Next Article in Journal
Acute Kidney Injury Biomarkers in Marathon Runners: Systematic Review and Meta-Analysis
Previous Article in Journal
ICU Admission-Related Factors Affecting the Duration of Mechanical Ventilation After Elective Cardiac Surgery—Retrospective Cohort Study from a Tertiary Center in Croatia
Previous Article in Special Issue
Improving Outcomes in Pancreatic Adenocarcinoma: A Systematic Review of Immunotherapy in Multimodal Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Microenvironment: Recent Advances in Immunotherapies of Pancreatic Cancer

by
Sharon Varghese Thankachan
1,†,
Vijayalakshmi Jayaraman
1,
Liza Datta
1,
Soniga Apthi
1,
Binish Fatima Zaman
1,
Raghav Gurunathan
1,
Anuppama Suresh
1,
Parthasarathy Chandrakesan
2,
Ramachandran Vinayagam
3,†,
Sang Gu Kang
3,*,
Kanagaraj Palaniyandi
1,† and
Dhanavathy Gnanasampanthapandian
1,*
1
Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India
2
Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
3
Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Medicina 2025, 61(10), 1776; https://doi.org/10.3390/medicina61101776
Submission received: 1 July 2025 / Revised: 18 September 2025 / Accepted: 25 September 2025 / Published: 1 October 2025
(This article belongs to the Special Issue Pancreatic Cancer: Advances in Treatment and Future Prospects)

Abstract

The progression of pancreatic cancer (PC) is significantly influenced by the immune system. In the United States, PC is the third leading cause of cancer-related mortality. The high lethality of PC is attributed to its immunological advantage, which is facilitated by an immunosuppressive microenvironment, a low mutational burden, and minimal T-cell infiltration. Although immunotherapies, such as checkpoint blockades or genetically engineered T cells, have not yet demonstrated viability, there is a growing body of evidence suggesting that innovative combinations of conventional therapies and various procedures may lead to effective immunotherapy in the treatment of PC. This review focuses on the importance of the tumor microenvironment and the promising role of immunotherapies in PC.

Graphical Abstract

1. Introduction

Pancreatic cancer (PC) is recognized as one of the most aggressive malignancies. The survival rate of PC is <5 years, with patients exhibiting a survival rate of only 8.2%. By 2030, PC is projected to become the second leading cause of cancer-related deaths. Diagnosed PC is higher in males (53%) than females (47%) [1]. In 2017, PC was the third most common cause of cancer-related mortality in the United States. Regrettably, ~80–85% of PC cases are unresectable. Despite significant advancements in PC research, the prognosis for this disease remains poor. PC demonstrates a high level of resistance to chemotherapy, radiotherapy, and immunotherapy. The tumor microenvironment (TME) of PC is characterized by a dense accumulation of immune cells that suppresses the host immune system. The TME significantly influences cancer progression, metastatic potential, and resistance to therapeutic interventions, which is a concerning aspect of PC. The microenvironment not only contributes to initial resistance to treatments but also to later acquired resistance, extending beyond traditional cytotoxic chemotherapies to include targeted therapies and immunomodulatory interventions (6). Furthermore, the TME’s dense stroma impedes immune cell infiltration and drug penetration [2]. Although chemotherapy has been previously employed, progress has been limited [3]. Consequently, immunotherapy has emerged as the fourth pillar of PC treatment [4]. The exploitation of the host immune system or the introduction of modified immune components into the host a potentially effective therapeutic approach for PC treatment with minimal or no side effects [5]. Currently, several strategies are employed to target the PC immune system: the use of monoclonal antibodies (mAbs) against the tumor antigens, targeting immune components [e.g., tumor-associated macrophages, tumor-associated neutrophils, pancreatic stellate cells (PSCs), tumor-infiltrating leukocytes, and cancer-associated fibroblasts (CAFs)], as well as blocking immune checkpoints, such as programmed cell death protein -1 (PD-1), programmed death-ligand 1 (PD-L1) and CD40 [6,7]. Rather than relying on a single agent, various immunotherapy approaches are utilized to combat PC. For instance, the combination of immunotherapy with chemotherapeutic agents has shown promising results, with most of these combination therapies currently in phase I and phase II clinical trials. Immunotherapy is regarded as more efficacious when combined with chemotherapy rather than with radiotherapy [8]. Although trials of immunotherapies have commenced recently, their effectiveness against PC is constrained by the TME. Adverse effects have been observed, attributed to the inflammation induced by immunotherapeutic agents on the host and the pre-existing immune cell population [9,10].
The immune system plays a crucial role in safeguarding the body against various pathogens and substances. However, translating fundamental immunological knowledge from animal models to humans has proven to be a significant challenge, often resulting in more setbacks than successes [11]. To enhance the understanding of the human immune system, immunologists are diligently employing diverse methodologies and developing novel therapeutic strategies. The fundamental components of the immune system comprise various immune cell types and cytokines, which facilitate intercellular communication. Fortunately, the majority of these components can be accessed using current technological advancements, and a representation of these elements is obtainable from a blood sample, which is readily accessible to researchers.
Over the past decade [12], various immunotherapeutic agents have been employed in the clinical management of tumors. These agents include diverse immune checkpoint blockers (ICBs) that target cytotoxic T lymphocyte–associated protein 4 (CTLA4), PD-1, or its primary ligand PD-L1. They are analogous to homologous T cells engineered to interact with CD19 through the targeting the chimeric antigen receptor (CAR) [13]. Ongoing clinical trials are investigating additional immunotherapies that demonstrate anticancer efficacy. These encompass numerous immunostimulatory mAbs and molecular fragments that counteract cancer-induced immunosuppression, similar to therapeutic vaccines targeting various cancers [14].
Stage I PC patients exhibit a survival rate of ~29%, whereas the overall 5-year survival rate remains at a mere 8% [15]. Relapses are inevitable and the therapeutic options for PC are limited, encompassing radiation, chemotherapy, and surgical interventions. Furthermore, a significant mortality rate is attributed to the delayed diagnosis of the disease. Recent advancements in immunotherapy have transformed cancer treatment paradigms, particularly for melanoma [16]. The interaction between the immune system and cancer cells occurs through the stages of elimination, equilibrium, and escape. During the elimination phase, the immune system identifies and eradicates altered cells [17]. As transformed cells transition from the elimination phase to the equilibrium phase, they undergo genomic alterations and establish a TME conducive to the development of early lesions. In the escape phase, cancer cells select immunosuppressive cells, including myeloid-derived suppressor cells (MDSCs), regulatory CD4+FOXP3+ T cells (Treg cells), and cancer-associated macrophages [18]. This review incorporates the latest research on various immunotherapies and examines the impact of TME on PC.

2. Tumor Microenvironment

The enhanced understanding of the critical characteristics of the TME in eliciting immune responses against cancer has significantly advanced the field of immuno-oncology [19,20]. The concept of “immune context” has been introduced and validated in light of these findings, leading to the classification of tumors into four categories: cold, immunosuppressed, excluded, and heated [21,22]. It has been suggested that “hot” tumors may respond effectively to immune checkpoint inhibitors (ICIs) if they possess growth-promoting lymphocytes, genomic susceptibility, and a prior antitumor immune response, as indicated by complementary determinations such as the expression of anti-PD-L1 on cancer-associated immune cells [23]. Cold tumors do not easily provoke an immune response and are frequently resistant to treatment [24]. Conversely, “cold” tumors have been identified as immunologically “ignorant,” characterized by insufficient invasion, increased proliferation, minimal expression of antigen presentation machinery, including major histocompatibility complex I (MHC I), and limited mutational perturbations (minimal expression of neoantigens) [25]. The transition from “cold” tumors to generative “hot” cancers that are susceptible to ICIs is a dynamic area of research. In this context, targeting the interaction between cancer and T cells may enhance resistance in an inhospitable microenvironment and contribute to the efficacy of immunotherapies [26]. Radiation and chemotherapy have been employed to potentially convert “cold” tumors into “hot” ones by increasing the antigenicity and priming ability of malignancies. Ionizing radiation-induced antigen release and immunogenic cell death may transform tumor cells into an in situ vaccine [27]. There is evidence that ICIs directly influence the metabolic landscape in the TME, thereby affecting the function of effector T cells (Figure 1). Increasing evidence suggests that the TME sustains inappropriate metabolic reprogramming, which impairs T-cell activity and diminishes the immunological response to tumors [28,29].
PC is characterized by a well-documented stromal response, which encompasses a diverse array of cells, including PSCs, immune cells, and other components that secrete growth factors [30]. Key stromal cell types implicated in cancer progression include PSCs, CAFs, regulatory T cells, and MDSCs. The mononuclear phagocyte within the bone marrow is capable of generating macrophages, a crucial subset of innate immune cells that contribute to PC immunosuppression and desmoplasia. Environmental signals, such as inflammatory cytokines like IL-8, IL-6, IL-1β, and IL-10, may activate TAMs [31]. Furthermore, a high infiltration of TAMs has been associated with tumor progression, prognosis, and patient survival in PC [32].
Metastasis is the primary factor contributing to the high mortality rate among patients with PC, with mechanisms of metastasis including hematogenous spread, local invasion, and lymphatic dissemination. The majority of patients diagnosed with pancreatic malignancy for the first time present with locally advanced and unresectable stage. It has been documented that most patients succumb to liver, lung, or peritoneal metastases [33]. The immune cells within the body, such as natural killer (NK) cells and CD8+ T cells, have the capacity to counteract or even eliminate malignant cells during metastasis, even at the early stage of epithelial–mesenchymal transition (EMT) [34]. Nevertheless, PC cells can evade immune surveillance and metastasize to distant sites due to the modulation of immunosuppressive components within the TME [35]. Although previous research has focused on the intrinsic properties of cancer cells, recent clinical investigations have identified the cancer stroma within the TME as playing a significant role in the pathogenesis of PC, potentially maintaining the malignant phenotype and promoting metastasis. Numerous studies have explored the mechanisms of PC metastasis, and recent findings indicate that key particles and mechanisms within the immunologic microenvironment are closely associated with pancreatic disease metastasis [36].
TME is crucial in the emergence of resistance to various treatments, including immune checkpoint blockage therapies (ICT) [37]. Patients receiving immune checkpoint inhibitor (ICIs) frequently show signs of immunological memory commonly [38]. Although initial ICIs treatments can yield improved outcomes in the early stages, resistance often arises when ICIs are used alone [39]. A variety of resistance mechanisms have been discovered [40].

2.1. The Role of Pancreatic Cancer TME in Metastasis

PC-TME is distinguished by a structure that includes PSCs, which are vital in metastasis, budding tumor cells, and a partial EMT state [41]. Tumor budding is indicative of a partial EMT state, as evidenced by features such as reduced expression of E-cadherin, loss of β-catenin expression, and increased expression of EMT markers, including zinc finger E-box-binding homeobox proteins 1/2 (ZEB-1/2), SNAIL, and N-cadherin [42,43]. The absence of both proliferative and apoptotic markers further confirms the presence of an EMT subtype, suggesting that proliferation is incompatible with EMT. Moreover, tumor budding has been linked to the dysregulation of the A-200 family miRNA, which is involved in tumor suppression mechanisms related to cell transformation, invasion, migration, and tumor growth and metastasis [44]. Hypoxia-inducible factor (HIF-1) regulates ATP utilization, reactive oxygen species (ROS) formation, and glucose redirection for energy production, thereby metabolically reprogramming the TME to facilitate disease progression [45]. TAMs influence metastasis by secreting macrophage-induced protein-3α (MIP-3α), which modifies the extracellular matrix (ECM) structure [35]. Hypoxia is also associated with the overexpression of heme oxygenase-1 (HO-1) in PC [46]. Recent findings suggest that inhibiting HO-1 may enhance the efficacy of chemotherapeutic agents such as gemcitabine and paclitaxel [47]. Furthermore, the development of an immunosuppressive TME and the lack of interaction between immune cells and pancreatic liver metastatic cells have been observed, promoting liver metastasis [48]. This phenomenon supports the establishment of a pro-TME during the metastatic process, involving CAFs RGS5+, neutrophils S100A8+, FOXP3+ regulatory T cells, and CCL18+ lipid-associated macrophages [48].

2.2. Lymphatic Metastasis

Metastatic hematopoiesis is frequently observed in PC, akin to other solid tumors. Acute surgical patients often exhibit inherent peripancreatic invasion, particularly into the post-pancreatic and extra-pancreatic networks. A recent study identifies lymph node metastasis as a predictive factor for pancreatic ductal adenocarcinoma (PDAC) [49]. Research by McKay et al. demonstrates that lymphatic metastasis significantly impacts survival following complete resection of pancreatic head cancer (PHC). Patients without lymphatic metastases show markedly higher 5-year survival rates compared to those with such metastases [50]. The invasion of lymphatics and subsequent transition to lymph nodes are critical events in the progression of PC. Although lymphatic invasion and lymph node metastasis do not directly increase morbidity in patients with PDAC, they are significant indicators of the disease’s metastatic potential [51]. Furthermore, PC cells derived from patients exhibit an enhanced ability to metastasize to lymph nodes in mouse models [52]. In clinical practice, lymph node status is utilized to predict survival, determine optimal treatment strategies, and monitor disease progression. PDAC carcinomas are often characterized by hypovascularity, with only a few lymphatic and blood vessels interspersed among the tumor cells [53].

2.3. Biomarkers for TME

The most prevalent dense structures within the TME are CAFs. CAFs typically constitute all fibrotic tissues that form the TME [54]. These fibroblasts primarily originate from pre-existing PSCs and represent potential therapeutic targets in PC treatment. The presence of glial fibrillary acidic protein (GFAP), desmin, vitamin A lipid droplets, and acetylcholine receptors signifies the existence of PSCs [55]. Given the intrinsic heterogeneity of CAFs, biomarkers are not exclusive to a single cell subtype. Nevertheless, specific biomarkers have been identified for CAF detection, including α-SMA, vimentin, fibroblast activation protein (FAP), platelet-derived growth factor receptor α/β (PDGFR α/β), and podoplanin (PDPN/gp38) [56,57]. Extracellular signal-regulated kinases (ERK) are highly expressed in PSCs, facilitating metastasis and interaction with the cancer stroma within the PC milieu [58]. Inhibition of ERK1/2 specifically results in diminished EMT processes, increased markers of cellular senescence, and activation of autophagy in PSCs of PC [58]. Leukemia inhibitory factor (LIF) serves as a more effective biomarker compared to carcinogenic embryonic antigen (CEA) and carbohydrate antigen 19-9 (CA 199), with case studies indicating high LIF expression in patients with lymph node metastases, correlating with decreased overall survival and recurrence-free survival [58,59]. Elevated expression of CD10+ PSCs is indicative of lymph node metastasis. Furthermore, vascular endothelial growth factor (VEGF) subtypes C/D are crucial for lymphatic vessel formation and thus represent viable targets [35]. Recently, kinesin family member 5B (KIF5B) and secreted frizzled-related protein (SFRP2) have been identified as components of the TME in PC [60].

3. Characteristics of TME in Pancreatic Cancer

PDAC is characterized by its aggressive nature. A prominent feature of TME is the dense stroma, which imparts both an aggressive phenotype and immunosuppressive properties. This is influenced by several factors, including CAFs, PSCs, and the hypoxic environment [61,62]. Smooth muscle antigen alpha (SMA-α) and FAP are integral to the wound healing process within the ECM [63]. In addition, the TME comprises predominant cell types, such as certain macrophages, cytotoxic immune cells, regulatory cells, endothelial cells, and PC-specific neuronal cells [64]. The ECM, rich in collagen, proteoglycans, hyaluronic acid, and fibronectin, is abundant within the TME [65]. It also serves as a source of hepatocyte growth factor (HGF) and fibroblast growth factor (FGF) [66].

3.1. Pancreatic Stellate Cells

PSCs represent a subset of cells integral to the dense TME of the PC [67]. These stellate cells, which store vitamins, are located around the periductal regions of the pancreas. In their quiescent stage, PSCs play a role in regulating endocrine and exocrine secretions, facilitating phagocytosis, and maintaining normal pancreatic morphology [68]. Upon activation, PSCs exhibit increased expression of ECM proteins, including laminin, collagen, and fibronectin, which contribute to the fibrotic nature of pancreatic tumor tissue. Furthermore, activated PSCs (aPSCs) facilitate the recruitment of immunosuppressive cells and are implicated in the progression of non-invasive pancreatic intraepithelial neoplasms to invasive PDAC subtypes [69]. The transition from PSC to aPSC is influenced by lifestyle factors (smoking and alcohol consumption), environmental stressors (hypoxia, hypoperfusion, and oxidative stress), which lead to the secretion of cellular factors (IL-1, IL-6, HIF-1α, TGF-β, and CTGF), and specific molecular signals, including Wnt/β-catenin and PI3 pathways [68]. In addition, aPSCs contribute to the recruitment of immunosuppressive cells through the secretion of IL-6 and macrophage colony-stimulating factor (M-CSF) [70]. The expression of autophagy factors is essential for aPSCs, and the presence of the autophagosomal marker microtubule-associated protein-1 (MAP1) in PSCs has been associated with poor patient survival rates [71].

