Treated-A Comprehensive Review. Cancers 2023, 15, 4275. [Google Scholar] [CrossRef]
  11. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef]
  12. Aung, K.L.; Fischer, S.E.; Denroche, R.E.; Jang, G.H.; Dodd, A.; Creighton, S.; Southwood, B.; Liang, S.B.; Chadwick, D.; Zhang, A.; et al. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin. Cancer Res. 2018, 24, 1344–1354. [Google Scholar] [CrossRef]
  13. Veenstra, V.L.; Garcia-Garijo, A.; van Laarhoven, H.W.; Bijlsma, M.F. Extracellular Influences: Molecular Subclasses and the Microenvironment in Pancreatic Cancer. Cancers 2018, 10, 34. [Google Scholar] [CrossRef] [PubMed]
  14. De Wever, O.; Mareel, M. Role of tissue stroma in cancer cell invasion. J. Pathol. 2003, 200, 429–447. [Google Scholar] [CrossRef] [PubMed]
  15. Thomas, D.; Radhakrishnan, P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol. Cancer 2019, 18, 14. [Google Scholar] [CrossRef]
  16. Schnittert, J.; Bansal, R.; Prakash, J. Targeting Pancreatic Stellate Cells in Cancer. Trends Cancer 2019, 5, 128–142. [Google Scholar] [CrossRef] [PubMed]
  17. Pandol, S.; Edderkaoui, M.; Gukovsky, I.; Lugea, A.; Gukovskaya, A. Desmoplasia of pancreatic ductal adenocarcinoma. Clin. Gastroenterol. Hepatol. 2009, 7, S44–S47. [Google Scholar] [CrossRef]
  18. Jang, J.E.; Hajdu, C.H.; Liot, C.; Miller, G.; Dustin, M.L.; Bar-Sagi, D. Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer. Cell Rep. 2017, 20, 558–571. [Google Scholar] [CrossRef]
  19. Sperb, N.; Tsesmelis, M.; Wirth, T. Crosstalk between Tumor and Stromal Cells in Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020, 21, 5486. [Google Scholar] [CrossRef]
  20. Dougan, S.K. The Pancreatic Cancer Microenvironment. Cancer J. 2017, 23, 321–325. [Google Scholar] [CrossRef]
  21. Apte, M.V.; Haber, P.S.; Darby, S.J.; Rodgers, S.C.; McCaughan, G.W.; Korsten, M.A.; Pirola, R.C.; Wilson, J.S. Pancreatic stellate cells are activated by proinflammatory cytokines: Implications for pancreatic fibrogenesis. Gut 1999, 44, 534–541. [Google Scholar] [CrossRef]
  22. Jin, G.; Hong, W.; Guo, Y.; Bai, Y.; Chen, B. Molecular Mechanism of Pancreatic Stellate Cells Activation in Chronic Pancreatitis and Pancreatic Cancer. J. Cancer 2020, 11, 1505–1515. [Google Scholar] [CrossRef]
  23. Schnittert, J.; Bansal, R.; Mardhian, D.F.; van Baarlen, J.; Östman, A.; Prakash, J. Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer. FASEB J. 2019, 33, 6609–6621. [Google Scholar] [CrossRef] [PubMed]
  24. Apte, M.V.; Park, S.; Phillips, P.A.; Santucci, N.; Goldstein, D.; Kumar, R.K.; Ramm, G.A.; Buchler, M.; Friess, H.; McCarroll, J.A.; et al. Desmoplastic reaction in pancreatic cancer: Role of pancreatic stellate cells. Pancreas 2004, 29, 179–187. [Google Scholar] [CrossRef]
  25. Apte, M.V.; Wilson, J.S.; Lugea, A.; Pandol, S.J. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology 2013, 144, 1210–1219. [Google Scholar] [CrossRef]
  26. Petruzzelli, M.; Wagner, E.F. Mechanisms of metabolic dysfunction in cancer-associated cachexia. Genes Dev. 2016, 30, 489–501. [Google Scholar] [CrossRef] [PubMed]
  27. Mantoni, T.S.; Lunardi, S.; Al-Assar, O.; Masamune, A.; Brunner, T.B. Pancreatic stellate cells radioprotect pancreatic cancer cells through β1-integrin signaling. Cancer Res. 2011, 71, 3453–3458. [Google Scholar] [CrossRef]
  28. Cabrera, M.C.; Tilahun, E.; Nakles, R.; Diaz-Cruz, E.S.; Charabaty, A.; Suy, S.; Jackson, P.; Ley, L.; Slack, R.; Jha, R.; et al. Human Pancreatic Cancer-Associated Stellate Cells Remain Activated after in vivo Chemoradiation. Front. Oncol. 2014, 4, 102. [Google Scholar] [CrossRef] [PubMed]
  29. Mękal, D.; Sobocki, J.; Badowska-Kozakiewicz, A.; Sygit, K.; Cipora, E.; Bandurska, E.; Czerw, A.; Deptała, A. Evaluation of Nutritional Status and the Impact of Nutritional Treatment in Patients with Pancreatic Cancer. Cancers 2023, 15, 3816. [Google Scholar] [CrossRef]
  30. 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]
  31. 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]
  32. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar] [CrossRef]
  33. Anderberg, C.; Pietras, K. On the origin of cancer-associated fibroblasts. Cell Cycle 2009, 8, 1461–1462. [Google Scholar] [CrossRef]
  34. 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]
  35. Iwano, M.; Plieth, D.; Danoff, T.M.; Xue, C.; Okada, H.; Neilson, E.G. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Investig. 2002, 110, 341–350. [Google Scholar] [CrossRef] [PubMed]
  36. Rimal, R.; Desai, P.; Daware, R.; Hosseinnejad, A.; Prakash, J.; Lammers, T.; Singh, S. Cancer-associated fibroblasts: Origin, function, imaging, and therapeutic targeting. Adv. Drug Deliv. Rev. 2022, 189, 114504. [Google Scholar] [CrossRef] [PubMed]
  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] [PubMed]
  38. Flaberg, E.; Markasz, L.; Petranyi, G.; Stuber, G.; Dicso, F.; Alchihabi, N.; Oláh, È.; Csízy, I.; Józsa, T.; Andrén, O.; et al. High-throughput live-cell imaging reveals differential inhibition of tumor cell proliferation by human fibroblasts. Int. J. Cancer 2011, 128, 2793–2802. [Google Scholar] [CrossRef]
  39. 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.e1622. [Google Scholar] [CrossRef]
  40. Guo, J.; Zeng, H.; Chen, Y. Emerging Nano Drug Delivery Systems Targeting Cancer-Associated Fibroblasts for Improved Antitumor Effect and Tumor Drug Penetration. Mol. Pharm. 2020, 17, 1028–1048. [Google Scholar] [CrossRef]
  41. Sharbeen, G.; McCarroll, J.A.; Akerman, A.; Kopecky, C.; Youkhana, J.; Kokkinos, J.; Holst, J.; Boyer, C.; Erkan, M.; Goldstein, D.; et al. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma Determine Response to SLC7A11 Inhibition. Cancer Res. 2021, 81, 3461–3479. [Google Scholar] [CrossRef]
  42. Sato, H.; Shiiya, A.; Kimata, M.; Maebara, K.; Tamba, M.; Sakakura, Y.; Makino, N.; Sugiyama, F.; Yagami, K.; Moriguchi, T.; et al. Redox imbalance in cystine/glutamate transporter-deficient mice. J. Biol. Chem. 2005, 280, 37423–37429. [Google Scholar] [CrossRef]
  43. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef]
  44. Cairns, R.A.; Harris, I.S.; Mak, T.W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 2011, 11, 85–95. [Google Scholar] [CrossRef]
  45. Sabharwal, S.S.; Schumacker, P.T. Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles’ heel? Nat. Rev. Cancer 2014, 14, 709–721. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, K.; Jiang, J.; Lei, Y.; Zhou, S.; Wei, Y.; Huang, C. Targeting Metabolic-Redox Circuits for Cancer Therapy. Trends Biochem. Sci. 2019, 44, 401–414. [Google Scholar] [CrossRef]
  47. Weber, C.K.; Liptay, S.; Wirth, T.; Adler, G.; Schmid, R.M. Suppression of NF-kappaB activity by sulfasalazine is mediated by direct inhibition of IkappaB kinases alpha and beta. Gastroenterology 2000, 119, 1209–1218. [Google Scholar] [CrossRef] [PubMed]
  48. Lo, M.; Ling, V.; Low, C.; Wang, Y.Z.; Gout, P.W. Potential use of the anti-inflammatory drug, sulfasalazine, for targeted therapy of pancreatic cancer. Curr. Oncol. 2010, 17, 9–16. [Google Scholar] [CrossRef]
  49. Chen, Y.; Yang, S.; Tavormina, J.; Tampe, D.; Zeisberg, M.; Wang, H.; Mahadevan, K.K.; Wu, C.J.; Sugimoto, H.; Chang, C.C.; et al. Oncogenic collagen I homotrimers from cancer cells bind to α3β1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell 2022, 40, 818–834.e819. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, X.; He, X.; Zhang, Y.; Hosaka, K.; Andersson, P.; Wu, J.; Wu, J.; Jing, X.; Du, Q.; Hui, X.; et al. Inflammatory cell-derived CXCL3 promotes pancreatic cancer metastasis through a novel myofibroblast-hijacked cancer escape mechanism. Gut 2022, 71, 129–147. [Google Scholar] [CrossRef]
  51. Maurer, C.; Holmstrom, S.R.; He, J.; Laise, P.; Su, T.; Ahmed, A.; Hibshoosh, H.; Chabot, J.A.; Oberstein, P.E.; Sepulveda, A.R.; et al. Experimental microdissection enables functional harmonisation of pancreatic cancer subtypes. Gut 2019, 68, 1034–1043. [Google Scholar] [CrossRef]
  52. 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.e546. [Google Scholar] [CrossRef]
  53. Thayer, S.P.; di Magliano, M.P.; Heiser, P.W.; Nielsen, C.M.; Roberts, D.J.; Lauwers, G.Y.; Qi, Y.P.; Gysin, S.; Fernández-del Castillo, C.; Yajnik, V.; et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003, 425, 851–856. [Google Scholar] [CrossRef] [PubMed]
  54. Kayed, H.; Kleeff, J.; Osman, T.; Keleg, S.; Büchler, M.W.; Friess, H. Hedgehog signaling in the normal and diseased pancreas. Pancreas 2006, 32, 119–129. [Google Scholar] [CrossRef] [PubMed]
  55. Porter, J.A.; Young, K.E.; Beachy, P.A. Cholesterol modification of hedgehog signaling proteins in animal development. Science 1996, 274, 255–259. [Google Scholar] [CrossRef] [PubMed]
  56. Ishii, G.; Sangai, T.; Ito, T.; Hasebe, T.; Endoh, Y.; Sasaki, H.; Harigaya, K.; Ochiai, A. In vivo and in vitro characterization of human fibroblasts recruited selectively into human cancer stroma. Int. J. Cancer 2005, 117, 212–220. [Google Scholar] [CrossRef]
  57. Yoo, Y.A.; Kang, M.H.; Kim, J.S.; Oh, S.C. Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Carcinogenesis 2008, 29, 480–490. [Google Scholar] [CrossRef]
  58. Olivares, O.; Mayers, J.R.; Gouirand, V.; Torrence, M.E.; Gicquel, T.; Borge, L.; Lac, S.; Roques, J.; Lavaut, M.N.; Berthezène, P.; et al. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat. Commun. 2017, 8, 16031. [Google Scholar] [CrossRef]
  59. Ohlund, D.; Lundin, C.; Ardnor, B.; Oman, M.; Naredi, P.; Sund, M. Type IV collagen is a tumour stroma-derived biomarker for pancreas cancer. Br. J. Cancer 2009, 101, 91–97. [Google Scholar] [CrossRef]
  60. Knapinska, A.M.; Estrada, C.A.; Fields, G.B. The Roles of Matrix Metalloproteinases in Pancreatic Cancer. Prog. Mol. Biol. Transl. Sci. 2017, 148, 339–354. [Google Scholar] [CrossRef]
  61. Ottaviano, A.J.; Sun, L.; Ananthanarayanan, V.; Munshi, H.G. Extracellular matrix-mediated membrane-type 1 matrix metalloproteinase expression in pancreatic ductal cells is regulated by transforming growth factor-beta1. Cancer Res. 2006, 66, 7032–7040. [Google Scholar] [CrossRef]
  62. Whatcott, C.J.; Ng, S.; Barrett, M.T.; Hostetter, G.; Von Hoff, D.D.; Han, H. Inhibition of ROCK1 kinase modulates both tumor cells and stromal fibroblasts in pancreatic cancer. PLoS ONE 2017, 12, e0183871. [Google Scholar] [CrossRef]
  63. Whatcott, C.J.; Diep, C.H.; Jiang, P.; Watanabe, A.; LoBello, J.; Sima, C.; Hostetter, G.; Shepard, H.M.; Von Hoff, D.D.; Han, H. Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clin. Cancer Res. 2015, 21, 3561–3568. [Google Scholar] [CrossRef] [PubMed]
  64. Grage-Griebenow, E.; Schäfer, H.; Sebens, S. The fatal alliance of cancer and T cells: How pancreatic tumor cells gather immunosuppressive T cells. Oncoimmunology 2014, 3, e29382. [Google Scholar] [CrossRef] [PubMed]
  65. Padoan, A.; Plebani, M.; Basso, D. Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int. J. Mol. Sci. 2019, 20, 676. [Google Scholar] [CrossRef] [PubMed]
  66. Brouwer, T.; Ijsselsteijn, M.; Oosting, J.; Ruano, D.; van der Ploeg, M.; Dijk, F.; Bonsing, B.; Fariña, A.; Morreau, H.; Vahrmeijer, A.; et al. A Paradoxical Role for Regulatory T Cells in the Tumor Microenvironment of Pancreatic Cancer. Cancers 2022, 14, 3862. [Google Scholar] [CrossRef]
  67. Karakhanova, S.; Link, J.; Heinrich, M.; Shevchenko, I.; Yang, Y.; Hassenpflug, M.; Bunge, H.; von Ahn, K.; Brecht, R.; Mathes, A.; et al. Characterization of myeloid leukocytes and soluble mediators in pancreatic cancer: Importance of myeloid-derived suppressor cells. Oncoimmunology 2015, 4, e998519. [Google Scholar] [CrossRef]
  68. Pergamo, M.; Miller, G. Myeloid-derived suppressor cells and their role in pancreatic cancer. Cancer Gene Ther. 2017, 24, 100–105. [Google Scholar] [CrossRef]
  69. Metzger, P.; Kirchleitner, S.V.; Kluge, M.; Koenig, L.M.; Hörth, C.; Rambuscheck, C.A.; Böhmer, D.; Ahlfeld, J.; Kobold, S.; Friedel, C.C.; et al. Immunostimulatory RNA leads to functional reprogramming of myeloid-derived suppressor cells in pancreatic cancer. J. Immunother. Cancer 2019, 7, 288. [Google Scholar] [CrossRef]
  70. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef]
  71. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef]
  72. Vincent, J.; Mignot, G.; Chalmin, F.; Ladoire, S.; Bruchard, M.; Chevriaux, A.; Martin, F.; Apetoh, L.; Rébé, C.; Ghiringhelli, F. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010, 70, 3052–3061. [Google Scholar] [CrossRef]
  73. Liu, R.; Li, J.; Liu, L.; Wang, W.; Jia, J. Tumor-associated macrophages (TAMs): Constructing an immunosuppressive microenvironment bridge for pancreatic ductal adenocarcinoma (PDAC). Cancer Pathog. Ther. 2024, 2, E01–E50. [Google Scholar] [CrossRef]
  74. Chen, S.J.; Lian, G.D.; Li, J.J.; Zhang, Q.B.; Zeng, L.J.; Yang, K.G.; Huang, C.M.; Li, Y.Q.; Chen, Y.T.; Huang, K.H. Tumor-driven like macrophages induced by conditioned media from pancreatic ductal adenocarcinoma promote tumor metastasis via secreting IL-8. Cancer Med. 2018, 7, 5679–5690. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, M.; Pan, X.; Fujiwara, K.; Jurcak, N.; Muth, S.; Zhou, J.; Xiao, Q.; Li, A.; Che, X.; Li, Z.; et al. Pancreatic cancer cells render tumor-associated macrophages metabolically reprogrammed by a GARP and DNA methylation-mediated mechanism. Signal Transduct. Target. Ther. 2021, 6, 366. [Google Scholar] [CrossRef] [PubMed]
  76. He, Z.; Wang, J.; Zhu, C.; Xu, J.; Chen, P.; Jiang, X.; Chen, Y.; Jiang, J.; Sun, C. Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Lett. 2022, 548, 215751. [Google Scholar] [CrossRef]
  77. Kratochvill, F.; Neale, G.; Haverkamp, J.M.; Van de Velde, L.A.; Smith, A.M.; Kawauchi, D.; McEvoy, J.; Roussel, M.F.; Dyer, M.A.; Qualls, J.E.; et al. TNF Counterbalances the Emergence of M2 Tumor Macrophages. Cell Rep. 2015, 12, 1902–1914. [Google Scholar] [CrossRef]
  78. Xiong, C.; Zhu, Y.; Xue, M.; Jiang, Y.; Zhong, Y.; Jiang, L.; Shi, M.; Chen, H. Tumor-associated macrophages promote pancreatic ductal adenocarcinoma progression by inducing epithelial-to-mesenchymal transition. Aging 2021, 13, 3386–3404. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Di Maggio, F.; Arumugam, P.; Delvecchio, F.R.; Batista, S.; Lechertier, T.; Hodivala-Dilke, K.; Kocher, H.M. Pancreatic stellate cells regulate blood vessel density in the stroma of pancreatic ductal adenocarcinoma. Pancreatology 2016, 16, 995–1004. [Google Scholar] [CrossRef]
  81. Fernández-Cortés, M.; Delgado-Bellido, D.; Oliver, F.J. Vasculogenic Mimicry: Become an Endothelial Cell “But Not So Much”. Front. Oncol. 2019, 9, 803. [Google Scholar] [CrossRef]
  82. Benjakul, N.; Prakobphol, N.; Tangshewinsirikul, C.; Dulyaphat, W.; Svasti, J.; Charngkaew, K.; Kangsamaksin, T. Notch signaling regulates vasculogenic mimicry and promotes cell morphogenesis and the epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma. PLoS ONE 2022, 17, e0279001. [Google Scholar] [CrossRef]
  83. Hawighorst, T.; Velasco, P.; Streit, M.; Hong, Y.K.; Kyriakides, T.R.; Brown, L.F.; Bornstein, P.; Detmar, M. Thrombospondin-2 plays a protective role in multistep carcinogenesis: A novel host anti-tumor defense mechanism. EMBO J. 2001, 20, 2631–2640. [Google Scholar] [CrossRef] [PubMed]
  84. Bausch, D.; Fritz, S.; Bolm, L.; Wellner, U.F.; Fernandez-Del-Castillo, C.; Warshaw, A.L.; Thayer, S.P.; Liss, A.S. Hedgehog signaling promotes angiogenesis directly and indirectly in pancreatic cancer. Angiogenesis 2020, 23, 479–492. [Google Scholar] [CrossRef] [PubMed]
  85. Folkman, J. Tumor angiogenesis: A possible control point in tumor growth. Ann. Intern. Med. 1975, 82, 96–100. [Google Scholar] [CrossRef] [PubMed]
  86. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef]
  87. 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]
  88. 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]