3.2. Cancer-Associated Fibroblasts

CAFs are integral components responsible for the dense desmoplasia characteristic of the TME, contributing to its aggressiveness [44]. These CAFs originate from various mesenchymal cell types, with PSCs serving as the primary source [72]. Predominantly, resident PSCs differentiate into CAFs. The transition of CAFs towards cancer is marked by a reduction in FAPs, the expression of α-SMA, and the acquisition of a contractile morphology [73]. In PCs, CAFs exhibit significant heterogeneity, comprising multiple subtypes derived from PSCs that secrete cytokines IL-6 and α-SMA at low levels [56]. This heterogeneity poses a challenge for therapeutic targeting, as it can lead to epigenetic regulation, induction of stem cells in PC, and metabolic reprogramming [57]. Recent investigations employing single-cell RNA sequencing have elucidated the heterogeneity of stromal components within the TME, with CAFs playing a pivotal role in TME regulation. Cancer-derived substances, such as interleukin-1 (IL-1) and transforming growth factor-beta (TGF-β), have been demonstrated to alter the phenotype of adjacent fibroblasts, inducing their differentiation into inflammatory and myofibroblastic CAFs, respectively. Inflammatory CAFs secrete IL-6 and facilitate tumor growth, whereas myofibroblastic CAFs contribute to the formation of the surrounding stroma. These distinct stromal cell subtypes correlate with specific cancer cell subtypes, reflecting the capacity of cancer cells to shape a microenvironment conducive to their survival [74].

3.3. Tumor-Associated Macrophages

Macrophages, which primarily differentiate from monocytes produced in the central bone marrow, are present in both healthy and inflamed tissues [75]. The metabolism of macrophages plays a crucial role in regulating the progression of PC. The promotion of glycolysis by HIF-1 leads to increased expression of IL-1. To enhance phagocytosis, M1 macrophages exhibit modifications in the tricarboxylic acid (TCA) cycle, increased glycolysis, and oxidative phosphorylation (OXPHOS). Phagocytosis and inflammation are more effectively facilitated by lipid metabolism. The progression of PDAC is affected by impaired glutamine metabolism. These findings underscore the significance of macrophage metabolism in PC [76]. Certain macrophages are specialized, such as Kupffer cells in the liver and splenocytes in the white pulp of the spleen. Circulating macrophages are also present in the TME, aided by various molecules, including chemokines and cytokines, along with high concentrations of lactic acid, local anoxia, and environmental factors [77,78]. Collectively, these cells constitute TAMs. TAMs are classified into M1 and M2 types based on their polarizing properties. Although M1 macrophages are involved in the secretion of pro-inflammatory cytokines, M2 macrophages are associated with the secretion of anti-inflammatory cytokines [79]. A previous study on the blockade of CCR4+ in TAMs demonstrated that TAMs are essential for the infiltration of immune cells into the tumor [80]. An alternate study of examining organ-specific isotypes of PDAC revealed a significant increase in Maf+ macrophages, which is directly associated with CD8+ T cells, thereby promoting an immunosuppressive microenvironment [81].

3.4. Hypoxia

In contrast to other solid tumors, PC is characterized by a substantial presence of stromal cells and an ECM, yet lacks vascularization, resulting in severe and persistent hypoxia within the tumor. This hypoxic microenvironment significantly influences the biological characteristics of PC, including metabolic reprogramming, cancer stem cell dynamics, invasion and metastasis, and pathological angiogenesis, all of which collectively contribute to disease progression and therapeutic resistance [62]. Consequently, hypoxia is a critical aspect of the pancreatic TME due to the dense stroma that impedes the infiltration of immune cells and other therapeutic agents [45]. A recent study employing a xenograft model demonstrated that under hypoxic conditions, HIF-1α induces LncRNA-BX111, which subsequently leads to the transcription of ZEB-1, an EMT inducer [82]. Inhibition of BX111 results in the suppression of tumor growth and metastasis [82]. In addition, the hypoxic environment facilitates pH balance and glycolysis regulation by enhancing the expression of activated KRAS, which in turn leads to the overexpression of carbonic anhydrase 9 (CA9) through modulation of HIF-1α and HIF-2α [83]. Recent studies have shown that specific molecular and cellular alterations occur in PC under hypoxic conditions. Additionally, hypoxia downregulates lncRNA-CF129, which plays a pivotal role in preventing invasion in a FOXC2-dependent manner via the ubiquitination and degradation of p53 [84,85,86,87]. CF129 is transcriptionally downregulated by hypoxic factors, such as the HIF-1α/ histone deacetylase 1 (HDAC1) complex, thereby contributing to disease progression [88]. Activation of the phosphatidylinositol-3-kinase (PI3K)-mTOR pathway under hypoxic conditions inhibits NK cells from eliminating tumor cells. Moreover, by upregulating the expression of matrix metalloproteinase10 (MMP10), hypoxia diminishes the expression of the tumor cell surface recognition molecule MICA, which downregulates the expression of NK and group 2D NK cells (NKG2D) on T cells, facilitating immune evasion from tumor cells [89,90]. Various cellular components contribute to the distinct properties of the TME. In addition, increased production of vascular endothelial growth factor (VEGF) under hypoxic conditions promotes angiogenesis by inducing endothelial cell proliferation and the formation of new blood vessels [91,92,93,94].

3.5. Lymphangiogenesis and Lymphatic Metastasis

One of the most critical components of PC metastasis is the TME. The disease disseminates either through local metastasis or lymph node metastasis [95]. Lymphatic metastasis, facilitated by lymphangiogenesis, represents a successful mechanism by which cancer metastasizes to both distant and local areas. A high density of immune cells, including regulatory T cells (Tregs), immature and tolerogenic dendritic cells (DCs), cytokine producers, and MDSCs, in the lymph nodes at tumor drainage sites, is associated with lymph node metastasis. M2-type TAMs have been identified as playing a crucial role in the regulation of lymphangiogenesis [96]. Furthermore, PSCs and PC cells initiate angiogenesis by secreting pro-angiogenic factors, leading to the formation of microcapillaries [93,97,98,99]. Extracellular vesicles (EVs) have been extensively reported to promote cancer progression and structural alterations in the TME, as well as metastasis to adjacent organs [100].

3.6. Functional Diversity of Tumor Lymphangiogenesis

The extent to which lymphangiogenesis contributes to the TME remains uncertain, despite its role in promoting tumor dissemination in both primary tumors and empty lymph nodes (LNs). Recent research employing phylogenetic reconstruction techniques has revealed that the majority of colorectal cancer metastases to distant organs bypass the LN [101]. Historically, lymphatic metastasis was considered a passive process. However, emerging evidence indicates that lymphatic endothelial cells (LECs) actively participate in the invasion of cancer cells into lymphatic vessels (LVs) and their subsequent infiltration into LNs [51]. One study showed that LEC extensively form filopodia in response to VEGF-C, which facilitates the shredding of tumor cells into the lymphatic vasculature [102]. Notably, LECs promote the migration of tumor cells into the lymphatics, with chemokines playing a contributory role. For instance, CXC-motif chemokine 12 (CXCL12)-expressing LVs have been shown to enhance the invasiveness of various cancer cell types expressing CXCR4. Similarly, the production of secondary lymphoid-tissue chemokine (CCL21) by LECs has been implicated in the relocation of CCR7+ malignant cells to a niche within the LV during tumor progression [103,104,105,106]. In PDAC, CD3+ markers (90%) are indicative of tumor-infiltrating lymphocytes (TILs) [88], predominantly of the CD4+ subtype. Most TILs are CD45RO+ and CCR7, suggesting that these TILs are effector cells with known antigenic determinants. In addition to TILs, CD8+ cells express CD28 and, to a lesser extent, Glucocorticoid-induced TNFR-related protein (GITR), a receptor that supports CD8+ proliferation [107]. TILs in PC can enhance therapeutic responses by recognizing and capturing tumor-associated antigens [108]. While TILs may also include B cells, these typically remain inactive within the tumor [109].

3.7. T Lymphocytes

T cells are systematically categorized based on their T cell receptor (TCR) subunits, similar to central lineage markers. T cells within the classification are capable of recognizing proteins presented on the cell surface via MHC I or II (CD8 or CD4 T cells, respectively) due to the αβ TCR complex [110]. To date, the majority of research has concentrated on CD4low CD8high T cells, a subset believed to originate from peripheral CD8 T cells that co-express low levels of CD4 following activation [111].
Owing to their well-documented antiviral and anticancer properties, CD8+ T cells are also referred to as cytotoxic T lymphocytes (CTLs). CTLs are capable of releasing substantial quantities of antitumor cytokines and cytotoxic particles, such as TNFα and granzymes [110]. The memory T cell subsets also exhibit a range of divisions, from cells with a more naïve phenotype to those with an effector-like phenotype. These cells typically align with the lineage of effector memory T (TEM), stem cell-like memory T, and effector memory RA+ T cells [112]. Tissue-resident memory T cells (TRM) in the surrounding tissues interact with canonical markers such as CD103, also called integrin αE, CXCR6, CD49a, and CD69 [113,114,115].

3.8. B Lymphocytes

In the realm of clinical immunotherapy, patient stratification and response evaluation have predominantly concentrated on T-cell responses. Recent research, however, has recognized the significance of B lymphocytes in immunotherapy, linking their quality to improved prognoses in various cancers, including melanoma, breast cancer, renal cell carcinoma, colorectal cancer, hepatocellular carcinoma, and squamous cell carcinoma of the head and neck [116,117,118,119]. Within the TME, B lymphocytes can exhibit either pro-tumor or antitumor characteristics [100]. In ectopic lymph nodes, such as tertiary lymphoid structures (TLS), tumor-infiltrating B cells differentiate into memory B cells and IgG1-producing plasma cells with the assistance of follicular Tfh cells. The IgG1 and IgE antibodies produced by memory and plasma B cells in response to tumor-associated antigens activate the complement system, phagocytosis, or antibody-dependent cytotoxicity of NK cells or macrophages [120,121]. A defining feature of proficient antigen-presenting cells (APCs) is their ability to internalize antigens and present the processed antigen on MHC class molecules to T lymphocytes [110]. Although B cells possess the capability to function as APCs, their efficacy appears inferior to that of DCs, likely due to their less effective nonspecific antigen uptake. B cells exhibit heightened sensitivity to antigens at lower concentrations compared to DCs, attributed to their typically high-binding affinity (multivalence) upon antigen encounter [122]. Prior to immunization, antigen-specific B cells are exceedingly rare in comparison to DCs, leading to the perception that B cells contribute only modestly to APC-mediated activation of naïve CD4+ T cells [123]. Nevertheless, utilizing virus-like particles identified by RNA phage Qβ as a model for nanoparticle antigens, Hong et al. [124] showed that B cells, rather than DCs, are responsible for the initial activation of CD4+ T cells and the subsequent differentiation of CD4+ T cells into CD4+ T follicular helper (TFH) cells. Furthermore, this model can induce a germinal center reaction in the absence of DCs [43].

4. Immune System and Immunobiology of Pancreatic Cancer

The immune system of PC primarily relies on the components of innate immune cells, including macrophages, myeloid cells, neutrophils, and T lymphocytes. Myeloid cells are present as MDSCs, which exist in two forms: polymorphonuclear MDSCs (PMN-MDSCs), phenotypically similar to neutrophils, and monocytic MDSCs (m-MDSCs), resembling monocytes (Figure 2) [125]. Their primary function is the suppression of the immune system through cysteine detachment mechanisms and the upregulation of arginase expression with concurrent downregulation of L-arginine, both essential for T-cell activation and T-cell protein synthesis, respectively [77]. Macrophages, in the form of TAMs, constitute another significant component of the immune system. The TME of PC induces primary macrophage M1 (pro-inflammatory) to differentiate into M2 (anti-inflammatory) subtypes. Tregs are abundant in the PC immune system and exert a suppressive role by secreting factors such as IL-10 and TGF-β and expressing CTLA-4 [126]. Typically, the immune environment of PC is characterized by reduced numbers of DCs, NK cells, and MHC class 1 molecules, which serve as the initial defense against cancer. The suppressive nature of the tumor results in a diminished presence of these cells [127]. NK cells, as innate immune cells, express CD56+ receptors and lack T-cell receptors. They are recognized for targeting virus-infected tumor cells. This anticancer activity of NK cells occurs independently of stimulation but it is facilitated by predominantly present receptors, such as CD16, NKG2D, DNAM1 (DNAX accessory molecule), and NK cell receptors [128]. NK cells can be activated by cytokines such as IL-2, IL-12, and IL-15 [129]. Furthermore, the tumor contains neutrophils, a type of granulocyte. PC can recruit neutrophil granulocytes into the solid tumor, but rather than eliciting an antitumor response, the neutrophils promote disease progression [80]. The neutrophil subtypes N1 and N2, referred to as tumor-associated neutrophils (TANs), are present in the early stages of PC and are known to regulate tumor development through various mechanisms, including proinflammatory cytokine release, angiogenesis, and invasion [77]. In a case study involving 112 patients with PDAC, 25 of whom were long-term survivors, the majority of components were identified as CAFs in a quiescent state. In addition, the long-term survivors exhibited increased levels of CD3+, CD4+, and iNOS+ cells (M1 macrophages and neutrophils) and a decrease in the expression of Tregs, CD68+ macrophages, and Foxp3 variants [130].
Several secretory factors such as cytokines (IL-6, IL-10 and IL-11, IL-12, GM-CSF and TNF-α), chemokines CXCL-8, CXCL-10, CXCL12 and CCL-3 and growth factors VEGF and TGF-β are important roles in tumor cell invasion and infiltration [131,132]. KRAS mutated PC caused several downstream signaling molecules to be activated and further inflammatory cytokines IL-6, IL-10 and chemokines CXCL2 were activated [133]. In addition, p53 mutation activates the immune evasion in PC [134].

5. Immunotherapy

The characteristic features of PC include an immunosuppressive milieu and a dense stroma, which serves both as a physical barrier to drug penetration and as a dynamic component in the regulation of the immune system. Consequently, the immune system is considered integral to the development of PC [135]. In cancer immunotherapy, specific components are administered to either stimulate the immune system to target a tumor or direct these immune components against the tumor, thereby exerting anticancer effects. The first successful immunotherapy was y carried out in 1981 using streptococcal organisms against bone and soft tissue tumors [136]. This was followed in 1984 by immunotherapy against metastatic melanoma in a 33-year-old woman, who underwent several treatments before receiving a recombinant IL2 (rIL2) infusion. After two months, the tumor size ceased to decrease [137,138]. Immunotherapy has now been established as the fourth pillar in the treatment of PC, alongside surgery, chemotherapy, and radiotherapy [139]. Most PC immunotherapies are currently in phase II trials, with a significant number of phase I trials completed (Table 1) [98,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]. These therapies show promise, particularly when used in combination with traditional drugs [156], rather than as standalone treatments. In general, the efficacy of immunotherapy depends on the immunosuppressive environment developed by the tumor, the tumor’s sensitivity to the immunotherapy, and its capacity to trigger an immune response. The generation of immune responses against tumors involves targeting tumor neoantigens. Reversal of immunosuppression had a limited impact on the intratumoral decline in CD8+ and NK cell populations, with PD-1 expression on immune cell surfaces being a significant impact factor in this impact [157]. Key determinants for the efficacy of immunotherapy encompass the tumor’s composition, the localization of infiltrated immune components, and their respective functionality, all of which are essential for the precise targeting of the tumor immune system. In addition, the characteristics of the tumor stroma and blood vessels offer valuable insights into the strategic application of immunotherapeutics against the disease [12].

6. Characteristics of Immunotherapy

Immune cells from both the innate and adaptive immune systems infiltrate the TME and play a crucial role in maintaining the equilibrium of tumor dynamics, underscoring the importance of the immune system in immune surveillance. Immune responses can effectively eliminate threatening cells or diminish their morphologies and capacities [158]. However, cancer cells possess several sophisticated mechanisms, including immunosuppressive cell populations, upregulated negative regulatory pathways, and defects in the antigen presentation machinery [158], which enable them to evade immune surveillance by blocking the effector capacity of immune cells and abrogating antitumor immune responses. Immunotherapy, which aims to bolster natural defenses in eradicating malignant cells, represents a significant advancement in cancer treatment and has transformed the field of oncology. Although the concept of utilizing the host’s immune system to eliminate cancerous growths dates back a century [158], substantial progress has been achieved in contemporary scientific and clinical research. Immunotherapy has elicited clinical responses in various malignancies, albeit with low response rates and complex underlying mechanisms [159]. Currently, immunotherapy is a primary focus for certain cancers, as it functions by restoring the patient’s immune responses to combat tumor cells [160,161].

7. Mechanism of Immunotherapy for Pancreatic Cancer

Building upon a well-established immune system, various forms of immunotherapy have been developed over time. These include adoptive cell transfer, oncolytic viruses, immunostimulatory cytokines, and anti-tumor (bispecific) antibodies immunotherapies, such as mAbs. mAbs, which inhibit immunosuppressive signals from immune cells or cancer, are the most widely used in clinical practice today, with numerous FDA approvals for solid tumors. Their anti-tumor effects are further enhanced by ICI treatment [162,163]. These therapies have demonstrated stimulatory and durable effects in certain patients by modulating their immune response [164]. The mechanisms of immunotherapy developed thus far include blocking immune checkpoint proteins, enhancing the efficiency of antigen recognition by immune cells, increasing intra-tumoral T cells, and administering tumor-specific antigens through vaccines to aid the immune system in recognizing tumor cell types [130,165]. Specific active immunotherapies target the adaptive immune system by activating T or B cells against the respective antigens of the PC [166]. mAbs, immune checkpoint-blocking antibodies directed against a specific protein/peptide of the PC, function as specific passive immunotherapies. T cells can prime antigenic targets when administered to patients to elicit an immediate response, thereby circumventing the need for antigen presentation. Recently, mRNAs have also been used as new targets for passive immunotherapy. Nonspecific adoptive immunotherapies, as the term suggests, involve the adoptive transfer of specifically engineered immune cells or immune cells coupled with cytokines or lymphokines to a patient with PC [167]. Recent studies have discussed in detail the potential of targeting DCLK1 to inhibit cancer stem cell self-renewal in tumors [168,169].