  89. Hwang, R.F.; Moore, T.; Arumugam, T.; Ramachandran, V.; Amos, K.D.; Rivera, A.; Ji, B.; Evans, D.B.; Logsdon, C.D. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008, 68, 918–926. [Google Scholar] [CrossRef]
  90. Nicolle, R.; Blum, Y.; Marisa, L.; Loncle, C.; Gayet, O.; Moutardier, V.; Turrini, O.; Giovannini, M.; Bian, B.; Bigonnet, M.; et al. Pancreatic Adenocarcinoma Therapeutic Targets Revealed by Tumor-Stroma Cross-Talk Analyses in Patient-Derived Xenografts. Cell Rep. 2017, 21, 2458–2470. [Google Scholar] [CrossRef]
  91. Sun, L.; Zheng, M.; Gao, Y.; Brigstock, D.R.; Gao, R. Retinoic acid signaling pathway in pancreatic stellate cells: Insight into the anti-fibrotic effect and mechanism. Eur. J. Pharmacol. 2024, 967, 176374. [Google Scholar] [CrossRef]
  92. Guan, J.; Zhang, H.; Wen, Z.; Gu, Y.; Cheng, Y.; Sun, Y.; Zhang, T.; Jia, C.; Lu, Z.; Chen, J. Retinoic acid inhibits pancreatic cancer cell migration and EMT through the downregulation of IL-6 in cancer associated fibroblast cells. Cancer Lett. 2014, 345, 132–139. [Google Scholar] [CrossRef]
  93. Go, Y.H.; Choi, W.H.; Bae, W.J.; Jung, S.I.; Cho, C.H.; Lee, S.A.; Park, J.S.; Ahn, J.M.; Kim, S.W.; Lee, K.J.; et al. Modeling Pancreatic Cancer with Patient-Derived Organoids Integrating Cancer-Associated Fibroblasts. Cancers 2022, 14, 2077. [Google Scholar] [CrossRef] [PubMed]
  94. Chia, L.; Wang, B.; Kim, J.H.; Luo, L.Z.; Shuai, S.; Herrera, I.; Chen, S.Y.; Li, L.; Xian, L.; Huso, T.; et al. HMGA1 induces FGF19 to drive pancreatic carcinogenesis and stroma formation. J. Clin. Investig. 2023, 133. [Google Scholar] [CrossRef]
  95. Parte, S.; Kaur, A.B.; Nimmakayala, R.K.; Ogunleye, A.O.; Chirravuri, R.; Vengoji, R.; Leon, F.; Nallasamy, P.; Rauth, S.; Alsafwani, Z.W.; et al. Cancer-Associated Fibroblast Induces Acinar-to-Ductal Cell Transdifferentiation and Pancreatic Cancer Initiation Via LAMA5/ITGA4 Axis. Gastroenterology 2024, 166, 842–858.e845. [Google Scholar] [CrossRef]
  96. Picard, F.S.R.; Lutz, V.; Brichkina, A.; Neuhaus, F.; Ruckenbrod, T.; Hupfer, A.; Raifer, H.; Klein, M.; Bopp, T.; Pfefferle, P.I.; et al. IL-17A-producing CD8(+) T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut 2023, 72, 1510–1522. [Google Scholar] [CrossRef] [PubMed]
  97. Salvioli, B.; Bovara, M.; Barbara, G.; De Ponti, F.; Stanghellini, V.; Tonini, M.; Guerrini, S.; Cremon, C.; Degli Esposti, M.; Koumandou, M.; et al. Neurology and neuropathology of the pancreatic innervation. JOP 2002, 3, 26–33. [Google Scholar] [PubMed]
  98. Demir, I.E.; Friess, H.; Ceyhan, G.O. Neural plasticity in pancreatitis and pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 649–659. [Google Scholar] [CrossRef]
  99. Boilly, B.; Faulkner, S.; Jobling, P.; Hondermarck, H. Nerve Dependence: From Regeneration to Cancer. Cancer Cell 2017, 31, 342–354. [Google Scholar] [CrossRef]
  100. Dai, H.; Li, R.; Wheeler, T.; Ozen, M.; Ittmann, M.; Anderson, M.; Wang, Y.; Rowley, D.; Younes, M.; Ayala, G.E. Enhanced survival in perineural invasion of pancreatic cancer: An in vitro approach. Hum. Pathol. 2007, 38, 299–307. [Google Scholar] [CrossRef]
  101. Gil, Z.; Cavel, O.; Kelly, K.; Brader, P.; Rein, A.; Gao, S.P.; Carlson, D.L.; Shah, J.P.; Fong, Y.; Wong, R.J. Paracrine regulation of pancreatic cancer cell invasion by peripheral nerves. J. Natl. Cancer Inst. 2010, 102, 107–118. [Google Scholar] [CrossRef]
  102. Liebl, F.; Demir, I.E.; Mayer, K.; Schuster, T.; D’Haese, J.G.; Becker, K.; Langer, R.; Bergmann, F.; Wang, K.; Rosenberg, R.; et al. The impact of neural invasion severity in gastrointestinal malignancies: A clinicopathological study. Ann. Surg. 2014, 260, 900–907; discussion 907–908. [Google Scholar] [CrossRef]
  103. Li, J.; Kang, R.; Tang, D. Cellular and molecular mechanisms of perineural invasion of pancreatic ductal adenocarcinoma. Cancer Commun. 2021, 41, 642–660. [Google Scholar] [CrossRef] [PubMed]
  104. Ceyhan, G.O.; Demir, I.E.; Altintas, B.; Rauch, U.; Thiel, G.; Müller, M.W.; Giese, N.A.; Friess, H.; Schäfer, K.H. Neural invasion in pancreatic cancer: A mutual tropism between neurons and cancer cells. Biochem. Biophys. Res. Commun. 2008, 374, 442–447. [Google Scholar] [CrossRef] [PubMed]
  105. Ketterer, K.; Rao, S.; Friess, H.; Weiss, J.; Büchler, M.W.; Korc, M. Reverse transcription-PCR analysis of laser-captured cells points to potential paracrine and autocrine actions of neurotrophins in pancreatic cancer. Clin. Cancer Res. 2003, 9, 5127–5136. [Google Scholar] [PubMed]
  106. Banh, R.S.; Biancur, D.E.; Yamamoto, K.; Sohn, A.S.W.; Walters, B.; Kuljanin, M.; Gikandi, A.; Wang, H.; Mancias, J.D.; Schneider, R.J.; et al. Neurons Release Serine to Support mRNA Translation in Pancreatic Cancer. Cell 2020, 183, 1202–1218.e1225. [Google Scholar] [CrossRef]
  107. Saloman, J.L.; Singhi, A.D.; Hartman, D.J.; Normolle, D.P.; Albers, K.M.; Davis, B.M. Systemic Depletion of Nerve Growth Factor Inhibits Disease Progression in a Genetically Engineered Model of Pancreatic Ductal Adenocarcinoma. Pancreas 2018, 47, 856–863. [Google Scholar] [CrossRef]
  108. Airaksinen, M.S.; Saarma, M. The GDNF family: Signalling, biological functions and therapeutic value. Nat. Rev. Neurosci. 2002, 3, 383–394. [Google Scholar] [CrossRef]
  109. Zeng, Q.; Cheng, Y.; Zhu, Q.; Yu, Z.; Wu, X.; Huang, K.; Zhou, M.; Han, S.; Zhang, Q. The relationship between overexpression of glial cell-derived neurotrophic factor and its RET receptor with progression and prognosis of human pancreatic cancer. J. Int. Med. Res. 2008, 36, 656–664. [Google Scholar] [CrossRef]
  110. Lian, E.Y.; Hyndman, B.D.; Moodley, S.; Maritan, S.M.; Mulligan, L.M. RET isoforms contribute differentially to invasive processes in pancreatic ductal adenocarcinoma. Oncogene 2020, 39, 6493–6510. [Google Scholar] [CrossRef]
  111. Marchesi, F.; Piemonti, L.; Fedele, G.; Destro, A.; Roncalli, M.; Albarello, L.; Doglioni, C.; Anselmo, A.; Doni, A.; Bianchi, P.; et al. The chemokine receptor CX3CR1 is involved in the neural tropism and malignant behavior of pancreatic ductal adenocarcinoma. Cancer Res. 2008, 68, 9060–9069. [Google Scholar] [CrossRef]
  112. Nan, L.; Qin, T.; Xiao, Y.; Qian, W.; Li, J.; Wang, Z.; Ma, J.; Ma, Q.; Wu, Z. Pancreatic Stellate Cells Facilitate Perineural Invasion of Pancreatic Cancer via HGF/c-Met Pathway. Cell Transplant. 2019, 28, 1289–1298. [Google Scholar] [CrossRef]
  113. Guo, K.; Ma, Q.; Li, J.; Wang, Z.; Shan, T.; Li, W.; Xu, Q.; Xie, K. Interaction of the sympathetic nerve with pancreatic cancer cells promotes perineural invasion through the activation of STAT3 signaling. Mol. Cancer Ther. 2013, 12, 264–273. [Google Scholar] [CrossRef] [PubMed]
  114. Renz, B.W.; Takahashi, R.; Tanaka, T.; Macchini, M.; Hayakawa, Y.; Dantes, Z.; Maurer, H.C.; Chen, X.; Jiang, Z.; Westphalen, C.B.; et al. β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell 2018, 33, 75–90.e77. [Google Scholar] [CrossRef] [PubMed]
  115. Swanson, B.J.; McDermott, K.M.; Singh, P.K.; Eggers, J.P.; Crocker, P.R.; Hollingsworth, M.A. MUC1 is a counter-receptor for myelin-associated glycoprotein (Siglec-4a) and their interaction contributes to adhesion in pancreatic cancer perineural invasion. Cancer Res. 2007, 67, 10222–10229. [Google Scholar] [CrossRef] [PubMed]
  116. Deborde, S.; Omelchenko, T.; Lyubchik, A.; Zhou, Y.; He, S.; McNamara, W.F. Schwann Cells Promote Cancer Cell Invasion. Cancer Discov. 2016, 6, 473. [Google Scholar] [CrossRef]
  117. Deborde, S.; Omelchenko, T.; Lyubchik, A.; Zhou, Y.; He, S.; McNamara, W.F.; Chernichenko, N.; Lee, S.Y.; Barajas, F.; Chen, C.H.; et al. Schwann cells induce cancer cell dispersion and invasion. J. Clin. Investig. 2016, 126, 1538–1554. [Google Scholar] [CrossRef]
  118. Na’ara, S.; Amit, M.; Gil, Z. L1CAM induces perineural invasion of pancreas cancer cells by upregulation of metalloproteinase expression. Oncogene 2019, 38, 596–608. [Google Scholar] [CrossRef]