7.1. T-Cell Mediated Therapy

Immunotherapy strategies for pancreatic adenocarcinomas, including the reactivation of NK cells, the promotion or reintroduction of DCs, vaccination techniques utilizing DCs, and the reconstruction of the TME with a focus on macrophages, as well as controllers mediated via cancer or stromal cells, have the potential to modify the cancer microenvironment and enhance T-cell immunity [170]. T-cell therapy employing chimeric antigen receptors (CARs) targeting CD19 and B-cell maturation antigen (BCMA) has significantly improved the treatment of hematologic malignancies. However, the transfer of CAR-T cells targeting overexpressed PC antigens such as mesothelin, CD133, and epidermal growth factor receptors (EGFRs) have proven ineffective in patients with PC [171]. The CAR modulator protein comprises an intracellular segment with specific signaling domains, such as CD28 and CD137 (4-1BB), for T-cell activation, and an extracellular portion that recognizes a cell surface protein [172]. It has been established that 72% of PDAC express CD24, a small, mucin-like, heavily glycosylated, glycosylphosphatidylinositol-anchored cell surface protein associated with higher tumor grades [173], along with CD44 and CD133 [174]. In a human PC xenograft model utilizing orthotopic transplantation, the application of CAR-T cells incorporating anti-CD24 single-chain Fv and CD28 extracellular domains, in conjunction with light exposure, IL-2 administration, and cancer elimination, has been demonstrated [170]. It has been shown that the combination of ICI and chemotherapy is more effective than either treatment alone. The administration of folfirinox and PD-L1 significantly increased overall survival rates and elevated the number of CD8+ T cells and PD-1 expression in tumor and immune cells [175]. Another study indicated that the combination of BL-8040 and pembrolizumab with chemotherapy resulted in a disease control rate of 77% [176].
A previous study has suggested that chemotherapy and radiation enhance the responsiveness of cancers to ICB therapy through the release of pathogen associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs), which drive TLR-dependent [177] and stimulator of interferon gene (STING)-dependent DC activation. In particular, the activation of the cyclic GMP-AMP synthase (cGAS)-STING pathway by cytosolic tumor DNA is essential for the formation of antitumor T effector cells and the efficacy of ICB therapy in preclinical cancer models [178]. Current research is exploring preclinical strategies to activate the cGAS-STING-IFN axis as an adjunct to chemotherapy, radiation, and immunotherapy, despite the lack of clinical data [179]. By enhancing MHC-I expression, antigen cross-presentation, and T-cell priming, the tumoricidal effects of chemotherapy and radiation synergize with immunotherapy to augment T-cell receptor (TCR) diversity and improve T effector cell infiltration [180]. The dosage and sequencing of chemotherapy and radiation are likely to significantly impact the antitumor immune response. In preclinical models of ovarian cancer, metronomic dosing of chemotherapy is considered the optimal dosing strategy, as it maintains immune function and enhances the activity of T effector cells [181]. This approach is commonly employed to improve cancer vaccine responses by reducing the abundance and inhibitory capacity of Tregs. Furthermore, hypofractionated irradiation diminishes the induction of the DNA exonuclease Trex1, facilitating the cytoplasmic accumulation of double-stranded DNA, thereby-promoting STING-mediated type I IFN release by cancer cells, which is superior to single-dose treatment regimens in preclinical mouse models concerning the CTLA-4 blockade [182]. In addition, patients receiving a combination of Ipilimumab and GVAX/CRS-207exhibited an expanded T-cell receptor repertoire in their peripheral blood [183]. Tumor-infiltrating cells (TILs) proliferate following the administration of PD-1 blockade, 4-1BB stimulation, and CD8+ induction therapy, characterized by IFN-γ expression [107,184]. Enzymes that degrade amino acids, such as arginase and indolamine 2,3-dioxygenase (IDO), are targeted to disrupt the TME of PC due to their detrimental effects on the tumor. Adenosine-reducing agents (e.g., oleclumab) and antineoplastic drugs (e.g., urelumab, varlilumab, and zanolimumab) influence anti-tumor T-cell responses [185]. The increased density of cytotoxic T cells in both inflamed and non-inflamed tumors indicates that the immune checkpoint inhibition is a promising strategy for enhancing T-cell immunity [186,187].

7.2. Macrophage Programming

Human PDAC is characterized by extensive immune cell infiltration and fibrosis within its microenvironment of. Among the infiltrating cells, macrophages represent a significant component, and their inhibition may offer a promising strategy for reconstituting the cancer microenvironment [170]. In a mouse model of PC driven by mutant KRAS/p53 [doxycycline-inducible Kras G12D and Trp53 R172H under the control of pancreatic transcription factor 1 (Ptf1), Cre recruiter], the depletion of CD11b+ myeloid cells resulted in the expansion of intratumoral CD8+ T cells [188]. The metabolic reprogramming of macrophages can lead to the methylation of the Nqo-1 gene, thereby initiating protumor activity in a pancreatic model [189]. Furthermore, CAFs frequently develop resistance to FLOFIRINOX treatment, and reprogrammed macrophages contribute to reduced PC survival [190].

7.3. Antibody and Immune Checkpoint Inhibitors

Innate anti-tumor immunity can be enhanced through the application of tumor-targeted mAbs. Tumor cells often evade immune surveillance by activating immune checkpoint pathways, which suppresses antitumor immune responses and allow for unchecked tumor progression. A notable advancement in cancer immunotherapy involves the use of FDA-approved mAbs that inhibit immunological checkpoints (ICIs), thereby augmenting antitumor immune responses to eliminate tumor cells [191,192]. mAbs utilized in PC immunotherapy include ipilimumab, nivolumab, tremelimumab, atezolizumab, and pembrolizumab, all of which function as ICIs [16,193]. In addition, the CD40 antibody, expressed in most hematopoietic and non-hematopoietic cells and tissues, is recognized for its role in regulating immunity [194]. A previous study has shown that CD40 signaling and activation lead to the generation of cytotoxic T lymphocytes [195]. Ipilimumab and tremelimumab specifically target Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The efficacy of single-agent therapies is generally lower compared to combined checkpoint inhibitor therapies [196]. The interaction between PD-1 and CD80 is disrupted through ICP therapy. Mesothelin (MSLN) is a protein ubiquitously expressed various cancer types. MUC16 serves as the ligand of MSLN, and mAbs targeting MSLN, and mAbs targeting MSLN can inhibit the binding of MSLN to mucin 16 (MUC16), thereby impeding its function. This also facilitates antibody-dependent cytotoxicity (ADCC) [88,197] (Figure 3).
The modulation of ICI receptors and co-inhibitory molecules on T cells is crucial for maintaining immunological homeostasis and regulating the functions of effector and regulatory cells [198]. Achieving a balance between immune effector and immunosuppressive cells within the TME is essential for effective cancer immunity. The immune response is regulated by the interactions between immune cell receptors and costimulatory ligands. The CD28 family also includes the T cell Ig and immunoreceptor tyrosine-based inhibition motif (ITIM) domain (TIGIT), a co-inhibitory particle that interacts with T cells and NK cells [199]. Although extensive clinical trials have been conducted using ICIs to treat PC, their overall effectiveness has not been satisfactory [200,201]. The immunosuppressive microenvironment of PC contributes to the limited efficacy of ICIs targeting PD-1/PD-L1 [202]. The PC microenvironment is characterized by a dense interstitial matrix, which acts as a physical barrier to T and NK cells, enabling PC cells to evade host immune surveillance. This highly repressive microenvironment not only facilitates immune invasion by tumor cells but also hinders drug penetration and their anti-tumor efficacy. PC is inherently less immunogenic [203,204,205]. As a result, numerous studies have sought to enhance the immunogenicity of PC to improve the effectiveness of immunotherapy [206].

7.4. Vaccines

A promising approach to hindering the progression of PC involves the development of cancer vaccines utilizing antigen-specific T cells, which are introduced into the TME [207]. T cells are capable of recognizing antigens when presented by APCs that degrade an endogenous or exogenous mutant peptide produced by the tumor, followed by its processing and presentation via the MHC complex on the cell. DCs serve as APCs involved in the processing and presentation of antigens to CD8+ T cells, which are recognized by the αβ T-cell receptor (TCR) [208]. The significance of DCs as a PC vaccination has been emphasized by numerous studies. The study by Jang et al. highlights the importance of DCs in PC regression [209], demonstrating that the suppression of Treg cells (which interact with and impair the function of DCs) enhances the anti-tumor activity of CD8+ T cells by restoring the immunogenic function of CD11+ DCs, thereby inhibiting tumor growth. Due to various limitations associated with the use of DCs as vaccines, mature DCs have been developed in vitro and employed as cancer vaccines because they can effectively produce tumor-specific antigens for recognition by CD4+ and CD8+ T cells. To enhance the specificity of DCs and their efficiency in antigen presentation, different cell components or whole cells have been fused with DCs. The fusion of DCs with tumor lysates has been shown to induce a generalized anti-tumoral T-cell response in patients with PC, thereby extending overall survival by 56 months in one patient and achieving a 1-year survival in 5 out of 12 patients [62]. DCs have also been loaded with tumor antigens, such as exosomes [84], and fused with cancer cells to form hybrids [85]. In addition, DCs have been utilized to target various components of the TME. Most clinical trials have been conducted with MUC1- and WT1-targeted DCs, primarily in phase I/II trials [210,211]. MUC1 was used either as a peptide or as cDNA with DCs. Either MHC I or MHC II or both classes, restricted WT1 in combination with DCs for therapies (Figure 4). The results indicated that patients were able to respond to treatment with minimal or very few side effects [86]. Based on several characteristics of WT1, such as its therapeutic efficacy, specificity, immunogenic activity, role in tumor progression, the presence of a high number of positive WT1 antigens, and expression of WT1 by cells, the National Cancer Institute (NCI) has ranked this antigen as the top antigen for use as a vaccine [87]. Another target, hTERT/DC mRNA, when administered for 3 years to patients who could not receive chemotherapy, resulted in no active disease and an effective immune response [91].
Carcinoembryonic antigen (CEA) is a glycosylated protein present in 90% of PCs and represents a potential therapeutic target [92]. KRAS mutations, which are expressed in over 99% of PCs, pose a significant challenge for therapeutic intervention [93]. Vaccination targeting with Kras has shown a 9-year memory response post-vaccination and a 10-year survival rate with an immune response following surgical resection [97]. Beyond antigenic components used in vaccines, whole pancreatic tumor cells are also employed to elicit an immune response against cancer. In a study involving 93 patients, GVAX was administered with cyclophosphamide in combination with CRS-207, with or without nivolumab. Although no significant results were observed, the outcomes were comparable to those of standard therapy. Notably, patients treated with arm A (Cy/GVAX+CRS-207+ nivolumab) exhibited a superior immune response, characterized by increased CD8+ T cells and decreased CD68+ myeloid cells [98].
GVAX, in conjunction with ipilimumab, facilitates the differentiation of T cells into effector variants and enhances the presence of M1 macrophages within the tumor, although the anticipated overall survival was not achieved [58]. Another investigation explored the combination of GVAX and cyclophosphamide with a chemotherapeutic agent selected by the physician in various combinations. No improvement in overall survival was observed, but side effects such as chills, pyrexia, fatigue, and nausea were reported [103]. An alternative approach to inducing an immune response involves chemotherapeutic agents that induce tumor cell death by releasing cellular components, thereby activating DCs and enhancing the processing and presentation of relevant tumor-associated antigens (TAA) [105]. Patients with advanced PC do not benefit from the telomerase peptide vaccine GV1001, the Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF)-transfected pancreatic tumor vaccine (GVAX), or an allogeneic whole-cell vaccine derived from a PDAC cell line genetically engineered to express GM-CSF [212]. Pre-vaccination before immune checkpoint therapy enhances the effect of the infiltrating immune checkpoints, thereby improving vaccine efficacy. In addition, immune checkpoint therapies have been combined with DC vaccines to augment the antigen-presenting efficiency of cytotoxic T cells. The limited number of neoantigens in tumors presents a challenge in the development of cancer vaccines [104]. Most cancer vaccines remain in clinical trials, as further understanding is required regarding the interplay between host and tumor immune systems, the TME, and administered therapeutics.

7.5. Cytokine Therapies

The TME can be profoundly affected by the targeted delivery of cytokines [213]. Due to their short half-life, cytokines function in an autocrine or paracrine manner, necessitating their concentration at specific sites. However, several cytokine-based therapies employed in clinical practice, such as IL-2 and IFN, are administered systemically, often resulting in serious dose-limiting toxicities [214]. The challenges in directing cytokines to the appropriate site and the incomplete understanding of the effects of specific cytokines across various cancer types constrain the effectiveness of cytokine-based therapies [215]. GM-CSF is instrumental in selecting and activating antigen-presenting cells that process and present cancer-associated antigens, thereby eliciting an effector T-cell response. Talimogene laherparepvec (T-VEC) is a modified herpes simplex virus type 1 (HSV-1) that encodes the gene for human GM-CSF [216]. The intratumoral administration of this engineered HSV prompts cancer cells to produce extended levels of GM-CSF locally, which attracts antigen-presenting cells through chemotaxis, initiating a local adaptive immune response to tumor antigens capable of cancer distant from the infusion site. In animal models, this approach confers protection against reinfection with the same tumor [217].

7.6. Novel Checkpoint Blockade Targets

T-cell immunoglobulin 3 (TIM3), lymphocyte enactment gene 3 (LAG3), B7-H3, the V-domain immunoglobulin suppressor of T-cell activation (VISTA), and T-cell immunoreceptors with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) are among the recently identified negative regulators of T-cell activation currently under investigation [218,219,220]. Two well-established mechanisms that mediate T-cell immune checkpoint suppression are the PD-1/PD-L1 and CTLA-4/B7 signaling pathways. Beyond these two pathways, the interaction between tumor cells and host immune cells involves numerous immunomodulatory receptor-ligand interactions, which can be targeted clinically through either monotherapy or combination therapy [221]. LAG3, an inhibitory ligand present on activated T cells and Treg cells, inhibits CD4 binding sites on MHC class II proteins, thereby attenuating T-cell activation. It induces cell cycle arrest, preventing excessive expansion of the T cell compartment [222]. TIM3 is another negative controller of T-cell responses, likely modulating apoptosis following galectin 9 binding by restricting cell cycle progression, akin to LAG3 [223]. The upregulation of TIM3 may contribute to resistance against anti-PD1 treatments, suggesting that combination therapy could enhance the efficacy of anti-PD1 treatments. Moreover, TIM3 expression is associated with poor prognosis in follicular lymphoma and non-small cell lung cancer, indicating its role in the cancer progression (201). B7-H3 is another target that negatively regulates T-cell responses and is highly expressed in various cancers, including non-small cell lung, pancreatic, prostate, colorectal, and ovarian cancers [219,220,224]. Due to the presence of two immunoreceptor tyrosine-based inhibitory motifs in its intracellular domain, TIGIT suppresses T-cell hyperactivation and is being explored as a potential checkpoint target. TIGIT is more strongly expressed in TILs than in peripheral cells, making it a promising target due to its higher specificity compared to other checkpoint molecules [220].

8. Suppression of Immunity

Through various mechanisms, cancer cells cause both local and systemic immunosuppression, which can significantly impact the efficacy of immunotherapy in clinical settings. The production of surface particles such as PD-L1, which leads to the exhaustion of tumor-infiltrating CD8+ T lymphocytes and NK cells, is a particularly critical factor [225]. In certain instances, such as patients with non-small lung cancer (NSCLC) undergoing treatment with ICBs, the expression of PD-L1 in malignant cells serves as a potent prognostic marker [226]. The plasma membrane protein 5′-nucleotidase ecto (NT5E; also known as CD73) plays a role in the conversion of extracellular ATP—an agent with strong chemotactic properties that targets DCs—to adenosine, contributing to the accumulation of malignant cells in an immunosuppressive microenvironment. CD73 also modulates the balance between immunostimulatory adrenergic signaling and its immunosuppressive adenosinergic counterpart by inhibiting resistance responses through various mechanisms [226,227]. When adenosine activates the A2A adenosine receptor (A2AR) receptor on effector T cells, it suppresses their activation and proliferation, leading to T-cell anergy [228,229]. In addition, A2AR activation impairs the maturation, activation, and cytotoxic functions of NK cells [230,231]. Additionally, naive CD4+ T cells activated by A2AR are more likely to differentiate into regulatory T cells (Treg) expressing the markers Foxp3+ and LAG-3+ [232,233]. Furthermore, through the A2BR receptor, adenosine produced by CD73 enhances the immunosuppressive effect and promotes the infiltration of MDSCs in tumors [234,235]. Regulatory B cells (B reg) are integral to the suppression of immune responses across a range of diseases [236,237]. Targeting B cells may reduce the burden of PC [238]. Induced B reg cells have natural killer function against tumor cells [239,240]. Recently CD38+ B cells reduced the antitumor immunoreactivity in PC [241]. Overall, CD73 attenuates the immune response to tumors by diminishing the cytotoxic activity of effector cells and enhancing the function of regulatory immune cells [234]. Consequently, CD73 represents a promising target for cancer immunotherapy (Figure 5). Various immunosuppressive agents in TME and their targets cells mentioned in Table 2.

9. Aptamer-Based Immunotherapy

To expand the applicability of immunotherapy in oncology, preclinical studies consistently indicate that multidimensional strategies employing a combination of various immunomodulatory agents should be considered. The potential for severe autoimmune-like reactions inevitably increases with the administration of multiple immunostimulatory drugs. Consequently, the development of target-specific therapeutic molecules with fewer adverse effects than mAbs is imperative. Oligonucleotide aptamers may address this limitation, as suggested by current preclinical research [261]. Aptamers are single-stranded DNA or RNA oligonucleotides with three-dimensional structures that exhibit high affinity for their targets. They are selected through a process known as Systematic Evolution of Ligands by Exponential Enrichment (SELEX) [262], an exceptional filtration technique that isolates rare particles from a vast, complex library [262]. The concept of aptamer-conjugated oligonucleotide backbone can be applied using DNA and/or RNA. To enhance the stability of aptamers in serum, several modifications can be implemented, such as substituting 20-amino, 20-fluoro, or 20-O-alkyl nucleotides for ribonucleotides. Oligonucleotide-mediated aptamers offer distinct advantages over mAbs alone. As cell-based products, mAbs are associated with higher production costs and a complex administrative burden to achieve good manufacturing practice (GMP), whereas aptamers, being synthetically derived molecules, resulting in lower production costs. Consequently, they offer the potential for less burdensome administrative approvals and most cost-effective alternative reagents for future clinical trials [261]. Aptamers have been employed as ligands in non-invasive diagnostic tests, such as ELISA and other immunoassays commonly used to analyze biomarkers in blood samples. These novel “aptasensors” or aptamer-based assays can be equipped with readout techniques, such as chemiluminescence (CL), electrochemiluminescence (ECL), fluorescence, surface plasmon resonance (SPR), surface-enhanced Raman spectroscopy (SERS), among others, to enhance the detection of existing biomarkers [263,264,265,266]. Numerous innovative therapeutic approaches have been developed to optimize immunotoxicity and can be intensified in combination with immunotherapy. To address this challenge, one strategy involves the targeted delivery of immunotherapy to tumor or immune cells. In this context, aptamer–drug conjugates (ApDCs) are at a promising stage [267]. ApDCs have been utilized to deliver immunomodulatory agents to restrict immune system co-stimulation in the tumor region, inhibit neoantigens in cancer, block exhaustion-inducing immune checkpoints to revive functional immune cells and trigger anti-tumor immunity [267]. The identification of effective molecular targets, such as immune checkpoints and those that stimulate the immune system, presents the opportunity to enhance immunotherapy by focusing on specific molecular sites. Several aptamers have been developed for immunological checkpoints associated with immunosuppression in cancer patients, including T-cell immunoglobulin-3 (TIM-3), cytotoxic T lymphocyte-associated protein 4 (CTLA4), and programmed death receptor I (PD-1) or its ligand, programmed death ligand I (PD-L1) [268,269,270].

10. Immunogenicity

PC is characterized by low immunogenicity, indicating that it is not readily recognized or targeted by the immune system. Several factors contribute to this low immunogenicity, including TME, immune checkpoint signaling, tumor heterogeneity, stromal barrier, and antigen presentation [261,262,267]. Cancer neoantigens, which arise from somatic nonsynonymous DNA alterations unique to cancer, are often the focus of effective adaptive immune responses against cancer cells [271]. Consequently, an increased mutational load, which enhances the likelihood of neoantigen emergence, has been correlated with heightened sensitivity to ICB-based immunotherapy in various clinical studies [225,272]. In addition, immunogenic chemotherapy and radiotherapy elicit multifaceted immune responses partially through pathogen mimicry. In this context, cancer cells undergoing treatment-induced stress responses release several endogenous molecules that function as adjuvants, including adenosine 5′-triphosphate (ATP), a downstream product of the autophagy process; calreticulin (CALR), a downstream product of endoplasmic reticulum stress; and type I interferon (IFN), a downstream product of Toll-like receptor 3 (TLR3) or cyclic GMP-AMP synthase signaling [273]. In addition, two considerations should be approached with caution as predictive biomarkers for cancer immunotherapy: (i) acute or chronic activation of certain signaling pathways may differentially influence immune responses in cancer, and (ii) the effects of a specific interaction on various types of immunotherapies may exhibit significant heterogeneity [274].

11. Factors That Limit the Efficacy of Immunotherapy

The effectiveness of immunotherapy in PC is limited by the relatively low number of aggregated mutations capable of inducing the expression of non-self-antigens, or “neoantigens,” which are recognized as foreign by the immune system. In comparison to malignancies with lower mutational loads, those with higher mutational burdens present a greater number of neoantigens, facilitating detection by the immune system [275,276]. There are three major limitations to immunotherapy for PC. Firstly, unlike lung cancer and melanoma, PC exhibits a very low mutational burden. Secondly, the immunosuppressive microenvironment, characterized by a dense desmoplastic reaction and a significant influx of tumorigenic macrophages and MDSCs, plays a significant role in the progression of pancreatic malignancy. Thirdly, the PC microenvironment is characterized by a paucity of infiltrating T cells, which hinders the development of adequate T-cell responses [277]. In addition to these factors limiting the efficacy of immunotherapy, other barriers to therapeutic success include the chronic inflammation associated with PC and the T-cell response to administered therapy [278]. When considering treatment with ICP blockade, only 4% of all tumor cells, including PC cells, CAFs, and CD163+ TAMs, express the PD-1 immune checkpoint [203].
In a groundbreaking study aimed at enhancing the efficacy of tumor immunotherapy, researchers focused on inhibiting the interferon gamma (IFN-γ) pathway in tumor cells. This suppression was found to enhance the overall effectiveness of immunotherapy and the body’s ability to identify and eliminate tumor cells through innate immune systems [203]. A recent study in the United States has made significant progress in understanding the role of IFNs in tumor immune responses. These findings suggest that IFNs influence the immune response to malignancies in two ways. Firstly, IFNs activate DCs, which are essential for delivering tumor-specific antigens to CD8+ T lymphocytes, thereby promoting cross-activation. However, prolonged exposure to IFNs can result in a negative feedback loop, leading to T-cell dysfunction and immunosuppression [278].

12. Future Perspectives

Although it is usually challenging to translate findings from preclinical models into clinical practice, the scarcity of new therapeutics being incorporated into the standard treatment for PDAC may particularly underscore the shortcomings of existing preclinical PDAC models. There are more ambitious translational research programs that run alongside clinical trials and enhance the understanding of PDAC immunology, even when clinical endpoints are not achieved. These programmes should ideally lead to “reverse translation” by experimentally establishing concepts that help overcome resistance to immunotherapeutic strategies. However, regarding translational methodology, it becomes evident that there are no established standards for sampling patient material and for evaluating immune responses. In addition, patient cohorts who have undergone novel treatment strategies have frequently received at least one previous line of therapy, involving repeated drug exposure. This feature, which can greatly influence the reaction to experimental treatments, is not replicated by most tumour models. Patient-derived organoid cultures or 3D cultures testing of the drugs/immunotherapeutic molecules may provide the better treatment options in patients with high degree of drug/immune resistance.

13. Conclusions

Recent advances in cancer immunotherapies, which stimulate the immune system to recognize and combat malignant tumors, have expanded the potential for effective treatment of such tumors. In recent years, the range of successful immunotherapies has increased, thereby broadening the therapeutic options available to oncologists. Despite significant progress in malignancy and translational research, PC continues to yield poor outcomes and retains highly lethal malignancy. The efficacy of immunotherapy in PC is currently under investigation in several early-phase clinical trials. These trials are evaluating the impact of adoptive cell transfer, ICIs, and their combinations with chemoradiotherapy or other molecularly targeted treatments. In many of these studies, cancer vaccines have consistently shown non-progressive outcomes. Future research should prioritize strategies to overcome immunotherapy resistance by addressing various immunodeficiencies and employing combined immunotherapy and cytotoxic approaches in patients with PC. At present, our understanding of the etiology of PC is limited, necessitating further comprehensive research. Large-scale studies are essential to elucidate the risk factors associated with PC. This study aims to identify novel diagnostic and therapeutic strategies that may enhance the survival of patients with PC.

Author Contributions

Conceptualization, methodology, S.V.T. and V.J.; Software, B.F.Z., R.G. and A.S.; Validation, formal analysis,; investigation, S.V.T., V.J., L.D., S.A., P.C. and D.G.; Data curation, R.V.; Writing—original draft preparation, S.V.T., V.J., L.D., S.A., P.C. and D.G.; Writing—review and editing, R.V. and S.G.K.; Visualization, K.P.; Supervision, P.C. and S.G.K.; Review analysis and data compilation, K.P. and D.G.; Funding acquisition, R.V. and K.P. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Department of Science and Technology, Science Engineering Research Board (DST-SERB), the Government of India, under grant number EEQ/2017/000567 to KP, as well as Industrial funding from BogaR Laboratories, Hyderabad.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions present in this study are included within the article. Further inquiries can be directed to the corresponding authors.