  119. Roger, E.; Martel, S.; Bertrand-Chapel, A.; Depollier, A.; Chuvin, N.; Pommier, R.M.; Yacoub, K.; Caligaris, C.; Cardot-Ruffino, V.; Chauvet, V.; et al. Schwann cells support oncogenic potential of pancreatic cancer cells through TGFβ signaling. Cell Death Dis. 2019, 10, 886. [Google Scholar] [CrossRef]
  120. Bakst, R.L.; Xiong, H.; Chen, C.-H.; Deborde, S.; Lyubchik, A.; Zhou, Y.; He, S.; McNamara, W.; Lee, S.-Y.; Olson, O.C.; et al. Inflammatory Monocytes Promote Perineural Invasion via CCL2-Mediated Recruitment and Cathepsin B Expression. Cancer Res. 2017, 77, 6400–6414. [Google Scholar] [CrossRef]
  121. Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef]
  122. Azad, A.; Yin Lim, S.; D’Costa, Z.; Jones, K.; Diana, A.; Sansom, O.J.; Kruger, P.; Liu, S.; McKenna, W.G.; Dushek, O.; et al. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol. Med. 2017, 9, 167–180. [Google Scholar] [CrossRef]
  123. Furuhashi, S.; Sakaguchi, T.; Murakami, T.; Fukushima, M.; Morita, Y.; Ikegami, K.; Kikuchi, H.; Setou, M.; Takeuchi, H. Tenascin C in the Tumor-Nerve Microenvironment Enhances Perineural Invasion and Correlates With Locoregional Recurrence in Pancreatic Ductal Adenocarcinoma. Pancreas 2020, 49, 442–454. [Google Scholar] [CrossRef] [PubMed]
  124. Li, X.; Wang, Z.; Ma, Q.; Xu, Q.; Liu, H.; Duan, W.; Lei, J.; Ma, J.; Wang, X.; Lv, S.; et al. Sonic hedgehog paracrine signaling activates stromal cells to promote perineural invasion in pancreatic cancer. Clin. Cancer Res. 2014, 20, 4326–4338. [Google Scholar] [CrossRef] [PubMed]
  125. Zeng, L.; Guo, Y.; Liang, J.; Chen, S.; Peng, P.; Zhang, Q.; Su, H.; Chen, Y.; Huang, K. Perineural Invasion and TAMs in Pancreatic Ductal Adenocarcinomas: Review of the Original Pathology Reports Using Immunohistochemical Enhancement and Relationships with Clinicopathological Features. J. Cancer 2014, 5, 754–760. [Google Scholar] [CrossRef] [PubMed]
  126. Cavel, O.; Shomron, O.; Shabtay, A.; Vital, J.; Trejo-Leider, L.; Weizman, N.; Krelin, Y.; Fong, Y.; Wong, R.J.; Amit, M.; et al. Endoneurial macrophages induce perineural invasion of pancreatic cancer cells by secretion of GDNF and activation of RET tyrosine kinase receptor. Cancer Res. 2012, 72, 5733–5743. [Google Scholar] [CrossRef]
  127. Yang, M.W.; Tao, L.Y.; Jiang, Y.S.; Yang, J.Y.; Huo, Y.M.; Liu, D.J.; Li, J.; Fu, X.L.; He, R.; Lin, C.; et al. Perineural Invasion Reprograms the Immune Microenvironment through Cholinergic Signaling in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2020, 80, 1991–2003. [Google Scholar] [CrossRef]
  128. Partecke, L.I.; Käding, A.; Trung, D.N.; Diedrich, S.; Sendler, M.; Weiss, F.; Kühn, J.P.; Mayerle, J.; Beyer, K.; von Bernstorff, W.; et al. Subdiaphragmatic vagotomy promotes tumor growth and reduces survival via TNFα in a murine pancreatic cancer model. Oncotarget 2017, 8, 22501–22512. [Google Scholar] [CrossRef]
  129. Koide, N.; Yamada, T.; Shibata, R.; Mori, T.; Fukuma, M.; Yamazaki, K.; Aiura, K.; Shimazu, M.; Hirohashi, S.; Nimura, Y.; et al. Establishment of perineural invasion models and analysis of gene expression revealed an invariant chain (CD74) as a possible molecule involved in perineural invasion in pancreatic cancer. Clin. Cancer Res. 2006, 12, 2419–2426. [Google Scholar] [CrossRef]
  130. Torer, N.; Kayaselcuk, F.; Nursal, T.Z.; Yildirim, S.; Tarim, A.; Nòyan, T.; Karakayali, H. Adhesion molecules as prognostic markers in pancreatic adenocarcinoma. J. Surg. Oncol. 2007, 96, 419–423. [Google Scholar] [CrossRef]
  131. Lebe, B.; Sağol, O.; Ulukuş, C.; Coker, A.; Karademir, S.; Astarcioglu, H.; Küpelioğlu, A.; Astarcioğlu, I.; Obuz, F. The importance of cyclin D1 and Ki67 expression on the biological behavior of pancreatic adenocarcinomas. Pathol. Res. Pract. 2004, 200, 389–396. [Google Scholar] [CrossRef]
  132. Bianchini, M.; Giambelluca, M.; Scavuzzo, M.C.; Di Franco, G.; Guadagni, S.; Palmeri, M.; Furbetta, N.; Gianardi, D.; Costa, A.; Gentiluomo, M.; et al. In Pancreatic Adenocarcinoma Alpha-Synuclein Increases and Marks Peri-Neural Infiltration. Int. J. Mol. Sci. 2022, 23, 3775. [Google Scholar] [CrossRef]
  133. Jurcak, N.R.; Rucki, A.A.; Muth, S.; Thompson, E.; Sharma, R.; Ding, D.; Zhu, Q.; Eshleman, J.R.; Anders, R.A.; Jaffee, E.M.; et al. Axon Guidance Molecules Promote Perineural Invasion and Metastasis of Orthotopic Pancreatic Tumors in Mice. Gastroenterology 2019, 157, 838–850.e836. [Google Scholar] [CrossRef] [PubMed]
  134. Selvaraj, D.; Hirth, M.; Gandla, J.; Kuner, R. A mouse model for pain and neuroplastic changes associated with pancreatic ductal adenocarcinoma. Pain 2017, 158, 1609–1621. [Google Scholar] [CrossRef] [PubMed]
  135. Demir, I.E.; Schorn, S.; Schremmer-Danninger, E.; Wang, K.; Kehl, T.; Giese, N.A.; Algül, H.; Friess, H.; Ceyhan, G.O. Perineural Mast Cells Are Specifically Enriched in Pancreatic Neuritis and Neuropathic Pain in Pancreatic Cancer and Chronic Pancreatitis. PLoS ONE 2013, 8, e60529. [Google Scholar] [CrossRef] [PubMed]
  136. Sun, L.; Zhang, X.; Song, Q.; Liu, L.; Forbes, E.; Tian, W.; Zhang, Z.; Kang, Y.; Wang, H.; Fleming, J.B.; et al. IGFBP2 promotes tumor progression by inducing alternative polarization of macrophages in pancreatic ductal adenocarcinoma through the STAT3 pathway. Cancer Lett. 2021, 500, 132–146. [Google Scholar] [CrossRef]
  137. Zhu, Y.; Herndon, J.M.; Sojka, D.K.; Kim, K.W.; Knolhoff, B.L.; Zuo, C.; Cullinan, D.R.; Luo, J.; Bearden, A.R.; Lavine, K.J.; et al. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017, 47, 323–338.e326. [Google Scholar] [CrossRef]
  138. Sperti, C.; Pasquali, C.; Piccoli, A.; Pedrazzoli, S. Survival after resection for ductal adenocarcinoma of the pancreas. Br. J. Surg. 1996, 83, 625–631. [Google Scholar] [CrossRef]
  139. Chen, J.W.; Bhandari, M.; Astill, D.S.; Wilson, T.G.; Kow, L.; Brooke-Smith, M.; Toouli, J.; Padbury, R.T. Predicting patient survival after pancreaticoduodenectomy for malignancy: Histopathological criteria based on perineural infiltration and lymphovascular invasion. HPB 2010, 12, 101–108. [Google Scholar] [CrossRef]
  140. Crippa, S.; Pergolini, I.; Javed, A.A.; Honselmann, K.C.; Weiss, M.J.; Di Salvo, F.; Burkhart, R.; Zamboni, G.; Belfiori, G.; Ferrone, C.R.; et al. Implications of Perineural Invasion on Disease Recurrence and Survival After Pancreatectomy for Pancreatic Head Ductal Adenocarcinoma. Ann. Surg. 2022, 276, 378–385. [Google Scholar] [CrossRef]
  141. Li, B.; Wang, Y.; Jiang, H.; Li, B.; Shi, X.; Gao, S.; Ni, C.; Zhang, Z.; Guo, S.; Xu, J.; et al. Pros and Cons: High Proportion of Stromal Component Indicates Better Prognosis in Patients With Pancreatic Ductal Adenocarcinoma-A Research Based on the Evaluation of Whole-Mount Histological Slides. Front. Oncol. 2020, 10, 1472. [Google Scholar] [CrossRef]
  142. Kondo, N.; Murakami, Y.; Uemura, K.; Hashimoto, Y.; Nakagawa, N.; Sasaki, H.; Sueda, T. An Increased Number of Perineural Invasions Is Independently Associated With Poor Survival of Patients With Resectable Pancreatic Ductal Adenocarcinoma. Pancreas 2015, 44, 1345–1351. [Google Scholar] [CrossRef]
  143. Zhou, W.; Jin, W.; Wang, D.; Lu, C.; Xu, X.; Zhang, R.; Kuang, T.; Zhou, Y.; Wu, W.; Jin, D.; et al. Laparoscopic versus open pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: A propensity score matching analysis. Cancer Commun. 2019, 39, 66. [Google Scholar] [CrossRef] [PubMed]
  144. McCracken, M.N.; Cha, A.C.; Weissman, I.L. Molecular Pathways: Activating T Cells after Cancer Cell Phagocytosis from Blockade of CD47 “Don’t Eat Me” Signals. Clin. Cancer Res. 2015, 21, 3597–3601. [Google Scholar] [CrossRef] [PubMed]
  145. Michaels, A.D.; Newhook, T.E.; Adair, S.J.; Morioka, S.; Goudreau, B.J.; Nagdas, S.; Mullen, M.G.; Persily, J.B.; Bullock, T.N.J.; Slingluff, C.L., Jr.; et al. CD47 Blockade as an Adjuvant Immunotherapy for Resectable Pancreatic Cancer. Clin. Cancer Res. 2018, 24, 1415–1425. [Google Scholar] [CrossRef] [PubMed]