Acknowledgments

We express gratitude to M. Vijayaraman for his assistance in editing the English language of this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ilic, I.; Ilic, M. International patterns in incidence and mortality trends of pancreatic cancer in the last three decades: A joinpoint regression analysis. World J. Gastroenterol. 2022, 28, 4698–4715. [Google Scholar] [CrossRef]
  2. Lavin, Y.; Kobayashi, S.; Leader, A.; Amir, E.D.; Elefant, N.; Bigenwald, C.; Remark, R.; Sweeney, R.; Becker, C.D.; Levine, J.H.; et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017, 169, 750–765.e17. [Google Scholar] [CrossRef]
  3. Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402. [Google Scholar] [CrossRef]
  4. Mangan, M.S.J.; Olhava, E.J.; Roush, W.R.; Seidel, H.M.; Glick, G.D.; Latz, E. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat. Rev. Drug Discov. 2018, 17, 588–606. [Google Scholar] [CrossRef]
  5. Irvine, D.J.; Dane, E.L. Enhancing cancer immunotherapy with nanomedicine. Nat. Rev. Immunol. 2020, 20, 321–334. [Google Scholar] [CrossRef]
  6. Mhaidly, R.; Mechta-Grigoriou, F. Role of cancer-associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer. Immunol. Rev. 2021, 302, 259–272. [Google Scholar] [CrossRef]
  7. Wu, L.; Zhang, X.H.-F. Tumor-Associated Neutrophils and Macrophages—Heterogenous but Not Chaotic. Front. Immunol. 2020, 11, 553967. [Google Scholar] [CrossRef]
  8. Paduch, R. The role of lymphangiogenesis and angiogenesis in tumor metastasis. Cell. Oncol. 2016, 39, 397–410. [Google Scholar] [CrossRef]
  9. Poma, A.M.; Torregrossa, L.; Bruno, R.; Basolo, F.; Fontanini, G. Hippo pathway affects survival of cancer patients: Extensive analysis of TCGA data and review of literature. Sci. Rep. 2018, 8, 10623. [Google Scholar] [CrossRef]
  10. Ansari, D.; Ohlsson, H.; Althini, C.; Bauden, M.; Zhou, Q.; Hu, D.; Andersson, R. The Hippo Signaling Pathway in Pancreatic Cancer. Anticancer Res. 2019, 39, 3317–3321. [Google Scholar] [CrossRef]
  11. Brodin, P.; Davis, M.M. Human immune system variation. Nat. Rev. Immunol. 2017, 17, 21–29. [Google Scholar] [CrossRef]
  12. Galluzzi, L.; Chan, T.A.; Kroemer, G.; Wolchok, J.D.; López-Soto, A. The hallmarks of successful anticancer immunotherapy. Sci. Transl. Med. 2018, 10, eaat7807. [Google Scholar] [CrossRef]
  13. Sadelain, M. CD19 CAR T Cells. Cell 2017, 171, 1471. [Google Scholar] [CrossRef] [PubMed]
  14. Hoos, A. Development of immuno-oncology drugs—From CTLA4 to PD1 to the next generations. Nat. Rev. Drug Discov. 2016, 15, 235–247. [Google Scholar] [CrossRef]
  15. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2017. CA Cancer J. Clin. 2017, 67, 7–30. [Google Scholar] [CrossRef] [PubMed]
  16. Banerjee, K.; Kumar, S.; Ross, K.A.; Gautam, S.; Poelaert, B.; Nasser, M.W.; Aithal, A.; Bhatia, R.; Wannemuehler, M.J.; Narasimhan, B.; et al. Emerging trends in the immunotherapy of pancreatic cancer. Cancer Lett. 2018, 417, 35–46. [Google Scholar] [CrossRef]
  17. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes. Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef]
  18. Ohue, Y.; Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089. [Google Scholar] [CrossRef]
  19. Pagès, F.; Mlecnik, B.; Marliot, F.; Bindea, G.; Ou, F.-S.; Bifulco, C.; Lugli, A.; Zlobec, I.; Rau, T.T.; Berger, M.D.; et al. International validation of the consensus Immunoscore for the classification of colon cancer: A prognostic and accuracy study. Lancet 2018, 391, 2128–2139. [Google Scholar] [CrossRef] [PubMed]
  20. Bader, J.E.; Voss, K.; Rathmell, J.C. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Mol. Cell 2020, 78, 1019–1033. [Google Scholar] [CrossRef]
  21. Gajewski, T.F.; Corrales, L.; Williams, J.; Horton, B.; Sivan, A.; Spranger, S. Cancer Immunotherapy Targets Based on Understanding the T Cell-Inflamed Versus Non-T Cell-Inflamed Tumor Microenvironment. Adv. Exp. Med. Biol. 2017, 1036, 19–31. [Google Scholar] [CrossRef]
  22. Fridman, W.H.; Zitvogel, L.; Sautes-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
  23. Hegde, P.S.; Karanikas, V.; Evers, S. The Where, the When, and the How of Immune Monitoring for Cancer Immunotherapies in the Era of Checkpoint Inhibition. Clin. Cancer Res. 2016, 22, 1865–1874. [Google Scholar] [CrossRef]
  24. Ouyang, P.; Wang, L.; Wu, J.; Tian, Y.; Chen, C.; Li, D.; Yao, Z.; Chen, R.; Xiang, G.; Gong, J.; et al. Overcoming cold tumors: A combination strategy of immune checkpoint inhibitors. Front. Immunol. 2024, 15, 1344272. [Google Scholar] [CrossRef]
  25. Balasubramanian, A.; John, T.; Asselin-Labat, M.L. Regulation of the antigen presentation machinery in cancer and its implication for immune surveillance. Biochem. Soc. Trans. 2022, 50, 825–837. [Google Scholar] [CrossRef]
  26. Esfahani, K.; Roudaia, L.; Buhlaiga, N.; Del Rincon, S.V.; Papneja, N.; Miller, W.H., Jr. A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol. 2020, 27, S87–S97. [Google Scholar] [CrossRef]
  27. Golden, E.B.; Apetoh, L. Radiotherapy and immunogenic cell death. Semin. Radiat. Oncol. 2015, 25, 11–17. [Google Scholar] [CrossRef]
  28. Martinez-Outschoorn, U.E.; Peiris-Pages, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 11–31. [Google Scholar] [CrossRef] [PubMed]
  29. Kouidhi, S.; Elgaaied, A.B.; Chouaib, S. Impact of Metabolism on T-Cell Differentiation and Function and Cross Talk with Tumor Microenvironment. Front. Immunol. 2017, 8, 270. [Google Scholar] [CrossRef] [PubMed]
  30. Xu, Z.; Vonlaufen, A.; Phillips, P.A.; Fiala-Beer, E.; Zhang, X.; Yang, L.; Biankin, A.V.; Goldstein, D.; Pirola, R.C.; Wilson, J.S.; et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am. J. Pathol. 2010, 177, 2585–2596. [Google Scholar] [CrossRef] [PubMed]
  31. Gefen, T.; Castro, I.; Muharemagic, D.; Puplampu-Dove, Y.; Patel, S.; Gilboa, E. A TIM-3 Oligonucleotide Aptamer Enhances T Cell Functions and Potentiates Tumor Immunity in Mice. Mol. Ther. 2017, 25, 2280–2288. [Google Scholar] [CrossRef]
  32. Wang, S.; Zheng, Y.; Yang, F.; Zhu, L.; Zhu, X.Q.; Wang, Z.F.; Wu, X.L.; Zhou, C.H.; Yan, J.Y.; Hu, B.Y.; et al. The molecular biology of pancreatic adenocarcinoma: Translational challenges and clinical perspectives. Signal Transduct. Target. Ther. 2021, 6, 249. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, S.; Li, Y.; Xing, C.; Ding, C.; Zhang, H.; Chen, L.; You, L.; Dai, M.; Zhao, Y. Tumor microenvironment in chemoresistance, metastasis and immunotherapy of pancreatic cancer. Am. J. Cancer Res. 2020, 10, 1937–1953. [Google Scholar]
  34. Bure, I.V.; Nemtsova, M.V.; Zaletaev, D.V. Roles of E-cadherin and Noncoding RNAs in the Epithelial-mesenchymal Transition and Progression in Gastric Cancer. Int. J. Mol. Sci. 2019, 20, 2870. [Google Scholar] [CrossRef] [PubMed]
  35. Ren, B.; Cui, M.; Yang, G.; Wang, H.; Feng, M.; You, L.; Zhao, Y. Tumor microenvironment participates in metastasis of pancreatic cancer. Mol. Cancer 2018, 17, 108. [Google Scholar] [CrossRef] [PubMed]
  36. Qian, D.; Lu, Z.; Xu, Q.; Wu, P.; Tian, L.; Zhao, L.; Cai, B.; Yin, J.; Wu, Y.; Staveley-O’Carroll, K.F.; et al. Galectin-1-driven upregulation of SDF-1 in pancreatic stellate cells promotes pancreatic cancer metastasis. Cancer Lett. 2017, 397, 43–51. [Google Scholar] [CrossRef]
  37. Alsaafeen, B.H.; Ali, B.R.; Elkord, E. Resistance mechanisms to immune checkpoint inhibitors: Updated insights. Mol. Cancer 2025, 24, 20. [Google Scholar] [CrossRef]
  38. Johnson, D.B.; Nebhan, C.A.; Moslehi, J.J.; Balko, J.M. Immune-checkpoint inhibitors: Long-term implications of toxicity. Nat. Rev. Clin. Oncol. 2022, 19, 254–267. [Google Scholar] [CrossRef]
  39. Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef]
  40. Nagasaki, J.; Ishino, T.; Togashi, Y. Mechanisms of resistance to immune checkpoint inhibitors. Cancer Sci. 2022, 113, 3303–3312. [Google Scholar] [CrossRef]
  41. Kuwada, K.; Kagawa, S.; Yoshida, R.; Sakamoto, S.; Ito, A.; Watanabe, M.; Ieda, T.; Kuroda, S.; Kikuchi, S.; Tazawa, H.; et al. The epithelial-to-mesenchymal transition induced by tumor-associated macrophages confers chemoresistance in peritoneally disseminated pancreatic cancer. J. Exp. Clin. Cancer Res. 2018, 37, 307. [Google Scholar] [CrossRef] [PubMed]
  42. Zhou, P.; Li, B.; Liu, F.; Zhang, M.; Wang, Q.; Liu, Y.; Yao, Y.; Li, D. The epithelial to mesenchymal transition (EMT) and cancer stem cells: Implication for treatment resistance in pancreatic cancer. Mol. Cancer 2017, 16, 52. [Google Scholar] [CrossRef] [PubMed]
  43. Recouvreux, M.V.; Moldenhauer, M.R.; Galenkamp, K.M.O.; Jung, M.; James, B.; Zhang, Y.; Lowy, A.; Bagchi, A.; Commisso, C. Glutamine depletion regulates Slug to promote EMT and metastasis in pancreatic cancer. J. Exp. Med. 2020, 217, e20200388. [Google Scholar] [CrossRef]
  44. Karamitopoulou, E. Tumour microenvironment of pancreatic cancer: Immune landscape is dictated by molecular and histopathological features. Br. J. Cancer 2019, 121, 5–14. [Google Scholar] [CrossRef]
  45. Erkan, M.; Kurtoglu, M.; Kleeff, J. The role of hypoxia in pancreatic cancer: A potential therapeutic target? Expert. Rev. Gastroenterol. Hepatol. 2016, 10, 301–316. [Google Scholar] [CrossRef]
  46. Abdalla, M.Y.; Ahmad, I.M.; Rachagani, S.; Banerjee, K.; Thompson, C.M.; Maurer, H.C.; Olive, K.P.; Bailey, K.L.; Britigan, B.E.; Kumar, S. Enhancing responsiveness of pancreatic cancer cells to gemcitabine treatment under hypoxia by heme oxygenase-1 inhibition. Transl. Res. 2019, 207, 56–69. [Google Scholar] [CrossRef]
  47. Ahmad, I.M.; Dafferner, A.J.; O’Connell, K.A.; Mehla, K.; Britigan, B.E.; Hollingsworth, M.A.; Abdalla, M.Y. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers 2021, 13, 2264. [Google Scholar] [CrossRef]
  48. Zhang, S.; Fang, W.; Zhou, S.; Zhu, D.; Chen, R.; Gao, X.; Li, Z.; Fu, Y.; Zhang, Y.; Yang, F.; et al. Single cell transcriptomic analyses implicate an immunosuppressive tumor microenvironment in pancreatic cancer liver metastasis. Nat. Commun. 2023, 14, 5123. [Google Scholar] [CrossRef]
  49. Aysal, A.; Agalar, C.; Cagaptay, S.; Safak, T.; Egeli, T.; Ozbilgin, M.; Unek, T.; Unek, T.; Sagol, O. The Site of Lymph Node Metastasis: A Significant Prognostic Factor in Pancreatic Ductal Adenocarcinoma. Turk. Patoloji Derg. 2022, 38, 284–291. [Google Scholar] [CrossRef]
  50. Li, Q.; Feng, Z.; Miao, R.; Liu, X.; Liu, C.; Liu, Z. Prognosis and survival analysis of patients with pancreatic cancer: Retrospective experience of a single institution. World J. Surg. Oncol. 2022, 20, 11. [Google Scholar] [CrossRef] [PubMed]
  51. Yuan, X.; Wu, H.; Xu, H.; Xiong, H.; Chu, Q.; Yu, S.; Wu, G.S.; Wu, K. Notch signaling: An emerging therapeutic target for cancer treatment. Cancer Lett. 2015, 369, 20–27. [Google Scholar] [CrossRef]
  52. Sugisawa, N.; Miyake, K.; Higuchi, T.; Oshiro, H.; Park, J.H.; Kawaguchi, K.; Bouvet, M.; Unno, M.; Hoffman, R.M. High Incidence of Lymph-node Metastasis in a Pancreatic-cancer Patient-derived Orthotopic Xenograft (PDOX) NOG-Mouse Model. Anticancer. Res. 2022, 42, 739–743. [Google Scholar] [CrossRef] [PubMed]
  53. Fink, D.M.; Steele, M.M.; Hollingsworth, M.A. The lymphatic system and pancreatic cancer. Cancer Lett. 2016, 381, 217–236. [Google Scholar] [CrossRef]
  54. von Ahrens, D.; Bhagat, T.D.; Nagrath, D.; Maitra, A.; Verma, A. The role of stromal cancer-associated fibroblasts in pancreatic cancer. J. Hematol. Oncol. 2017, 10, 76. [Google Scholar] [CrossRef]
  55. Ferdek, P.E.; Jakubowska, M.A. Biology of pancreatic stellate cells-more than just pancreatic cancer. Pflug. Arch. 2017, 469, 1039–1050. [Google Scholar] [CrossRef]
  56. 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]
  57. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef] [PubMed]
  58. Yan, Z.; Ohuchida, K.; Fei, S.; Zheng, B.; Guan, W.; Feng, H.; Kibe, S.; Ando, Y.; Koikawa, K.; Abe, T.; et al. Inhibition of ERK1/2 in cancer-associated pancreatic stellate cells suppresses cancer-stromal interaction and metastasis. J. Exp. Clin. Cancer Res. 2019, 38, 221. [Google Scholar] [CrossRef] [PubMed]
  59. Jiang, W.; Bai, W.; Li, J.; Liu, J.; Zhao, K.; Ren, L. Leukemia inhibitory factor is a novel biomarker to predict lymph node and distant metastasis in pancreatic cancer. Int. J. Cancer 2021, 148, 1006–1013. [Google Scholar] [CrossRef]
  60. Charles Jacob, H.K.; Signorelli, R.; Charles Richard, J.L.; Kashuv, T.; Lavania, S.; Middleton, A.; Gomez, B.A.; Ferrantella, A.; Amirian, H.; Tao, J.; et al. Identification of novel early pancreatic cancer biomarkers KIF5B and SFRP2 from “first contact” interactions in the tumor microenvironment. J. Exp. Clin. Cancer Res. 2022, 41, 258. [Google Scholar] [CrossRef]
  61. Javadi, S.; Karbasian, N.; Bhosale, P.; de Castro Faria, S.; Le, O.; Katz, M.H.; Koay, E.J.; Tamm, E.P. Imaging findings of recurrent pancreatic cancer following resection. Abdom. Radiol. 2018, 43, 489–496. [Google Scholar] [CrossRef]
  62. Tao, J.; Yang, G.; Zhou, W.; Qiu, J.; Chen, G.; Luo, W.; Zhao, F.; You, L.; Zheng, L.; Zhang, T.; et al. Targeting hypoxic tumor microenvironment in pancreatic cancer. J. Hematol. Oncol. 2021, 14, 14. [Google Scholar] [CrossRef] [PubMed]
  63. Tomasek, J.J.; Gabbiani, G.; Hinz, B.; Chaponnier, C.; Brown, R.A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat. Rev. Mol. Cell Biol. 2002, 3, 349–363. [Google Scholar] [CrossRef]
  64. Garg, B.; Giri, B.; Modi, S.; Sethi, V.; Castro, I.; Umland, O.; Ban, Y.; Lavania, S.; Dawra, R.; Banerjee, S.; et al. NFκB in Pancreatic Stellate Cells Reduces Infiltration of Tumors by Cytotoxic T Cells and Killing of Cancer Cells, via Up-regulation of CXCL12. Gastroenterology 2018, 155, 880–891.e8. [Google Scholar] [CrossRef]
  65. Jiang, H.; Torphy, R.J.; Steiger, K.; Hongo, H.; Ritchie, A.J.; Kriegsmann, M.; Horst, D.; Umetsu, S.E.; Joseph, N.M.; McGregor, K.; et al. Pancreatic ductal adenocarcinoma progression is restrained by stromal matrix. J. Clin. Investig. 2020, 130, 4704–4709. [Google Scholar] [CrossRef]
  66. Hessmann, E.; Buchholz, S.M.; Demir, I.E.; Singh, S.K.; Gress, T.M.; Ellenrieder, V.; Neesse, A. Microenvironmental Determinants of Pancreatic Cancer. Physiol. Rev. 2020, 100, 1707–1751. [Google Scholar] [CrossRef]
  67. Thomas, D.; Radhakrishnan, P. Pancreatic Stellate Cells: The Key Orchestrator of the Pancreatic Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1234, 57–70. [Google Scholar] [CrossRef] [PubMed]
  68. Schnittert, J.; Bansal, R.; Prakash, J. Targeting Pancreatic Stellate Cells in Cancer. Trends Cancer 2019, 5, 128–142. [Google Scholar] [CrossRef] [PubMed]
  69. Tang, D.; Wang, D.; Yuan, Z.; Xue, X.; Zhang, Y.; An, Y.; Chen, J.; Tu, M.; Lu, Z.; Wei, J.; et al. Persistent activation of pancreatic stellate cells creates a microenvironment favorable for the malignant behavior of pancreatic ductal adenocarcinoma. Int. J. Cancer 2013, 132, 993–1003. [Google Scholar] [CrossRef]
  70. Saka, D.; Gokalp, M.; Piyade, B.; Cevik, N.C.; Arik Sever, E.; Unutmaz, D.; Ceyhan, G.O.; Demir, I.E.; Asimgil, H. Mechanisms of T-Cell Exhaustion in Pancreatic Cancer. Cancers 2020, 12, 2274. [Google Scholar] [CrossRef]
  71. Endo, S.; Nakata, K.; Ohuchida, K.; Takesue, S.; Nakayama, H.; Abe, T.; Koikawa, K.; Okumura, T.; Sada, M.; Horioka, K.; et al. Autophagy Is Required for Activation of Pancreatic Stellate Cells, Associated with Pancreatic Cancer Progression and Promotes Growth of Pancreatic Tumors in Mice. Gastroenterology 2017, 152, 1492–1506.e24. [Google Scholar] [CrossRef] [PubMed]
  72. Sunami, Y.; Häußler, J.; Kleeff, J. Cellular Heterogeneity of Pancreatic Stellate Cells, Mesenchymal Stem Cells, and Cancer-Associated Fibroblasts in Pancreatic Cancer. Cancers 2020, 12, 3770. [Google Scholar] [CrossRef]
  73. Piersma, B.; Hayward, M.K.; Weaver, V.M. Fibrosis and cancer: A strained relationship. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188356. [Google Scholar] [CrossRef]
  74. Miyabayashi, K.; Ijichi, H.; Fujishiro, M. The Role of the Microbiome in Pancreatic Cancer. Cancers 2022, 14, 4479. [Google Scholar] [CrossRef]
  75. Gardian, K.; Janczewska, S.; Olszewski, W.L.; Durlik, M. Analysis of pancreatic cancer microenvironment: Role of macrophage infiltrates and growth factors expression. J. Cancer 2012, 3, 285–291. [Google Scholar] [CrossRef] [PubMed]
  76. Xiang, H.; Yang, R.; Tu, J.; Xi, Y.; Yang, S.; Lv, L.; Zhai, X.; Zhu, Y.; Dong, D.; Tao, X. Metabolic reprogramming of immune cells in pancreatic cancer progression. Biomed. Pharmacother. 2023, 157, 113992. [Google Scholar] [CrossRef] [PubMed]
  77. Huber, M.; Brehm, C.U.; Gress, T.M.; Buchholz, M.; Alashkar Alhamwe, B.; von Strandmann, E.P.; Slater, E.P.; Bartsch, J.W.; Bauer, C.; Lauth, M. The Immune Microenvironment in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7307. [Google Scholar] [CrossRef]
  78. Santoni, M.; Bracarda, S.; Nabissi, M.; Massari, F.; Conti, A.; Bria, E.; Tortora, G.; Santoni, G.; Cascinu, S. CXC and CC chemokines as angiogenic modulators in nonhaematological tumors. Biomed. Res. Int. 2014, 2014, 768758. [Google Scholar] [CrossRef]
  79. Bolli, E.; Movahedi, K.; Laoui, D.; Van Ginderachter, J.A. Novel insights in the regulation and function of macrophages in the tumor microenvironment. Curr. Opin. Oncol. 2017, 29, 55–61. [Google Scholar] [CrossRef]
  80. Nywening, T.M.; Belt, B.A.; Cullinan, D.R.; Panni, R.Z.; Han, B.J.; Sanford, D.E.; Jacobs, R.C.; Ye, J.; Patel, A.A.; Gillanders, W.E.; et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 2018, 67, 1112–1123. [Google Scholar] [CrossRef]
  81. Filippini, D.; Agosto, S.D.; Delfino, P.; Simbolo, M.; Piro, G.; Rusev, B.; Veghini, L.; Cantù, C.; Lupo, F.; Ugel, S.; et al. Immunoevolution of mouse pancreatic organoid isografts from preinvasive to metastatic disease. Sci. Rep. 2019, 9, 12286. [Google Scholar] [CrossRef]
  82. Deng, S.J.; Chen, H.Y.; Ye, Z.; Deng, S.C.; Zhu, S.; Zeng, Z.; He, C.; Liu, M.L.; Huang, K.; Zhong, J.X.; et al. Hypoxia-induced LncRNA-BX111 promotes metastasis and progression of pancreatic cancer through regulating ZEB1 transcription. Oncogene 2018, 37, 5811–5828. [Google Scholar] [CrossRef]
  83. McDonald, P.C.; Chafe, S.C.; Brown, W.S.; Saberi, S.; Swayampakula, M.; Venkateswaran, G.; Nemirovsky, O.; Gillespie, J.A.; Karasinska, J.M.; Kalloger, S.E.; et al. Regulation of pH by Carbonic Anhydrase 9 Mediates Survival of Pancreatic Cancer Cells with Activated KRAS in Response to Hypoxia. Gastroenterology 2019, 157, 823–837. [Google Scholar] [CrossRef]
  84. Pitt, J.M.; Charrier, M.; Viaud, S.; Andre, F.; Besse, B.; Chaput, N.; Zitvogel, L. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J. Immunol. 2014, 193, 1006–1011. [Google Scholar] [CrossRef]
  85. Gong, J.; Chen, D.; Kashiwaba, M.; Kufe, D. Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat. Med. 1997, 3, 558–561. [Google Scholar] [CrossRef] [PubMed]
  86. Kajihara, M.; Takakura, K.; Kanai, T.; Ito, Z.; Matsumoto, Y.; Shimodaira, S.; Okamoto, M.; Ohkusa, T.; Koido, S. Advances in inducing adaptive immunity using cell-based cancer vaccines: Clinical applications in pancreatic cancer. World J. Gastroenterol. 2016, 22, 4446–4458. [Google Scholar] [CrossRef]
  87. Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; et al. The prioritization of cancer antigens: A national cancer institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15, 5323–5337. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, M.; Zhong, J.; Zeng, Z.; Huang, K.; Ye, Z.; Deng, S.; Chen, H.; Xu, F.; Li, Q.; Zhao, G. Hypoxia-induced feedback of HIF-1α and lncRNA-CF129 contributes to pancreatic cancer progression through stabilization of p53 protein. Theranostics 2019, 9, 4795–4810. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, G.; Wu, K.; Li, H.; Xia, D.; He, T. Role of hypoxia in the tumor microenvironment and targeted therapy. Front. Oncol. 2022, 12, 961637. [Google Scholar] [CrossRef]
  90. Torres, N.; Regge, M.V.; Secchiari, F.; Friedrich, A.D.; Spallanzani, R.G.; Raffo Iraolagoitia, X.L.; Nunez, S.Y.; Sierra, J.M.; Ziblat, A.; Santilli, M.C.; et al. Restoration of antitumor immunity through anti-MICA antibodies elicited with a chimeric protein. J. Immunother. Cancer 2020, 8, e000233. [Google Scholar] [CrossRef]
  91. Turriziani, M.; Fantini, M.; Benvenuto, M.; Izzi, V.; Masuelli, L.; Sacchetti, P.; Modesti, A.; Bei, R. Carcinoembryonic antigen (CEA)-based cancer vaccines: Recent patents and antitumor effects from experimental models to clinical trials. Recent. Pat. Anticancer. Drug Discov. 2012, 7, 265–296. [Google Scholar] [CrossRef] [PubMed]
  92. Thompson, J.A.; Grunert, F.; Zimmermann, W. Carcinoembryonic antigen gene family: Molecular biology and clinical perspectives. J. Clin. Lab. Anal. 1991, 5, 344–366. [Google Scholar] [CrossRef]
  93. Kanda, M.; Matthaei, H.; Wu, J.; Hong, S.M.; Yu, J.; Borges, M.; Hruban, R.H.; Maitra, A.; Kinzler, K.; Vogelstein, B.; et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 2012, 142, 730–733.e9. [Google Scholar] [CrossRef]
  94. 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]
  95. Kanda, M.; Fujii, T.; Nagai, S.; Kodera, Y.; Kanzaki, A.; Sahin, T.T.; Hayashi, M.; Yamada, S.; Sugimoto, H.; Nomoto, S.; et al. Pattern of lymph node metastasis spread in pancreatic cancer. Pancreas 2011, 40, 951–955. [Google Scholar] [CrossRef] [PubMed]
  96. Riabov, V.; Gudima, A.; Wang, N.; Mickley, A.; Orekhov, A.; Kzhyshkowska, J. Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front. Physiol. 2014, 5, 75. [Google Scholar] [CrossRef]
  97. Weden, S.; Klemp, M.; Gladhaug, I.P.; Moller, M.; Eriksen, J.A.; Gaudernack, G.; Buanes, T. Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int. J. Cancer 2011, 128, 1120–1128. [Google Scholar] [CrossRef]
  98. Tsujikawa, T.; Crocenzi, T.; Durham, J.N.; Sugar, E.A.; Wu, A.A.; Onners, B.; Nauroth, J.M.; Anders, R.A.; Fertig, E.J.; Laheru, D.A.; et al. Evaluation of Cyclophosphamide/GVAX Pancreas Followed by Listeria-Mesothelin (CRS-207) with or without Nivolumab in Patients with Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 3578–3588. [Google Scholar] [CrossRef]
  99. Zhang, Z.; Ji, S.; Zhang, B.; Liu, J.; Qin, Y.; Xu, J.; Yu, X. Role of angiogenesis in pancreatic cancer biology and therapy. Biomed. Pharmacother. 2018, 108, 1135–1140. [Google Scholar] [CrossRef]
  100. Yang, E.; Wang, X.; Gong, Z.; Yu, M.; Wu, H.; Zhang, D. Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct. Target. Ther. 2020, 5, 242. [Google Scholar] [CrossRef] [PubMed]
  101. Naxerova, K.; Reiter, J.G.; Brachtel, E.; Lennerz, J.K.; van de Wetering, M.; Rowan, A.; Cai, T.; Clevers, H.; Swanton, C.; Nowak, M.A.; et al. Origins of lymphatic and distant metastases in human colorectal cancer. Science 2017, 357, 55–60. [Google Scholar] [CrossRef]
  102. Jacquemet, G.; Hamidi, H.; Ivaska, J. Filopodia in cell adhesion, 3D migration and cancer cell invasion. Curr. Opin. Cell Biol. 2015, 36, 23–31. [Google Scholar] [CrossRef] [PubMed]
  103. Le, D.T.; Picozzi, V.J.; Ko, A.H.; Wainberg, Z.A.; Kindler, H.; Wang-Gillam, A.; Oberstein, P.; Morse, M.A.; Zeh, H.J., 3rd; Weekes, C.; et al. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study). Clin. Cancer Res. 2019, 25, 5493–5502. [Google Scholar] [CrossRef]
  104. Bassani-Sternberg, M.; Digklia, A.; Huber, F.; Wagner, D.; Sempoux, C.; Stevenson, B.J.; Thierry, A.C.; Michaux, J.; Pak, H.; Racle, J.; et al. A Phase Ib Study of the Combination of Personalized Autologous Dendritic Cell Vaccine, Aspirin, and Standard of Care Adjuvant Chemotherapy Followed by Nivolumab for Resected Pancreatic Adenocarcinoma-A Proof of Antigen Discovery Feasibility in Three Patients. Front. Immunol. 2019, 10, 1832. [Google Scholar] [CrossRef]
  105. Vacchelli, E.; Aranda, F.; Eggermont, A.; Galon, J.; Sautes-Fridman, C.; Cremer, I.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial Watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2014, 3, e27878. [Google Scholar] [CrossRef] [PubMed]
  106. Hu, X.; Luo, J. Heterogeneity of tumor lymphangiogenesis: Progress and prospects. Cancer Sci. 2018, 109, 3005–3012. [Google Scholar] [CrossRef]
  107. Hall, M.; Liu, H.; Malafa, M.; Centeno, B.; Hodul, P.J.; Pimiento, J.; Pilon-Thomas, S.; Sarnaik, A.A. Expansion of tumor-infiltrating lymphocytes (TIL) from human pancreatic tumors. J. Immunother. Cancer 2016, 4, 61. [Google Scholar] [CrossRef]
  108. Smid, M.; Wilting, S.M.; Uhr, K.; Rodríguez-González, F.G.; de Weerd, V.; Prager-Van der Smissen, W.J.C.; van der Vlugt-Daane, M.; van Galen, A.; Nik-Zainal, S.; Butler, A.; et al. The circular RNome of primary breast cancer. Genome Res. 2019, 29, 356–366. [Google Scholar] [CrossRef]
  109. Meng, Q.; Valentini, D.; Rao, M.; Maeurer, M. KRAS RENAISSANCE(S) in Tumor Infiltrating B Cells in Pancreatic Cancer. Front. Oncol. 2018, 8, 384. [Google Scholar] [CrossRef]
  110. Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell. Mol. Immunol. 2021, 18, 842–859. [Google Scholar] [CrossRef]
  111. Parrot, T.; Oger, R.; Allard, M.; Desfrançois, J.; Raingeard de la Blétière, D.; Coutolleau, A.; Preisser, L.; Khammari, A.; Dréno, B.; Delneste, Y.; et al. Transcriptomic features of tumour-infiltrating CD4lowCD8high double positive αβ T cells in melanoma. Sci. Rep. 2020, 10, 5900. [Google Scholar] [CrossRef]
  112. Egelston, C.A.; Avalos, C.; Tu, T.Y.; Simons, D.L.; Jimenez, G.; Jung, J.Y.; Melstrom, L.; Margolin, K.; Yim, J.H.; Kruper, L.; et al. Human breast tumor-infiltrating CD8+ T cells retain polyfunctionality despite PD-1 expression. Nat. Commun. 2018, 9, 4297. [Google Scholar] [CrossRef] [PubMed]
  113. Cheuk, S.; Schlums, H.; Gallais Sérézal, I.; Martini, E.; Chiang, S.C.; Marquardt, N.; Gibbs, A.; Detlofsson, E.; Introini, A.; Forkel, M.; et al. CD49a Expression Defines Tissue-Resident CD8+ T Cells Poised for Cytotoxic Function in Human Skin. Immunity 2017, 46, 287–300. [Google Scholar] [CrossRef]
  114. Zaid, A.; Hor, J.L.; Christo, S.N.; Groom, J.R.; Heath, W.R.; Mackay, L.K.; Mueller, S.N. Chemokine Receptor-Dependent Control of Skin Tissue-Resident Memory T Cell Formation. J. Immunol. 2017, 199, 2451–2459. [Google Scholar] [CrossRef]
  115. Oja, A.E.; Piet, B.; van der Zwan, D.; Blaauwgeers, H.; Mensink, M.; de Kivit, S.; Borst, J.; Nolte, M.A.; van Lier, R.A.W.; Stark, R.; et al. Functional Heterogeneity of CD4+ Tumor-Infiltrating Lymphocytes with a Resident Memory Phenotype in NSCLC. Front. Immunol. 2018, 9, 2654. [Google Scholar] [CrossRef]
  116. Berntsson, J.; Nodin, B.; Eberhard, J.; Micke, P.; Jirström, K. Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer. Int. J. Cancer 2016, 139, 1129–1139. [Google Scholar] [CrossRef]
  117. Cillo, A.R.; Kürten, C.H.L.; Tabib, T.; Qi, Z.; Onkar, S.; Wang, T.; Liu, A.; Duvvuri, U.; Kim, S.; Soose, R.J.; et al. Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. Immunity 2020, 52, 183–199.e9. [Google Scholar] [CrossRef] [PubMed]
  118. Garaud, S.; Buisseret, L.; Solinas, C.; Gu-Trantien, C.; de Wind, A.; Van den Eynden, G.; Naveaux, C.; Lodewyckx, J.N.; Boisson, A.; Duvillier, H.; et al. Tumor infiltrating B-cells signal functional humoral immune responses in breast cancer. JCI Insight 2019, 5, e129641. [Google Scholar] [CrossRef]
  119. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
  120. Senturk, Z.N.; Akdag, I.; Deniz, B.; Sayi-Yazgan, A. Pancreatic cancer: Emerging field of regulatory B-cell-targeted immunotherapies. Front. Immunol. 2023, 14, 1152551. [Google Scholar] [CrossRef] [PubMed]
  121. Pylayeva-Gupta, Y.; Das, S.; Handler, J.S.; Hajdu, C.H.; Coffre, M.; Koralov, S.B.; Bar-Sagi, D. IL35-Producing B Cells Promote the Development of Pancreatic Neoplasia. Cancer Discov. 2016, 6, 247–255. [Google Scholar] [CrossRef]
  122. Hua, Z.; Hou, B. The role of B cell antigen presentation in the initiation of CD4+ T cell response. Immunol. Rev. 2020, 296, 24–35. [Google Scholar] [CrossRef]
  123. Huang, H.; Wang, Z.; Zhang, Y.; Pradhan, R.N.; Ganguly, D.; Chandra, R.; Murimwa, G.; Wright, S.; Gu, X.; Maddipati, R.; et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 2022, 40, 656–673.e7. [Google Scholar] [CrossRef] [PubMed]
  124. Hong, S.; Zhang, Z.; Liu, H.; Tian, M.; Zhu, X.; Zhang, Z.; Wang, W.; Zhou, X.; Zhang, F.; Ge, Q.; et al. B Cells Are the Dominant Antigen-Presenting Cells that Activate Naive CD4+ T Cells upon Immunization with a Virus-Derived Nanoparticle Antigen. Immunity 2018, 49, 695–708.e4. [Google Scholar] [CrossRef]
  125. Thyagarajan, A.; Alshehri, M.S.A.; Miller, K.L.R.; Sherwin, C.M.; Travers, J.B.; Sahu, R.P. Myeloid-Derived Suppressor Cells and Pancreatic Cancer: Implications in Novel Therapeutic Approaches. Cancers 2019, 11, 1627. [Google Scholar] [CrossRef] [PubMed]
  126. Mota Reyes, C.; Demir, E.; Cifcibasi, K.; Istvanffy, R.; Friess, H.; Demir, I.E. Regulatory T Cells in Pancreatic Cancer: Of Mice and Men. Cancers 2022, 14, 4582. [Google Scholar] [CrossRef] [PubMed]
  127. Amedei, A.; Niccolai, E.; Prisco, D. Pancreatic cancer: Role of the immune system in cancer progression and vaccine-based immunotherapy. Hum. Vaccin. Immunother. 2014, 10, 3354–3368. [Google Scholar] [CrossRef]
  128. Duffey, D.C.; Crowl-Bancroft, C.V.; Chen, Z.; Ondrey, F.G.; Nejad-Sattari, M.; Dong, G.; Van Waes, C. Inhibition of transcription factor nuclear factor-kappaB by a mutant inhibitor-kappaBalpha attenuates resistance of human head and neck squamous cell carcinoma to TNF-alpha caspase-mediated cell death. Br. J. Cancer 2000, 83, 1367–1374. [Google Scholar] [CrossRef]
  129. Zwirner, N.W.; Domaica, C.I. Cytokine regulation of natural killer cell effector functions. Biofactors 2010, 36, 274–288. [Google Scholar] [CrossRef]
  130. Sadozai, H.; Acharjee, A.; Eppenberger-Castori, S.; Gloor, B.; Gruber, T.; Schenk, M.; Karamitopoulou, E. Distinct Stromal and Immune Features Collectively Contribute to Long-Term Survival in Pancreatic Cancer. Front. Immunol. 2021, 12, 643529. [Google Scholar] [CrossRef]
  131. Xu, C.; Sui, S.; Shang, Y.; Yu, Z.; Han, J.; Zhang, G.; Ntim, M.; Hu, M.; Gong, P.; Chen, H.; et al. The landscape of immune cell infiltration and its clinical implications of pancreatic ductal adenocarcinoma. J. Adv. Res. 2020, 24, 139–148. [Google Scholar] [CrossRef] [PubMed]
  132. Zhu, T.; Wu, X.; Liao, Y.; Yan, Y.; Yu, M.; Wang, L.; Xia, Q. The role of innate immune cells as modulators of the tumor microenvironment in the metastasis and treatment of pancreatic cancer. Clin. Cancer Bull. 2023, 2, 2. [Google Scholar] [CrossRef]