  146. Kadera, B.E.; Sunjaya, D.B.; Isacoff, W.H.; Li, L.; Hines, O.J.; Tomlinson, J.S.; Dawson, D.W.; Rochefort, M.M.; Donald, G.W.; Clerkin, B.M.; et al. Locally Advanced Pancreatic Cancer: Association Between Prolonged Preoperative Treatment and Lymph-Node Negativity and Overall Survival. JAMA Surg. 2014, 149, 145–153. [Google Scholar] [CrossRef]
  147. Zhu, J.; Miao, X.-R.; Tao, K.-M.; Zhu, H.; Liu, Z.-Y.; Yu, D.-W.; Chen, Q.-B.; Qiu, H.-B.; Lu, Z.-J. Trypsin-protease activated receptor-2 signaling contributes to pancreatic cancer pain. Oncotarget 2017, 8, 61810. [Google Scholar] [CrossRef]
  148. Karamitopoulou, E.; Shoni, M.; Theoharides, T.C. Increased number of non-degranulated mast cells in pancreatic ductal adenocarcinoma but not in acute pancreatitis. Int. J. Immunopathol. Pharmacol. 2014, 27, 213–220. [Google Scholar] [CrossRef]
  149. Demir, I.E.; Kujundzic, K.; Pfitzinger, P.L.; Saricaoglu, Ö.C.; Teller, S.; Kehl, T.; Reyes, C.M.; Ertl, L.S.; Miao, Z.; Schall, T.J.; et al. Early pancreatic cancer lesions suppress pain through CXCL12-mediated chemoattraction of Schwann cells. Proc. Natl. Acad. Sci. USA 2017, 114, E85–E94. [Google Scholar] [CrossRef]
  150. Demir, I.E.; Tieftrunk, E.; Schorn, S.; Saricaoglu, Ö.C.; Pfitzinger, P.L.; Teller, S.; Wang, K.; Waldbaur, C.; Kurkowski, M.U.; Wörmann, S.M.; et al. Activated Schwann cells in pancreatic cancer are linked to analgesia via suppression of spinal astroglia and microglia. Gut 2016, 65, 1001–1014. [Google Scholar] [CrossRef]
  151. Chatterjee, D.; Katz, M.H.; Rashid, A.; Wang, H.; Iuga, A.C.; Varadhachary, G.R.; Wolff, R.A.; Lee, J.E.; Pisters, P.W.; Crane, C.H.; et al. Perineural and intraneural invasion in posttherapy pancreaticoduodenectomy specimens predicts poor prognosis in patients with pancreatic ductal adenocarcinoma. Am. J. Surg. Pathol. 2012, 36, 409–417. [Google Scholar] [CrossRef]
  152. Bakst, R.L.; Lee, N.; He, S.; Chernichenko, N.; Chen, C.H.; Linkov, G.; Le, H.C.; Koutcher, J.; Vakiani, E.; Wong, R.J. Radiation impairs perineural invasion by modulating the nerve microenvironment. PLoS ONE 2012, 7, e39925. [Google Scholar] [CrossRef]
  153. Lu, Z.; Dong, T.H.; Si, P.R.; Shen, W.; Bi, Y.L.; Min, M.; Chen, X.; Liu, Y. Continuous Low-dose-rate Irradiation of Iodine-125 Seeds Inhibiting Perineural Invasion in Pancreatic Cancer. Chin. Med. J. 2016, 129, 2460–2468. [Google Scholar] [CrossRef] [PubMed]
  154. Koujima, T.; Tazawa, H.; Ieda, T.; Araki, H.; Fushimi, T.; Shoji, R.; Kuroda, S.; Kikuchi, S.; Yoshida, R.; Umeda, Y.; et al. Oncolytic Virus-Mediated Targeting of the ERK Signaling Pathway Inhibits Invasive Propensity in Human Pancreatic Cancer. Mol. Ther. Oncolytics 2020, 17, 107–117. [Google Scholar] [CrossRef] [PubMed]
  155. Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806.e712. [Google Scholar] [CrossRef] [PubMed]
  156. Aykut, B.; Pushalkar, S.; Chen, R.; Li, Q.; Abengozar, R.; Kim, J.I.; Shadaloey, S.A.; Wu, D.; Preiss, P.; Verma, N.; et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574, 264–267. [Google Scholar] [CrossRef]
  157. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
  158. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  159. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef]
  160. Kim, Y.I.; Park, J.E.; Brand, D.D.; Fitzpatrick, E.A.; Yi, A.K. Protein kinase D1 is essential for the proinflammatory response induced by hypersensitivity pneumonitis-causing thermophilic actinomycetes Saccharopolyspora rectivirgula. J. Immunol. 2010, 184, 3145–3156. [Google Scholar] [CrossRef]
  161. Hezaveh, K.; Shinde, R.S.; Klötgen, A.; Halaby, M.J.; Lamorte, S.; Ciudad, M.T.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022, 55, 324–340.e328. [Google Scholar] [CrossRef]
  162. Beatty, G.L.; Delman, D.; Yu, J.; Liu, M.; Li, J.H.; Zhang, L.; Lee, J.W.; Chang, R.B.; Bahary, N.; Kennedy, E.P.; et al. Treatment Response in First-Line Metastatic Pancreatic Ductal Adenocarcinoma Is Stratified By a Composite Index of Tumor Proliferation and CD8 T-Cell Infiltration. Clin. Cancer Res. 2023, 29, 3514–3525. [Google Scholar] [CrossRef]
  163. Tintelnot, J.; Xu, Y.; Lesker, T.R.; Schönlein, M.; Konczalla, L.; Giannou, A.D.; Pelczar, P.; Kylies, D.; Puelles, V.G.; Bielecka, A.A.; et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic cancer. Nature 2023, 615, 168–174. [Google Scholar] [CrossRef]
  164. Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef]
  165. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  166. Pan, Y.; Lu, F.; Fei, Q.; Yu, X.; Xiong, P.; Yu, X.; Dang, Y.; Hou, Z.; Lin, W.; Lin, X.; et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J. Hematol. Oncol. 2019, 12, 124. [Google Scholar] [CrossRef] [PubMed]
  167. Ito, M.; Watanabe, T.; Oga, Y.; Matsumoto, S.; Kimura, N.; Nagamori, M.; Tanaka, H.; Shibuya, K.; Yoshioka, I.; Fujii, T. A case of MSI-high pancreatic body-tail cancer successfully treated with radical resection after pembrolizumab. Clin. J. Gastroenterol. 2025, 18, 208–213. [Google Scholar] [CrossRef] [PubMed]
  168. Fudalej, M.; Kwaśniewska, D.; Nurzyński, P.; Badowska-Kozakiewicz, A.; Mękal, D.; Czerw, A.; Sygit, K.; Deptała, A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers 2023, 15, 2327. [Google Scholar] [CrossRef] [PubMed]
  169. McCarthy, P.M.; Rendo, M.J.; Uy, M.D.; Adams, A.M.; O’Shea, A.E.; Nelson, D.W.; Fenderson, J.L.; Cebe, K.M.; Krell, R.W.; Clifton, G.T.; et al. Near Complete Pathologic Response to PD-1 Inhibitor and Radiotherapy in a Patient with Locally Advanced Pancreatic Ductal Adenocarcinoma. Onco Targets Ther. 2021, 14, 3537–3544. [Google Scholar] [CrossRef]
  170. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  171. Fan, J.Q.; Wang, M.F.; Chen, H.L.; Shang, D.; Das, J.K.; Song, J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol. Cancer 2020, 19, 32. [Google Scholar] [CrossRef]
  172. Poschke, I.; Faryna, M.; Bergmann, F.; Flossdorf, M.; Lauenstein, C.; Hermes, J.; Hinz, U.; Hank, T.; Ehrenberg, R.; Volkmar, M.; et al. Identification of a tumor-reactive T-cell repertoire in the immune infiltrate of patients with resectable pancreatic ductal adenocarcinoma. Oncoimmunology 2016, 5, e1240859. [Google Scholar] [CrossRef]
  173. Stromnes, I.M.; Hulbert, A.; Pierce, R.H.; Greenberg, P.D.; Hingorani, S.R. T-cell Localization, Activation, and Clonal Expansion in Human Pancreatic Ductal Adenocarcinoma. Cancer Immunol. Res. 2017, 5, 978–991. [Google Scholar] [CrossRef] [PubMed]
  174. Tassi, E.; Gavazzi, F.; Albarello, L.; Senyukov, V.; Longhi, R.; Dellabona, P.; Doglioni, C.; Braga, M.; Di Carlo, V.; Protti, M.P. Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. J. Immunol. 2008, 181, 6595–6603. [Google Scholar] [CrossRef]
  175. Turk, J.L.; Parker, D. Effect of cyclophosphamide on immunological control mechanisms. Immunol. Rev. 1982, 65, 99–113. [Google Scholar] [CrossRef] [PubMed]
  176. Madondo, M.T.; Quinn, M.; Plebanski, M. Low dose cyclophosphamide: Mechanisms of T cell modulation. Cancer Treat. Rev. 2016, 42, 3–9. [Google Scholar] [CrossRef] [PubMed]
  177. Heylmann, D.; Bauer, M.; Becker, H.; van Gool, S.; Bacher, N.; Steinbrink, K.; Kaina, B. Human CD4+CD25+ regulatory T cells are sensitive to low dose cyclophosphamide: Implications for the immune response. PLoS ONE 2013, 8, e83384. [Google Scholar] [CrossRef]
  178. Beatty, G.L.; Chiorean, E.G.; Fishman, M.P.; Saboury, B.; Teitelbaum, U.R.; Sun, W.; Huhn, R.D.; Song, W.; Li, D.; Sharp, L.L.; et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011, 331, 1612–1616. [Google Scholar] [CrossRef]
  179. 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]
  180. Haen, S.P.; Löffler, M.W.; Rammensee, H.G.; Brossart, P. Towards new horizons: Characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 2020, 17, 595–610. [Google Scholar] [CrossRef]