  133. Hamarsheh, S.; Gross, O.; Brummer, T.; Zeiser, R. Immune modulatory effects of oncogenic KRAS in cancer. Nat. Commun. 2020, 11, 5439. [Google Scholar] [CrossRef]
  134. Wang, C.; Tan, J.Y.M.; Chitkara, N.; Bhatt, S. TP53 Mutation-Mediated Immune Evasion in Cancer: Mechanisms and Therapeutic Implications. Cancers 2024, 16, 3069. [Google Scholar] [CrossRef]
  135. Aroldi, F.; Zaniboni, A. Immunotherapy for pancreatic cancer: Present and future. Immunotherapy 2017, 9, 607–616. [Google Scholar] [CrossRef]
  136. McCarthy, E.F. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar]
  137. Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196. [Google Scholar] [CrossRef]
  138. Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [PubMed]
  139. Emens, L.A.; Ascierto, P.A.; Darcy, P.K.; Demaria, S.; Eggermont, A.M.M.; Redmond, W.L.; Seliger, B.; Marincola, F.M. Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur. J. Cancer 2017, 81, 116–129. [Google Scholar] [CrossRef]
  140. Katz, M.H.G.; Petroni, G.R.; Bauer, T.; Reilley, M.J.; Wolpin, B.M.; Stucky, C.C.; Bekaii-Saab, T.S.; Elias, R.; Merchant, N.; Dias Costa, A.; et al. Multicenter randomized controlled trial of neoadjuvant chemoradiotherapy alone or in combination with pembrolizumab in patients with resectable or borderline resectable pancreatic adenocarcinoma. J. Immunother. Cancer 2023, 11, e007586. [Google Scholar] [CrossRef] [PubMed]
  141. Abbas, A.A.; Davelaar, J.; Gai, J.; Brown, Z.; Levi, A.; Linden, S.; Minasyan, A.; Rodriguez, C.; Pachter, J.A.; Gong, J.; et al. Preliminary translational immune and stromal correlates in a randomized phase II trial of pembrolizumab with or without defactinib for resectable pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2023, 41, 4024. [Google Scholar] [CrossRef]
  142. Mueller, S.; Engleitner, T.; Maresch, R.; Zukowska, M.; Lange, S.; Kaltenbacher, T.; Konukiewitz, B.; Ollinger, R.; Zwiebel, M.; Strong, A.; et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 2018, 554, 62–68. [Google Scholar] [CrossRef]
  143. 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]
  144. Chick, R.C.; Gunderson, A.J.; Rahman, S.; Cloyd, J.M. Neoadjuvant Immunotherapy for Localized Pancreatic Cancer: Challenges and Early Results. Cancers 2023, 15, 3967. [Google Scholar] [CrossRef]
  145. Balsano, R.; Zanuso, V.; Pirozzi, A.; Rimassa, L.; Bozzarelli, S. Pancreatic Ductal Adenocarcinoma and Immune Checkpoint Inhibitors: The Gray Curtain of Immunotherapy and Spikes of Lights. Curr. Oncol. 2023, 30, 3871–3885. [Google Scholar] [CrossRef]
  146. Wang, J.; Gai, J.; Zhang, T.; Niu, N.; Qi, H.; Thomas, D.L., 2nd; Li, K.; Xia, T.; Rodriguez, C.; Parkinson, R.; et al. Neoadjuvant radioimmunotherapy in pancreatic cancer enhances effector T cell infiltration and shortens their distances to tumor cells. Sci. Adv. 2024, 10, eadk1827. [Google Scholar] [CrossRef]
  147. Sahin, I.; Turen, S.; Santapuram, P.; Sahin, I.H. The tumor microenvironment of pancreatic adenocarcinoma and immune checkpoint inhibitor resistance: A perplex relationship. Cancer Drug Resist. 2020, 3, 699–709. [Google Scholar] [CrossRef] [PubMed]
  148. Fu, Q.; Chen, Y.; Huang, D.; Guo, C.; Zhang, X.; Xiao, W.; Xue, X.; Zhang, Q.; Li, X.; Gao, S.; et al. Sintilimab Plus Modified FOLFIRINOX in Metastatic or Recurrent Pancreatic Cancer: The Randomized Phase II CISPD3 Trial. Ann. Surg. Oncol. 2023, 30, 5071–5080. [Google Scholar] [CrossRef] [PubMed]
  149. Yang, S.H.; Kuo, S.H.; Lee, J.C.; Chen, B.B.; Shan, Y.S.; Tien, Y.W.; Chiu, S.C.; Cheng, A.L.; Yeh, K.H. Adding-on nivolumab to chemotherapy-stabilized patients is associated with improved survival in advanced pancreatic ductal adenocarcinoma. Cancer Immunol. Immunother. 2024, 73, 227. [Google Scholar] [CrossRef]
  150. Olaoba, O.T.; Yang, M.; Adelusi, T.I.; Maidens, T.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G. Targeted Therapy for Highly Desmoplastic and Immunosuppressive Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers 2024, 16, 1470. [Google Scholar] [CrossRef]
  151. Shin, S.M.; Hernandez, A.; Coyne, E.; Zhang, Z.; Mitchell, S.; Durham, J.; Yuan, X.; Yang, H.; Fertig, E.J.; Jaffee, E.M.; et al. Abstract 2270: Combination of CXCR4 antagonist and anti-PD1 therapy results in significant mobilization and increased infiltration of myeloid cells into the metastatic liver microenvironment of PDAC. Cancer Res. 2023, 83, 2270. [Google Scholar] [CrossRef]
  152. Reiss, K.A.; Mick, R.; Teitelbaum, U.; O’Hara, M.; Schneider, C.; Massa, R.; Karasic, T.; Tondon, R.; Onyiah, C.; Gosselin, M.K.; et al. Niraparib plus nivolumab or niraparib plus ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: A randomised, phase 1b/2 trial. Lancet Oncol. 2022, 23, 1009–1020. [Google Scholar] [CrossRef]
  153. Macarulla Mercade, T.; McAndrews, K.; Pazo Cid, R.A.; Medina Rodríguez, L.; Gil-Negrete, A.; Rivera Herrero, F.; Varela Pose, V.; Martín-Muñoz, A.; Ruiz-Heredia, Y.; Kalluri, R.; et al. 331P Phase Ib/IIa study to evaluate safety and efficacy of priming treatment with the hedgehog inhibitor NLM-001 prior to gemcitabine and nab-paclitaxel plus zalifrelimab as first-line treatment in patients with advanced pancreatic cancer: NUMANTIA study. Ann. Oncol. 2024, 35, S139–S140. [Google Scholar] [CrossRef]
  154. Gross, N.E.; Zhang, Z.; Mitchell, J.T.; Charmsaz, S.; Hernandez, A.G.; Coyne, E.M.; Shin, S.M.; Vargas Carvajal, D.C.; Sidiropoulos, D.N.; Cho, Y.; et al. Phosphodiesterase-5 inhibition collaborates with vaccine-based immunotherapy to reprogram myeloid cells in pancreatic ductal adenocarcinoma. JCI Insight 2024, 9, e179292. [Google Scholar] [CrossRef]
  155. Piatek, M.; Bienkowski, M.; Kusnierz, K.; Pilch-Kowalczyk, J.; Imielska-Zdunek, D.; Mrowiec, S.; Lampe, P.; Radecka, B.; Nawrocki, S. Combination of modified FOLFIRINOX with stereotactic body radiotherapy as an induction therapy for locally advanced pancreatic adenocarcinoma—A prospective single-arm study. Contemp. Oncol. 2024, 28, 15–30. [Google Scholar] [CrossRef]
  156. Schizas, D.; Charalampakis, N.; Kole, C.; Economopoulou, P.; Koustas, E.; Gkotsis, E.; Ziogas, D.; Psyrri, A.; Karamouzis, M.V. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat. Rev. 2020, 86, 102016. [Google Scholar] [CrossRef] [PubMed]
  157. Foucher, E.D.; Ghigo, C.; Chouaib, S.; Galon, J.; Iovanna, J.; Olive, D. Pancreatic Ductal Adenocarcinoma: A Strong Imbalance of Good and Bad Immunological Cops in the Tumor Microenvironment. Front. Immunol. 2018, 9, 1044. [Google Scholar] [CrossRef]
  158. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef]
  159. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef]
  160. Oiseth, S.J.; Aziz, M.S. Cancer immunotherapy: A brief review of the history, possibilities, and challenges ahead. J. Cancer Metastasis Treat. 2017, 3, 250–261. [Google Scholar] [CrossRef]
  161. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef]
  162. Sureban, S.M.; Berahovich, R.; Zhou, H.; Xu, S.; Wu, L.; Ding, K.; May, R.; Qu, D.; Bannerman-Menson, E.; Golubovskaya, V.; et al. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers 2019, 12, 54. [Google Scholar] [CrossRef] [PubMed]
  163. Liu, L.; Huang, X.; Shi, F.; Song, J.; Guo, C.; Yang, J.; Liang, T.; Bai, X. Combination therapy for pancreatic cancer: Anti-PD-(L)1-based strategy. J. Exp. Clin. Cancer Res. 2022, 41, 56. [Google Scholar] [CrossRef] [PubMed]
  164. Mukherji, R.; Debnath, D.; Hartley, M.L.; Noel, M.S. The Role of Immunotherapy in Pancreatic Cancer. Curr. Oncol. 2022, 29, 6864–6892. [Google Scholar] [CrossRef] [PubMed]
  165. Feng, M.; Xiong, G.; Cao, Z.; Yang, G.; Zheng, S.; Song, X.; You, L.; Zheng, L.; Zhang, T.; Zhao, Y. PD-1/PD-L1 and immunotherapy for pancreatic cancer. Cancer Lett. 2017, 407, 57–65. [Google Scholar] [CrossRef]
  166. Akce, M.; Zaidi, M.Y.; Waller, E.K.; El-Rayes, B.F.; Lesinski, G.B. The Potential of CAR T Cell Therapy in Pancreatic Cancer. Front. Immunol. 2018, 9, 2166. [Google Scholar] [CrossRef]
  167. Chulpanova, D.S.; Kitaeva, K.V.; Green, A.R.; Rizvanov, A.A.; Solovyeva, V.V. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front. Cell Dev. Biol. 2020, 8, 402. [Google Scholar] [CrossRef]
  168. Cao, Z.; Weygant, N.; Chandrakesan, P.; Houchen, C.W.; Peng, J.; Qu, D. Tuft and Cancer Stem Cell Marker DCLK1: A New Target to Enhance Anti-Tumor Immunity in the Tumor Microenvironment. Cancers 2020, 12, 3801. [Google Scholar] [CrossRef]
  169. Ding, L.; Weygant, N.; Ding, C.; Lai, Y.; Li, H. DCLK1 and tuft cells: Immune-related functions and implications for cancer immunotherapy. Crit. Rev. Oncol. Hematol. 2023, 191, 104118. [Google Scholar] [CrossRef]
  170. Sunami, Y.; Kleeff, J. Immunotherapy of pancreatic cancer. Prog. Mol. Biol. Transl. Sci. 2019, 164, 189–216. [Google Scholar] [CrossRef]
  171. Leidner, R.; Sanjuan Silva, N.; Huang, H.; Sprott, D.; Zheng, C.; Shih, Y.P.; Leung, A.; Payne, R.; Sutcliffe, K.; Cramer, J.; et al. Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. N. Engl. J. Med. 2022, 386, 2112–2119. [Google Scholar] [CrossRef] [PubMed]
  172. Beatty, G.L.; O’Hara, M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: Defining the challenges and next steps. Pharmacol. Ther. 2016, 166, 30–39. [Google Scholar] [CrossRef] [PubMed]
  173. Malfertheiner, P.; Megraud, F.; O’Morain, C.A.; Gisbert, J.P.; Kuipers, E.J.; Axon, A.T.; Bazzoli, F.; Gasbarrini, A.; Atherton, J.; Graham, D.Y.; et al. Management of Helicobacter pylori infection-the Maastricht V/Florence Consensus Report. Gut 2017, 66, 6–30. [Google Scholar] [CrossRef] [PubMed]
  174. Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34. [Google Scholar] [CrossRef]
  175. Hu, Z.I.; Hellmann, M.D.; Wolchok, J.D.; Vyas, M.; Shia, J.; Stadler, Z.K.; Diaz, L.A., Jr.; O’Reilly, E.M. Acquired resistance to immunotherapy in MMR-D pancreatic cancer. J. Immunother. Cancer 2018, 6, 127. [Google Scholar] [CrossRef]
  176. 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]
  177. Pfirschke, C.; Engblom, C.; Rickelt, S.; Cortez-Retamozo, V.; Garris, C.; Pucci, F.; Yamazaki, T.; Poirier-Colame, V.; Newton, A.; Redouane, Y.; et al. Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. Immunity 2016, 44, 343–354. [Google Scholar] [CrossRef]
  178. Wang, Q.; Douglass, J.; Hwang, M.S.; Hsiue, E.H.; Mog, B.J.; Zhang, M.; Papadopoulos, N.; Kinzler, K.W.; Zhou, S.; Vogelstein, B. Direct Detection and Quantification of Neoantigens. Cancer Immunol. Res. 2019, 7, 1748–1754. [Google Scholar] [CrossRef]
  179. Yum, S.; Li, M.; Frankel, A.E.; Chen, Z.J. Roles of the cGAS-STING Pathway in Cancer Immunosurveillance and Immunotherapy. Annu. Rev. Cancer Biol. 2019, 3, 323–344. [Google Scholar] [CrossRef]
  180. Rech, A.J.; Dada, H.; Kotzin, J.J.; Henao-Mejia, J.; Minn, A.J.; Twyman-Saint Victor, C.; Vonderheide, R.H. Radiotherapy and CD40 Activation Separately Augment Immunity to Checkpoint Blockade in Cancer. Cancer Res. 2018, 78, 4282–4291. [Google Scholar] [CrossRef]
  181. Bear, A.S.; Vonderheide, R.H.; O’Hara, M.H. Challenges and Opportunities for Pancreatic Cancer Immunotherapy. Cancer Cell 2020, 38, 788–802. [Google Scholar] [CrossRef] [PubMed]
  182. Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef]
  183. Hopkins, A.C.; Yarchoan, M.; Durham, J.N.; Yusko, E.C.; Rytlewski, J.A.; Robins, H.S.; Laheru, D.A.; Le, D.T.; Lutz, E.R.; Jaffee, E.M. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight 2018, 3, e122092. [Google Scholar] [CrossRef]
  184. Curran, M.A.; Kim, M.; Montalvo, W.; Al-Shamkhani, A.; Allison, J.P. Combination CTLA-4 Blockade and 4-1BB Activation Enhances Tumor Rejection by Increasing T-Cell Infiltration, Proliferation, and Cytokine Production. PLoS ONE 2011, 6, e19499. [Google Scholar] [CrossRef]
  185. Zahavi, D.; Hodge, J.W. Targeting Immunosuppressive Adenosine Signaling: A Review of Potential Immunotherapy Combination Strategies. Int. J. Mol. Sci. 2023, 24, 8871. [Google Scholar] [CrossRef]
  186. Chiorean, E.G.; Coveler, A.L. Pancreatic cancer: Optimizing treatment options, new, and emerging targeted therapies. Drug Des. Devel Ther. 2015, 9, 3529–3545. [Google Scholar] [CrossRef]
  187. Trujillo, J.A.; Sweis, R.F.; Bao, R.; Luke, J.J. T Cell-Inflamed versus Non-T Cell-Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection. Cancer Immunol. Res. 2018, 6, 990–1000. [Google Scholar] [CrossRef] [PubMed]
  188. Zhang, Y.; Velez-Delgado, A.; Mathew, E.; Li, D.; Mendez, F.M.; Flannagan, K.; Rhim, A.D.; Simeone, D.M.; Beatty, G.L.; Pasca di Magliano, M. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017, 66, 124–136. [Google Scholar] [CrossRef] [PubMed]
  189. 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]
  190. Hussain, Z.; Bertran, T.; Finetti, P.; Lohmann, E.; Mamessier, E.; Bidaut, G.; Bertucci, F.; Rego, M.; Tomasini, R. Macrophages reprogramming driven by cancer-associated fibroblasts under FOLFIRINOX treatment correlates with shorter survival in pancreatic cancer. Cell Commun. Signal 2024, 22, 1. [Google Scholar] [CrossRef]
  191. Topalian, S.L.; Weiner, G.J.; Pardoll, D.M. Cancer immunotherapy comes of age. J. Clin. Oncol. 2011, 29, 4828–4836. [Google Scholar] [CrossRef]
  192. Tsang, K.Y.; Fantini, M.; Mavroukakis, S.A.; Zaki, A.; Annunziata, C.M.; Arlen, P.M. Development and Characterization of an Anti-Cancer Monoclonal Antibody for Treatment of Human Carcinomas. Cancers 2022, 14, 3037. [Google Scholar] [CrossRef] [PubMed]
  193. Hafeez, U.; Parakh, S.; Gan, H.K.; Scott, A.M. Antibody-Drug Conjugates for Cancer Therapy. Molecules 2020, 25, 4764. [Google Scholar] [CrossRef] [PubMed]
  194. Grewal, I.S.; Flavell, R.A. CD40 and CD154 in cell-mediated immunity. Annu. Rev. Immunol. 1998, 16, 111–135. [Google Scholar] [CrossRef]
  195. Bennett, S.R.; Carbone, F.R.; Karamalis, F.; Flavell, R.A.; Miller, J.F.; Heath, W.R. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 1998, 393, 478–480. [Google Scholar] [CrossRef]
  196. Henriksen, A.; Dyhl-Polk, A.; Chen, I.; Nielsen, D. Checkpoint inhibitors in pancreatic cancer. Cancer Treat. Rev. 2019, 78, 17–30. [Google Scholar] [CrossRef] [PubMed]
  197. Chen, S.H.; Hung, W.C.; Wang, P.; Paul, C.; Konstantopoulos, K. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci. Rep. 2013, 3, 1870. [Google Scholar] [CrossRef]
  198. Marshall, H.T.; Djamgoz, M.B.A. Immuno-Oncology: Emerging Targets and Combination Therapies. Front. Oncol. 2018, 8, 315. [Google Scholar] [CrossRef]
  199. Chen, W.; Yuan, Y.; Jiang, X. Antibody and antibody fragments for cancer immunotherapy. J. Control Release 2020, 328, 395–406. [Google Scholar] [CrossRef]
  200. Bian, J.; Almhanna, K. Pancreatic cancer and immune checkpoint inhibitors-still a long way to go. Transl. Gastroenterol. Hepatol. 2021, 6, 6. [Google Scholar] [CrossRef]
  201. Tran, L.C.; Ozdemir, B.C.; Berger, M.D. The Role of Immune Checkpoint Inhibitors in Metastatic Pancreatic Cancer: Current State and Outlook. Pharmaceuticals 2023, 16, 1411. [Google Scholar] [CrossRef]
  202. Smyth, M.J.; Ngiow, S.F.; Ribas, A.; Teng, M.W. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 2016, 13, 143–158. [Google Scholar] [CrossRef]
  203. 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]