  181. Zuo, C.; Baer, J.M.; Knolhoff, B.L.; Belle, J.I.; Liu, X.; Alarcon De La Lastra, A.; Fu, C.; Hogg, G.D.; Kingston, N.L.; Breden, M.A.; et al. Stromal and therapy-induced macrophage proliferation promotes PDAC progression and susceptibility to innate immunotherapy. J. Exp. Med. 2023, 220, e20212062. [Google Scholar] [CrossRef]
  182. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
  183. Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
  184. Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.M.; Ries, C.H.; Rüttinger, D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef] [PubMed]
  185. 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] [PubMed]
  186. Felix, K.; Gaida, M.M. Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression. Int. J. Biol. Sci. 2016, 12, 302–313. [Google Scholar] [CrossRef] [PubMed]
  187. Bayne, L.J.; Beatty, G.L.; Jhala, N.; Clark, C.E.; Rhim, A.D.; Stanger, B.Z.; Vonderheide, R.H. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012, 21, 822–835. [Google Scholar] [CrossRef]
  188. Ray, U.; Pathoulas, C.L.; Thirusangu, P.; Purcell, J.W.; Kannan, N.; Shridhar, V. Exploiting LRRC15 as a Novel Therapeutic Target in Cancer. Cancer Res. 2022, 82, 1675–1681. [Google Scholar] [CrossRef]
  189. Challita-Eid, P.M.; Satpayev, D.; Yang, P.; An, Z.; Morrison, K.; Shostak, Y.; Raitano, A.; Nadell, R.; Liu, W.; Lortie, D.R.; et al. Enfortumab Vedotin Antibody-Drug Conjugate Targeting Nectin-4 Is a Highly Potent Therapeutic Agent in Multiple Preclinical Cancer Models. Cancer Res. 2016, 76, 3003–3013. [Google Scholar] [CrossRef]
  190. Suarez-Carmona, M.; Williams, A.; Schreiber, J.; Hohmann, N.; Pruefer, U.; Krauss, J.; Jäger, D.; Frömming, A.; Beyer, D.; Eulberg, D.; et al. Combined inhibition of CXCL12 and PD-1 in MSS colorectal and pancreatic cancer: Modulation of the microenvironment and clinical effects. J. Immunother. Cancer 2021, 9, e002505. [Google Scholar] [CrossRef]
  191. 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]
  192. Angevin, E.; Tabernero, J.; Elez, E.; Cohen, S.J.; Bahleda, R.; van Laethem, J.L.; Ottensmeier, C.; Lopez-Martin, J.A.; Clive, S.; Joly, F.; et al. A phase I/II, multiple-dose, dose-escalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors. Clin. Cancer Res. 2014, 20, 2192–2204. [Google Scholar] [CrossRef]
  193. Abdel-Wahab, R.; Varadhachary, G.R.; Bhosale, P.R.; Wang, X.; Fogelman, D.R.; Shroff, R.T.; Overman, M.J.; Wolff, R.A.; Javle, M. 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. J. Hematol. Oncol. 2018, 11, 71. [Google Scholar] [CrossRef] [PubMed]
  194. Kundranda, M.; Gracian, A.C.; Zafar, S.F.; Meiri, E.; Bendell, J.; Algül, H.; Rivera, F.; Ahn, E.R.; Watkins, D.; Pelzer, U.; et al. Randomized, double-blind, placebo-controlled phase II study of istiratumab (MM-141) plus nab-paclitaxel and gemcitabine versus nab-paclitaxel and gemcitabine in front-line metastatic pancreatic cancer (CARRIE). Ann. Oncol. 2020, 31, 79–87. [Google Scholar] [CrossRef]
  195. Fuchs, C.S.; Azevedo, S.; Okusaka, T.; Van Laethem, J.L.; Lipton, L.R.; Riess, H.; Szczylik, C.; Moore, M.J.; Peeters, M.; Bodoky, G.; 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. Ann. Oncol. 2015, 26, 921–927. [Google Scholar] [CrossRef] [PubMed]
  196. Ko, A.H.; LoConte, N.; Tempero, M.A.; Walker, E.J.; Kate Kelley, R.; Lewis, S.; Chang, W.C.; Kantoff, E.; Vannier, M.W.; Catenacci, D.V.; et al. A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma. Pancreas 2016, 45, 370–375. [Google Scholar] [CrossRef] [PubMed]
  197. De Jesus-Acosta, A.; Sugar, E.A.; O’Dwyer, P.J.; Ramanathan, R.K.; Von Hoff, D.D.; Rasheed, Z.; Zheng, L.; Begum, A.; Anders, R.; Maitra, A.; et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br. J. Cancer 2020, 122, 498–505. [Google Scholar] [CrossRef]
  198. Kim, E.J.; Sahai, V.; Abel, E.V.; Griffith, K.A.; Greenson, J.K.; Takebe, N.; Khan, G.N.; Blau, J.L.; Craig, R.; Balis, U.G.; et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin. Cancer Res. 2014, 20, 5937–5945. [Google Scholar] [CrossRef]
  199. Carr, R.M.; Duma, N.; McCleary-Wheeler, A.L.; Almada, L.L.; Marks, D.L.; Graham, R.P.; Smyrk, T.C.; Lowe, V.; Borad, M.J.; Kim, G.; et al. Targeting of the Hedgehog/GLI and mTOR pathways in advanced pancreatic cancer, a phase 1 trial of Vismodegib and Sirolimus combination. Pancreatology 2020, 20, 1115–1122. [Google Scholar] [CrossRef]
  200. Pijnappel, E.N.; Wassenaar, N.P.M.; Gurney-Champion, O.J.; Klaassen, R.; van der Lee, K.; Pleunis-van Empel, M.C.H.; Richel, D.J.; Legdeur, M.C.; Nederveen, A.J.; van Laarhoven, H.W.M.; et al. Phase I/II Study of LDE225 in Combination with Gemcitabine and Nab-Paclitaxel in Patients with Metastatic Pancreatic Cancer. Cancers 2021, 13, 4869. [Google Scholar] [CrossRef]
  201. Nagaraju, G.P.; Saddala, M.S.; Foote, J.B.; Khaliq, A.M.; Masood, A.; Golivi, Y.; Bandi, D.S.R.; Sarvesh, S.; Reddy, S.P.; Switchenko, J.; et al. Mechanism of enhancing chemotherapy efficacy in pancreatic ductal adenocarcinoma with paricalcitol and hydroxychloroquine. Cell Rep. Med. 2025, 6, 101881. [Google Scholar] [CrossRef]
  202. Chung, V.; Alistar, A.; Becerra, C.; Kasi, A.; Borazanci, E.; Jameson, G.S.; Roe, D.J.; Wertheim, B.C.; Cridebring, D.; Truitt, M.; et al. Pembrolizumab ± paricalcitol in metastatic pancreatic cancer postmaximal cytoreduction. Oncologist 2025, 30, oyae323. [Google Scholar] [CrossRef]
  203. Grierson, P.M.; Suresh, R.; Tan, B.; Pedersen, K.S.; Amin, M.; Park, H.; Trikalinos, N.A.; Liu, J.; Boice, N.; Brown, A.; et al. A Pilot Study of Paricalcitol plus Nanoliposomal Irinotecan and 5-FU/LV in Advanced Pancreatic Cancer Patients after Progression on Gemcitabine-Based Therapy. Clin. Cancer Res. 2023, 29, 4733–4739. [Google Scholar] [CrossRef] [PubMed]
  204. Kocher, H.M.; Basu, B.; Froeling, F.E.M.; Sarker, D.; Slater, S.; Carlin, D.; deSouza, N.M.; De Paepe, K.N.; Goulart, M.R.; Hughes, C.; et al. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nat. Commun. 2020, 11, 4841. [Google Scholar] [CrossRef] [PubMed]
  205. Kocher, H.M.; Sasieni, P.; Corrie, P.; McNamara, M.G.; Sarker, D.; Froeling, F.E.M.; Christie, A.; Gillmore, R.; Khan, K.; Propper, D. Study protocol: Multi-centre, randomised controlled clinical trial exploring stromal targeting in locally advanced pancreatic cancer; STARPAC2. BMC Cancer 2025, 25, 106. [Google Scholar] [CrossRef] [PubMed]
  206. Melisi, D.; Oh, D.Y.; Hollebecque, A.; Calvo, E.; Varghese, A.; Borazanci, E.; Macarulla, T.; Merz, V.; Zecchetto, C.; Zhao, Y.; et al. Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer. J. Immunother. Cancer 2021, 9, e002068. [Google Scholar] [CrossRef] [PubMed]
  207. Gardner, F.P.; Wainberg, Z.A.; Fountzilas, C.; Bahary, N.; Womack, M.S.; Macarulla, T.; Garrido-Laguna, I.; Peterson, P.M.; Borazanci, E.; Johnson, M.; et al. Results of a Randomized, Double-Blind, Placebo-Controlled, Phase 1b/2 Trial of Nabpaclitaxel + Gemcitabine ± Olaratumab in Treatment-Naïve Participants with Metastatic Pancreatic Cancer. Cancers 2024, 16, 1323. [Google Scholar] [CrossRef]
  208. Murphy, J.E.; Wo, J.Y.; Ryan, D.P.; Clark, J.W.; Jiang, W.; Yeap, B.Y.; Drapek, L.C.; Ly, L.; Baglini, C.V.; Blaszkowsky, L.S.; et al. Total Neoadjuvant Therapy With FOLFIRINOX in Combination With Losartan Followed by Chemoradiotherapy for Locally Advanced Pancreatic Cancer: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 1020–1027. [Google Scholar] [CrossRef]
  209. Mayer, P.; Jiang, Y.; Kuder, T.A.; Bergmann, F.; Khristenko, E.; Steinle, V.; Kaiser, J.; Hackert, T.; Kauczor, H.U.; Klauß, M.; et al. Diffusion Kurtosis Imaging-A Superior Approach to Assess Tumor-Stroma Ratio in Pancreatic Ductal Adenocarcinoma. Cancers 2020, 12, 1656. [Google Scholar] [CrossRef]
  210. Erkan, M.; Adler, G.; Apte, M.V.; Bachem, M.G.; Buchholz, M.; Detlefsen, S.; Esposito, I.; Friess, H.; Gress, T.M.; Habisch, H.J.; et al. StellaTUM: Current consensus and discussion on pancreatic stellate cell research. Gut 2012, 61, 172–178. [Google Scholar] [CrossRef]
Figure 1. Scheme of mutual influence between PDAC cells, IL-6+ CAFs, and TAM M1 cells.
Figure 1. Scheme of mutual influence between PDAC cells, IL-6+ CAFs, and TAM M1 cells.
Cancers 17 01599 g001
Figure 2. Scheme of PDAC, stroma cell clusters, and the most frequent metastasis locations (the liver is presented with subsegments according to the Couinaud system).
Figure 2. Scheme of PDAC, stroma cell clusters, and the most frequent metastasis locations (the liver is presented with subsegments according to the Couinaud system).
Cancers 17 01599 g002
Table 1. Examples of cells secreting cytokines and chemokines responsible for the activation of PSCs [16,21,22,23,24,25,26,27,28].
Table 1. Examples of cells secreting cytokines and chemokines responsible for the activation of PSCs [16,21,22,23,24,25,26,27,28].
Type of CellsCytokines and Chemokines
Cancer cellsTGF-β
FGF
PDGF
IGF-1
IL-1β
IL-6
Acinar cellsTGF-β
CTGF
IL-1β
TNF-α
PSCTGF-β
CTGF
IL-10
PDGF
IL-6
IGF-1
CXCL12
Inflammatory cellsIL-6
IL-10
TGF-β
IL-6
IL-1β
TNF-α
IGF-1
PlateletsPDGF
TGF-β
Table 2. Example of proteins associated with PNI.
Table 2. Example of proteins associated with PNI.
Expression
UpregulatedDownregulated
Correlation with PNIPositiveBIRC5, CCND1, CDH1, CD74, COL6A3, CTHRC1, CXCR4, IL-13RA2, LRP1, L1CAM, MAP1LC3A/B, MDK, MYBL2, MYC, PODXL, PTN, SDC2, SDC3, SNCG, TIMP2, VGF, WASL
NegativeFAS, POP1CADM4, HPGD
Table 3. Summary of TME components and their impact on tumor behavior, metastasis, and treatment response.
Table 3. Summary of TME components and their impact on tumor behavior, metastasis, and treatment response.
Component of TMEDescriptionImpact on Tumor BehaviorImpact on MetastasisImpact on Treatment Response
PSCsQuiescent cells in the exocrine pancreas; upon activation, they express markers like α-SMA and desmin and produce ECM components such as collagen and fibronectin.Promote cancer cell proliferation and migration. Inhibit apoptosis.Contribute to ECM remodeling, aiding tumor invasion and metastasis.Contribute to chemotherapy and radiotherapy resistance.
CAFsHeterogeneous cells responsible for ECM deposition, arise from fibroblasts, mesenchymal stem cells, or PSCs, and express markers like FAP, α-SMA, and vimentin.Promote tumor growth, neovascularization, and invasion through the secretion of growth factors.Enhance metastasis Via ECM remodeling and secretion of pro-invasive cytokines.Suppress anti-tumor immune responses, affecting immunotherapy efficacy.
TregsCD4+CD25+FOXP3+ cells that suppress immune responses and maintain self-tolerance.Impair anti-tumor immune responses, promoting tumor immune evasion.Facilitate metastasis by suppressing immune responses to circulating tumor cells.Correlate with poorer prognosis; suppress immune checkpoint responses.
MDSCsImmature myeloid cells that are activated by cytokines like GM-CSF and IL-13.Suppress anti-tumor immunity by activating Tregs and inhibiting CD8+ T cells.Promote immune tolerance, aiding tumor cell survival in distant sites.Limit the effectiveness of immunotherapies by suppressing T cell function.
TAMsMonocytes that differentiate into M1 or M2 macrophages; M1 promotes inflammation, but M2 is more pro-tumor.M1 promotes inflammation, M2 promotes tissue remodeling, immune suppression, and tumor progression.M2 phenotype enhances metastasis by facilitating ECM remodeling and immune evasion.M2 macrophages are linked to poor prognosis, suppressing response to treatment.
VMCancer cells form vessel-like structures independent of endothelial cells and angiogenesis.Provides an alternative tumor blood supply, facilitating nutrient and oxygen supply to the tumor.Complicates tumor vascular architecture and enhances metastasis by forming new blood pathways.Linked to poor prognosis and chemotherapy resistance.
Table 4. Clinical trials encompassing some components of TME in PC.
Table 4. Clinical trials encompassing some components of TME in PC.
Clinical TrialStatusTested DrugTargetAdditional InterventionsShort Description, Objective of the Study, Outcome, If Available
NCT06904378Phase 1 (Finished)/
Phase 2 (Not yet recruiting)
OntegimodCD11bNab-Paclitaxel, GemcitabineAn open-label phase I/II clinical trial of Ontegimod with gemcitabine and nab-paclitaxel in unresectable PDAC prior to future studies incorporating anti-PD1 checkpoint immunotherapy. Results are not available yet.
NCT06825546Phase 2 (Recruiting)IcaritinTNF-α, IL-6, PD-L1Nab-Paclitaxel, GemcitabineRegulating the tumor immune microenvironment by reducing the secretion of TNFa and IL-6 and inhibiting PD-L1 expression through decreasing MDSC proportion. Results are not available yet.
NCT06639724Phase 1 (Recruiting)FostamatinibSyk kinaseNab-Paclitaxel, GemcitabineFostamatinib is a Syk kinase inhibitor currently FDA-approved for chronic idiopathic thrombocytopenia purpura, but it has not been studied in PDAC. The investigators hypothesize that Syk inhibition reprograms macrophages to an immunostimulatory phenotype in the TME. Thus, Syk inhibition with fostamatinib in combination with chemotherapy could improve outcomes for patients with PDAC while having a favorable safety profile. Results are not available yet.
NCT06492915Phase 2 (Recruiting)ChiauranibVEGFR2, VEGFR1, VEGFR3, PDGFRa and c-Kit, Aurora B kinase, CSF-1RNab-Paclitaxel, GemcitabineChiauranib, which simultaneously targets VEGFR/Aurora B/CSF-1R, several key kinases involved in tumor angiogenesis, tumor cell mitosis, and chronic inflammatory microenvironment. Results are not available yet.
NCT06145074Phase 2 (Recruiting) Propanololβ-adrenergic receptors Assessment of density and subtypes of tumor-infiltrating lymphocytes, desmoplasia, adrenergic receptor expression, and spatial distribution of immune cells in the TME. Results are not available yet.
NCT05927142Phase 1 (Recruiting), Phase 2 (Recruiting)RintatolimodTLR3DurvalumabImmunotherapy effectiveness is improved by the agonist effect of Rintatolimod, which enhances dendritic cell maturation. Results are not available yet.
NCT05546853Phase 1 (Active, not recruiting)NP137Netrin-1FOLFIRINOXInhibiting Netrin-1 impedes EMT, thereby reducing tumor progression and metastasis. Results are not available yet.
NCT04493060Phase 2 (Active, not recruiting)NiraparibPARP1, PARP2DostarlimabThis phase II trial studies how well niraparib and dostarlimab work in treating patients with germline or somatic BRCA1/2 and PALB2 mutated pancreatic cancer that has spread to other places in the body (metastatic). Results are not available yet.
NCT03727880Phase 2 (Active, not recruiting) Defacitinib FAK kinasePembrolizumabEvaluating if reprograming the TME by targeting FAK following chemotherapy can potentiate anti-PD-1 antibody. Results are not available yet.
NCT02651727Phase 1 (Terminated)Defacitinib, FAK kinase (VS-4718)Nab-Paclitaxel, GemcitabineThe company’s decision to de-prioritize 4718 development.
NCT03085914Phase 1 (Completed), Phase 2 (Completed)Epacadostat, Indoleamine 2,3-dioxygenase 1Nab-Paclitaxel, Gemcitabine, FOLFIRINOX, Pembrolizumab, Pemetrexed, Cyclophosphamide, Carboplatin, Cisplatin,Additional cohorts (i.e., the mandatory biopsy cohorts) were designed to evaluate changes in the TME in participants with any advanced or metastatic solid tumor who had progressed on previous therapy with a PD-1 or a PD-L1 inhibitor. Results are not available yet.
NCT02600949Phase 1 (Recruiting)Synthetic Tumoor-Associated Peptide Vaccine Therapy, Personalized peptide vaccine targeting antigenes of PDAC tumorImiquimod, Pembrolizumab, SotigalimabA personalized peptide vaccine is developed from a patient’s tumor cells and blood to be used as a biological therapy. Biological therapies, such as personalized peptide vaccines, may attack tumor cells and stop them from growing or kill them. Results are not available yet.
NCT02565758Phase 1 (Completed)ABBV-085LRRC15 Targeting leucine-rich repeat-containing protein 15 (LRRC15) using specific antibody-drug conjugates (ABBV-085) has the potential to improve the outcome of patients with LRRC15+ cancers of mesenchymal origin or stromal desmoplasia [188].
NCT03932565Phase 1 (Status Unknown)CAR-T therapyFAP/Nectin-4 Targeting Nectin-4 transmembrane protein, which is highly expressed on the surface of breast cancer, bladder cancer, non-small lung cancer, and pancreatic cancer [189].
NCT03168139Phase 1/Phase 2 (Completed)NOX-A12, CXCL12PembrolizumabOlaptesed pegol (NOX-A12) targets a key chemokine in tumor TME—CXCL12, which is involved in the homeostasis of blood and immune cells. The hypothesis is that inactivation of CXCL12 by NOX-A12 makes pancreatic tumors more susceptible to immunotherapy [190].
NCT02765165Phase 1/Phase 2 (Terminated)USL311, CXCR4LomustineTerminated because of business reasons not related to safety.
NCT03277209Phase 1 (Terminated)Plerixafor, CXCR4Cemiplimab, LomustineTerminated due to slow accrual.
NCT02907099Phase 2 (Completed)BL-8040 (motixafortide)CXCR4 Blocking CXCR4 with BL-8040 may stop the growth of tumor cells by blocking some of the enzymes needed for cell growth [191].
NCT00841191Phase 1/Phase 2 (Completed)SiltuximabIL-6 IL-6 is one of the main agents responsible for inflammation. Blocking IL-6 with siltuximab was tolerated, but without clinical activity in solid tumors, including ovarian and KRAS-mutant cancers [192].
NCT00769483Phase 1/Phase 2 (Completed)MK-0646, IGF1RGemcitabine, ErlotinibMK-0646 and Ganitumab (AMG 479) are monoclonal antibodies, while Istiratumab is a bispecific antibody, with all three targeting IGF1R (with additional blockade of ErbB3 by Istiratumab), preventing its binding with IGF1 and IGF2 ligands and inhibiting downstream signaling pathways, such as Pl3K/Akt and MAPK, which are involved in promoting tumor cell proliferation and survival and resistance to apoptosis. All three trials did not show prolonged PFS or OS compared to standard chemotherapy regimens [193,194,195].
NCT02399137Phase 2 (Completed)MM-141 (istiratumab), Nab-Paclitaxel, Gemcitabine
NCT01231347Phase 3 (Completed)AMG 479 (ganitumab), Gemcitabine
NCT01383538Phase 1 (Completed)IPI-926HedgehogFOLFIRINOXIPI-926 is an oral SHH pathway inhibitor. The initial response rate was high, and patients receiving IPI-926 maintenance showed further declines in CA19-9 levels even after FOLFIRINOX discontinuation. However, the trial was closed early, as a separate phase II trial of IPI-926 + gemcitabine indicated the detrimental effects of this combination [196].
NCT01537107Phase 1 (Completed)VismodegibSirolimusVismodegib is an SHH pathway inhibitor. SHH signaling is predominantly active in stromal cells rather than tumor cells, leading to desmoplasia and creating a dense TME. By inhibiting this pathway, vismodegib was supposed to remodel TME and enhance chemotherapy efficacy. However, all three studies showed that adding vismodegib did not enhance efficacy compared to standard chemotherapy regimens [197,198,199].
NCT01088815Phase 2 (Completed)VismodegibNab-Paclitaxel, Gemcitabine
NCT01195415Phase 2 (Completed)Vismodegib, Gemcitabine hydrochloride
NCT01485744Phase 1 (Completed)LDE225 FOLFIRINOXLDE225, in combination with gemcitabine and nab-paclitaxel, was well-tolerated in patients with metastatic PDAC and has a promising efficacy after prior treatment with FOLFIRINOX. Quantitative MRI suggested that LDE225 causes increased tumor diffusion and works particularly well in patients with poor baseline tumor perfusion [200].
NCT02358161Phase 1/Phase 2 (Completed)LDE225Nab-Paclitaxel, Gemcitabine
NCT03472833Phase 3 (Terminated)High dose vitamin DVitamin D metabolism Slow recruitment and patients lost to follow-up due to the COVID-19 pandemic.
NCT04524702Phase 2 (Active, not recruiting)ParicalcitolNab-Paclitaxel, Gemcitabine, HydroxychloroquineIn preclinical models, the combination of paricalcitol and hydroxychloroquine has been shown to remodel TME by reducing stromal activation, decreasing cancer-associated fibroblasts, and enhancing immune cell infiltration. However, the efficacy of this regimen in the clinical trial was hard to establish due to the early termination of the trial due to COVID-19 and lower-than-expected enrolment. No published data on OS or PFS outcomes from this are available [201].
NCT04617067Phase 2 (Completed)Nab-Paclitaxel, Gemcitabine Results are not available at the moment.
NCT03520790Phase 1 (Terminated) Phase II was not pursued due to futility based on the NAPOLI-3 therapeutic clinical trial results.
NCT03519308Early Phase 1 (TerminatedNab-Paclitaxel, Gemcitabine, NivolumabThe accrual goal was unmet, and the drug manufacturer pulled support.
NCT03331562Phase 2 (Completed)PembrolizumabParicalcitol did not improve pembrolizumab’s efficacy, likely related to its short half-life of only 5–7 h [202].
NCT03883919Phase 1 (Completed)5-Fluorouracil, Liposomal Irinotecan, LeucovorinThe study showed increased tumor vascularity, potentially enhancing treatment efficacy. However, more insight is needed [203].
NCT03307148Phase 1 (Completed)ATRAVitamin A metabolismNab-Paclitaxel, GemcitabineThe study showed that the combination of ATRA and Nab-Paclitaxel + Gemcitabine was safe and tolerable, establishing the recommended phase 2 dose. Pharmacodynamic studies indicated stromal modulation consistent with the proposed mechanism of action [204].
NCT04241276Phase 2 (Active, not recruiting)No clinical outcomes such as OS or PFS are reported at this stage [205].
NCT04390763Phase 2 (Terminated)NIS793TGF-βNab-Paclitaxel, Gemcitabine, FOLFIRINOX, SpartalizumabThe study was terminated early following the NIS793 treatment halt and urgent safety measure issued in July 2023, as the continued evaluation of Standard of Care alone will not support the original purpose of this phase 2 clinical trial.
NCT04935359Phase 3 (Completed)Nab-Paclitaxel, GemcitabineResults are not available at the moment.
NCT05417386Phase 1 (Terminated)FOLFIRINOXNIS793 is no longer being developed.
NCT02734160Phase 1 (Completed)Galunisertib Durvalumab, GemcitabineMedian OS was 5.72 months, and PFS was 1.87 months. The study concluded that, while the combination was tolerable, its clinical efficacy was modest, indicating that future research might focus on earlier lines of treatment or patient selection based on predictive biomarkers for TGF-β inhibition [206].
NCT04327986Phase 1/Phase 2 (Terminated) M7824GemcitabineThe study closed to accrual due to the worsening risk-benefit ratio for participants receiving bintrafusp alfa (M7824).
NCT03451773Phase 1/Phase 2 (Terminated)The study was closed after one treatment-related death.
NCT03086369Phase 1/Phase 2 (Completed)Olaratumab PDGF-αNab-Paclitaxel, GemcitabineThe primary endpoint, OS, was not met: median OS was 9.1 months in the olaratumab arm versus 10.8 months in the placebo arm. Similarly, median PFS was 5.5 months with olaratumab compared to 6.4 months with placebo [207].
NCT02550327Early Phase 1 (Completed)AnakinraIL-1Nab-Paclitaxel, Gemcitabine, CisplatinResults are not available at the moment.
NCT04999969Phase 2 (Active, not recruiting)AZD0171LIFNab-Paclitaxel, Gemcitabine, DurvalumabResults are not available at the moment.
NCT02119663Phase 3 (Terminated)RuxolitinibJAKCapecitabineThe safety committee found no safety issues, but recommended halting the study based on a lack of efficacy in a similar trial. The sponsor terminated the trial.
NCT02117479Phase 3 (Terminated)The study was terminated early based on the results of the planned interim analysis.
NCT03563248Phase 2 (Active, not recruiting)Losartan Collagen and HA degradationFOLFIRINOXResults are not available at the moment.
NCT01821729Phase 2 (Unknown status)In this single-arm phase 2 trial of 49 patients, the R0 resection rate was 61% among all eligible participants, with a PFS of 17.5 months and OS of 31.4 months, which suggests that this total neoadjuvant approach may offer improved respectability and survival benefits in this patient population [208].
NCT02715804Phase 3 (Terminated) PEGPH20, nab-paclitaxel, gemcitabineHyaluronan acid Sponsor decision.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Glapiński, F.; Zając, W.; Fudalej, M.; Deptała, A.; Czerw, A.; Sygit, K.; Kozłowski, R.; Badowska-Kozakiewicz, A. The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers 2025, 17, 1599. https://doi.org/10.3390/cancers17101599

AMA Style

Glapiński F, Zając W, Fudalej M, Deptała A, Czerw A, Sygit K, Kozłowski R, Badowska-Kozakiewicz A. The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers. 2025; 17(10):1599. https://doi.org/10.3390/cancers17101599

Chicago/Turabian Style

Glapiński, Franciszek, Weronika Zając, Marta Fudalej, Andrzej Deptała, Aleksandra Czerw, Katarzyna Sygit, Remigiusz Kozłowski, and Anna Badowska-Kozakiewicz. 2025. "The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies" Cancers 17, no. 10: 1599. https://doi.org/10.3390/cancers17101599

APA Style

Glapiński, F., Zając, W., Fudalej, M., Deptała, A., Czerw, A., Sygit, K., Kozłowski, R., & Badowska-Kozakiewicz, A. (2025). The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers, 17(10), 1599. https://doi.org/10.3390/cancers17101599

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