  204. Sugiura, D.; Shimizu, K.; Maruhashi, T.; Okazaki, I.M.; Okazaki, T. T-cell-intrinsic and -extrinsic regulation of PD-1 function. Int. Immunol. 2021, 33, 693–698. [Google Scholar] [CrossRef] [PubMed]
  205. Galluzzi, L.; Buque, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015, 28, 690–714. [Google Scholar] [CrossRef] [PubMed]
  206. Song, D.; Yang, X.; Guo, X.; Sun, H. Safety and efficacy analysis of PD-1 inhibitors in combination with chemotherapy for advanced pancreatic cancer. Immunotherapy 2022, 14, 1307–1313. [Google Scholar] [CrossRef]
  207. Lutz, E.R.; Wu, A.A.; Bigelow, E.; Sharma, R.; Mo, G.; Soares, K.; Solt, S.; Dorman, A.; Wamwea, A.; Yager, A.; et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol. Res. 2014, 2, 616–631. [Google Scholar] [CrossRef]
  208. Steinman, R.M.; Swanson, J. The endocytic activity of dendritic cells. J. Exp. Med. 1995, 182, 283–288. [Google Scholar] [CrossRef] [PubMed]
  209. 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]
  210. Rong, Y.; Qin, X.; Jin, D.; Lou, W.; Wu, L.; Wang, D.; Wu, W.; Ni, X.; Mao, Z.; Kuang, T.; et al. A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin. Exp. Med. 2012, 12, 173–180. [Google Scholar] [CrossRef]
  211. Katsuda, M.; Miyazawa, M.; Ojima, T.; Katanuma, A.; Hakamada, K.; Sudo, K.; Asahara, S.; Endo, I.; Ueno, M.; Hara, K.; et al. A double-blind randomized comparative clinical trial to evaluate the safety and efficacy of dendritic cell vaccine loaded with WT1 peptides (TLP0-001) in combination with S-1 in patients with advanced pancreatic cancer refractory to standard chemotherapy. Trials 2019, 20, 242. [Google Scholar] [CrossRef]
  212. Yoon, J.H.; Jung, Y.J.; Moon, S.H. Immunotherapy for pancreatic cancer. World J. Clin. Cases 2021, 9, 2969–2982. [Google Scholar] [CrossRef]
  213. Dranoff, G. Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 2004, 4, 11–22. [Google Scholar] [CrossRef] [PubMed]
  214. Floros, T.; Tarhini, A.A. Anticancer Cytokines: Biology and Clinical Effects of Interferon-alpha2, Interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin. Oncol. 2015, 42, 539–548. [Google Scholar] [CrossRef] [PubMed]
  215. Dougan, M.; Ingram, J.R.; Jeong, H.J.; Mosaheb, M.M.; Bruck, P.T.; Ali, L.; Pishesha, N.; Blomberg, O.; Tyler, P.M.; Servos, M.M.; et al. Targeting Cytokine Therapy to the Pancreatic Tumor Microenvironment Using PD-L1-Specific VHHs. Cancer Immunol. Res. 2018, 6, 389–401. [Google Scholar] [CrossRef]
  216. Conry, R.M.; Westbrook, B.; McKee, S.; Norwood, T.G. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum. Vaccin. Immunother. 2018, 14, 839–846. [Google Scholar] [CrossRef] [PubMed]
  217. Silk, A.W.; Margolin, K. Cytokine Therapy. Hematol. Oncol. Clin. North Am. 2019, 33, 261–274. [Google Scholar] [CrossRef]
  218. Burugu, S.; Dancsok, A.R.; Nielsen, T.O. Emerging targets in cancer immunotherapy. Semin. Cancer Biol. 2018, 52, 39–52. [Google Scholar] [CrossRef]
  219. Donini, C.; D’Ambrosio, L.; Grignani, G.; Aglietta, M.; Sangiolo, D. Next generation immune-checkpoints for cancer therapy. J. Thorac. Dis. 2018, 10, S1581–S1601. [Google Scholar] [CrossRef]
  220. Marin-Acevedo, J.A.; Dholaria, B.; Soyano, A.E.; Knutson, K.L.; Chumsri, S.; Lou, Y. Next generation of immune checkpoint therapy in cancer: New developments and challenges. J. Hematol. Oncol. 2018, 11, 39. [Google Scholar] [CrossRef]
  221. Zhu, Y.H.; Zheng, J.H.; Jia, Q.Y.; Duan, Z.H.; Yao, H.F.; Yang, J.; Sun, Y.W.; Jiang, S.H.; Liu, D.J.; Huo, Y.M. Immunosuppression, immune escape, and immunotherapy in pancreatic cancer: Focused on the tumor microenvironment. Cell. Oncol. 2023, 46, 17–48. [Google Scholar] [CrossRef]
  222. He, Y.; Rivard, C.J.; Rozeboom, L.; Yu, H.; Ellison, K.; Kowalewski, A.; Zhou, C.; Hirsch, F.R. Lymphocyte-activation gene-3, an important immune checkpoint in cancer. Cancer Sci. 2016, 107, 1193–1197. [Google Scholar] [CrossRef]
  223. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  224. Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef]
  225. Inthagard, J.; Edwards, J.; Roseweir, A.K. Immunotherapy: Enhancing the efficacy of this promising therapeutic in multiple cancers. Clin. Sci. 2019, 133, 181–193. [Google Scholar] [CrossRef] [PubMed]
  226. Herbst, R.S.; Baas, P.; Kim, D.W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.Y.; Molina, J.; Kim, J.H.; Arvis, C.D.; Ahn, M.J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef]
  227. Qiao, G.; Chen, M.; Bucsek, M.J.; Repasky, E.A.; Hylander, B.L. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front. Immunol. 2018, 9, 164. [Google Scholar] [CrossRef] [PubMed]
  228. Huang, S.; Apasov, S.; Koshiba, M.; Sitkovsky, M. Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood 1997, 90, 1600–1610. [Google Scholar] [CrossRef] [PubMed]
  229. Bono, M.R.; Fernandez, D.; Flores-Santibanez, F.; Rosemblatt, M.; Sauma, D. CD73 and CD39 ectonucleotidases in T cell differentiation: Beyond immunosuppression. FEBS Lett. 2015, 589, 3454–3460. [Google Scholar] [CrossRef]
  230. Chambers, A.M.; Wang, J.; Lupo, K.B.; Yu, H.; Atallah Lanman, N.M.; Matosevic, S. Adenosinergic Signaling Alters Natural Killer Cell Functional Responses. Front. Immunol. 2018, 9, 2533. [Google Scholar] [CrossRef]
  231. Hausler, S.F.; Montalban del Barrio, I.; Strohschein, J.; Chandran, P.A.; Engel, J.B.; Honig, A.; Ossadnik, M.; Horn, E.; Fischer, B.; Krockenberger, M.; et al. Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunol. Immunother. 2011, 60, 1405–1418. [Google Scholar] [CrossRef]
  232. Ohta, A.; Kini, R.; Ohta, A.; Subramanian, M.; Madasu, M.; Sitkovsky, M. The development and immunosuppressive functions of CD4+ CD25+ FoxP3+ regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012, 3, 190. [Google Scholar] [CrossRef] [PubMed]
  233. Di Gennaro, P.; Gerlini, G.; Caporale, R.; Sestini, S.; Brandani, P.; Urso, C.; Pimpinelli, N.; Borgognoni, L. T regulatory cells mediate immunosuppresion by adenosine in peripheral blood, sentinel lymph node and TILs from melanoma patients. Cancer Lett. 2018, 417, 124–130. [Google Scholar] [CrossRef] [PubMed]
  234. Roh, M.; Wainwright, D.A.; Wu, J.D.; Wan, Y.; Zhang, B. Targeting CD73 to augment cancer immunotherapy. Curr. Opin. Pharmacol. 2020, 53, 66–76. [Google Scholar] [CrossRef] [PubMed]
  235. Xia, C.; Yin, S.; To, K.K.W.; Fu, L. CD39/CD73/A2AR pathway and cancer immunotherapy. Mol. Cancer 2023, 22, 44. [Google Scholar] [CrossRef]
  236. Catalan, D.; Mansilla, M.A.; Ferrier, A.; Soto, L.; Oleinika, K.; Aguillon, J.C.; Aravena, O. Immunosuppressive Mechanisms of Regulatory B Cells. Front. Immunol. 2021, 12, 611795. [Google Scholar] [CrossRef]
  237. Mirlekar, B.; Wang, Y.; Li, S.; Zhou, M.; Entwistle, S.; De Buysscher, T.; Morrison, A.; Herrera, G.; Harris, C.; Vincent, B.G.; et al. Balance between immunoregulatory B cells and plasma cells drives pancreatic tumor immunity. Cell Rep. Med. 2022, 3, 100744. [Google Scholar] [CrossRef]
  238. Philip, P.A. Targeting B cells in pancreatic adenocarcinoma: Does RESOLVE resolve the question? Ann. Oncol. 2021, 32, 582–583. [Google Scholar] [CrossRef]
  239. Li, S.; Mirlekar, B.; Johnson, B.M.; Brickey, W.J.; Wrobel, J.A.; Yang, N.; Song, D.; Entwistle, S.; Tan, X.; Deng, M.; et al. STING-induced regulatory B cells compromise NK function in cancer immunity. Nature 2022, 610, 373–380. [Google Scholar] [CrossRef]
  240. Michaud, D.; Steward, C.R.; Mirlekar, B.; Pylayeva-Gupta, Y. Regulatory B cells in cancer. Immunol. Rev. 2021, 299, 74–92. [Google Scholar] [CrossRef]
  241. Zhu, H.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Liang, C.; Hua, J.; Meng, Q.; Yu, X.; Shi, S. Intratumoral CD38+ CD19+ B cells associate with poor clinical outcomes and immunosuppression in patients with pancreatic ductal adenocarcinoma. eBioMedicine 2024, 103, 105098. [Google Scholar] [CrossRef]
  242. Geng, X.; Chen, H.; Zhao, L.; Hu, J.; Yang, W.; Li, G.; Cheng, C.; Zhao, Z.; Zhang, T.; Li, L.; et al. Cancer-Associated Fibroblast (CAF) Heterogeneity and Targeting Therapy of CAFs in Pancreatic Cancer. Front. Cell Dev. Biol. 2021, 9, 655152. [Google Scholar] [CrossRef] [PubMed]
  243. Mhaidly, R.; Mechta-Grigoriou, F. Fibroblast heterogeneity in tumor micro-environment: Role in immunosuppression and new therapies. Semin. Immunol. 2020, 48, 101417. [Google Scholar] [CrossRef]
  244. Cho, H.; Seo, Y.; Loke, K.M.; Kim, S.W.; Oh, S.M.; Kim, J.H.; Soh, J.; Kim, H.S.; Lee, H.; Kim, J.; et al. Cancer-Stimulated CAFs Enhance Monocyte Differentiation and Protumoral TAM Activation via IL6 and GM-CSF Secretion. Clin. Cancer Res. 2018, 24, 5407–5421. [Google Scholar] [CrossRef]
  245. Nakamura, K.; Smyth, M.J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell. Mol. Immunol. 2020, 17, 1–12. [Google Scholar] [CrossRef]
  246. Ostrand-Rosenberg, S.; Fenselau, C. Myeloid-Derived Suppressor Cells: Immune-Suppressive Cells That Impair Antitumor Immunity and Are Sculpted by Their Environment. J. Immunol. 2018, 200, 422–431. [Google Scholar] [CrossRef] [PubMed]
  247. Li, T.; Liu, T.; Zhu, W.; Xie, S.; Zhao, Z.; Feng, B.; Guo, H.; Yang, R. Targeting MDSC for Immune-Checkpoint Blockade in Cancer Immunotherapy: Current Progress and New Prospects. Clin. Med. Insights Oncol. 2021, 15, 11795549211035540. [Google Scholar] [CrossRef]
  248. Li, L.; Yu, R.; Cai, T.; Chen, Z.; Lan, M.; Zou, T.; Wang, B.; Wang, Q.; Zhao, Y.; Cai, Y. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int. Immunopharmacol. 2020, 88, 106939. [Google Scholar] [CrossRef]
  249. Medvedeva, G.F.; Kuzmina, D.O.; Nuzhina, J.; Shtil, A.A.; Dukhinova, M.S. How Macrophages Become Transcriptionally Dysregulated: A Hidden Impact of Antitumor Therapy. Int. J. Mol. Sci. 2021, 22, 2662. [Google Scholar] [CrossRef]
  250. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  251. Dong, L.; Chen, C.; Zhang, Y.; Guo, P.; Wang, Z.; Li, J.; Liu, Y.; Liu, J.; Chang, R.; Li, Y.; et al. The loss of RNA N6-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8+ T cell dysfunction and tumor growth. Cancer Cell 2021, 39, 945–957.e10. [Google Scholar] [CrossRef]
  252. Seifert, A.M.; Eymer, A.; Heiduk, M.; Wehner, R.; Tunger, A.; von Renesse, J.; Decker, R.; Aust, D.E.; Welsch, T.; Reissfelder, C.; et al. PD-1 Expression by Lymph Node and Intratumoral Regulatory T Cells Is Associated with Lymph Node Metastasis in Pancreatic Cancer. Cancers 2020, 12, 2756. [Google Scholar] [CrossRef] [PubMed]
  253. Whiteside, T.L. FOXP3+ Treg as a therapeutic target for promoting anti-tumor immunity. Expert. Opin. Ther. Targets 2018, 22, 353–363. [Google Scholar] [CrossRef]
  254. Keeley, T.; Costanzo-Garvey, D.L.; Cook, L.M. Unmasking the Many Faces of Tumor-Associated Neutrophils and Macrophages: Considerations for Targeting Innate Immune Cells in Cancer. Trends Cancer 2019, 5, 789–798. [Google Scholar] [CrossRef]
  255. Hu, F.; Lou, N.; Jiao, J.; Guo, F.; Xiang, H.; Shang, D. Macrophages in pancreatitis: Mechanisms and therapeutic potential. Biomed. Pharmacother. 2020, 131, 110693. [Google Scholar] [CrossRef] [PubMed]
  256. Masamune, A.; Shimosegawa, T. Pancreatic stellate cells: A dynamic player of the intercellular communication in pancreatic cancer. Clin. Res. Hepatol. Gastroenterol. 2015, 39, S98–S103. [Google Scholar] [CrossRef]
  257. Wang, X.; Sun, B.; Wang, Y.; Gao, P.; Song, J.; Chang, W.; Xiao, Z.; Xi, Y.; Li, Z.; An, F.; et al. Research progress of targeted therapy regulating Th17/Treg balance in bone immune diseases. Front. Immunol. 2024, 15, 1333993. [Google Scholar] [CrossRef] [PubMed]
  258. Eisenstein, E.M.; Williams, C.B. The Treg/Th17 cell balance: A new paradigm for autoimmunity. Pediatr. Res. 2009, 65, 26R–31R. [Google Scholar] [CrossRef]
  259. Sivori, S.; Pende, D.; Quatrini, L.; Pietra, G.; Della Chiesa, M.; Vacca, P.; Tumino, N.; Moretta, F.; Mingari, M.C.; Locatelli, F.; et al. NK cells and ILCs in tumor immunotherapy. Mol. Asp. Med. 2021, 80, 100870. [Google Scholar] [CrossRef]
  260. Wu, S.Y.; Fu, T.; Jiang, Y.Z.; Shao, Z.M. Natural killer cells in cancer biology and therapy. Mol. Cancer 2020, 19, 120. [Google Scholar] [CrossRef]
  261. Pastor, F. Aptamers: A New Technological Platform in Cancer Immunotherapy. Pharmaceuticals 2016, 9, 64. [Google Scholar] [CrossRef]
  262. Soldevilla, M.M.; Villanueva, H.; Pastor, F. Aptamers: A Feasible Technology in Cancer Immunotherapy. J. Immunol. Res. 2016, 2016, 1083738. [Google Scholar] [CrossRef]
  263. Munzar, J.D.; Ng, A.; Juncker, D. Duplexed aptamers: History, design, theory, and application to biosensing. Chem. Soc. Rev. 2019, 48, 1390–1419. [Google Scholar] [CrossRef] [PubMed]
  264. Halder, A.; Sun, Y. Biocompatible propulsion for biomedical micro/nano robotics. Biosens. Bioelectron. 2019, 139, 111334. [Google Scholar] [CrossRef]
  265. Yousefi, M.; Dehghani, S.; Nosrati, R.; Zare, H.; Evazalipour, M.; Mosafer, J.; Tehrani, B.S.; Pasdar, A.; Mokhtarzadeh, A.; Ramezani, M. Aptasensors as a new sensing technology developed for the detection of MUC1 mucin: A review. Biosens. Bioelectron. 2019, 130, 1–19. [Google Scholar] [CrossRef] [PubMed]
  266. Li, Q.; Maier, S.H.; Li, P.; Peterhansl, J.; Belka, C.; Mayerle, J.; Mahajan, U.M. Aptamers: A novel targeted theranostic platform for pancreatic ductal adenocarcinoma. Radiat. Oncol. 2020, 15, 189. [Google Scholar] [CrossRef]
  267. Zhu, G.; Chen, X. Aptamer-based targeted therapy. Adv. Drug Deliv. Rev. 2018, 134, 65–78. [Google Scholar] [CrossRef]
  268. Benaduce, A.P.; Brenneman, R.; Schrand, B.; Pollack, A.; Gilboa, E.; Ishkanian, A. 4-1BB Aptamer-Based Immunomodulation Enhances the Therapeutic Index of Radiation Therapy in Murine Tumor Models. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 458–461. [Google Scholar] [CrossRef] [PubMed]
  269. Huang, B.-T.; Lai, W.-Y.; Chang, Y.-C.; Wang, J.-W.; Yeh, S.-D.; Lin, E.P.-Y.; Yang, P.-C. A CTLA-4 Antagonizing DNA Aptamer with Antitumor Effect. Mol. Ther.—Nucleic Acids 2017, 8, 520–528. [Google Scholar] [CrossRef]
  270. Soldevilla, M.M.; Hervas, S.; Villanueva, H.; Lozano, T.; Rabal, O.; Oyarzabal, J.; Lasarte, J.J.; Bendandi, M.; Inoges, S.; López-Díaz de Cerio, A.; et al. Identification of LAG3 high affinity aptamers by HT-SELEX and Conserved Motif Accumulation (CMA). PLoS ONE 2017, 12, e0185169. [Google Scholar] [CrossRef]
  271. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef]
  272. Nishino, M.; Ramaiya, N.H.; Hatabu, H.; Hodi, F.S. Monitoring immune-checkpoint blockade: Response evaluation and biomarker development. Nat. Rev.—Clin. Oncol. 2017, 14, 655–668. [Google Scholar] [CrossRef] [PubMed]
  273. Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 2017, 17, 97–111. [Google Scholar] [CrossRef]
  274. Ventola, C.L. Cancer Immunotherapy, Part 3: Challenges and Future Trends. P T—A Peer-Rev. J. Formul. Manag. 2017, 42, 514–521. [Google Scholar]
  275. Ventola, C.L. Cancer Immunotherapy, Part 2: Efficacy, Safety, and Other Clinical Considerations. P T 2017, 42, 452–463. [Google Scholar] [PubMed]
  276. Evans, R.A.; Diamond, M.S.; Rech, A.J.; Chao, T.; Richardson, M.W.; Lin, J.H.; Bajor, D.L.; Byrne, K.T.; Stanger, B.Z.; Riley, J.L.; et al. Lack of immunoediting in murine pancreatic cancer reversed with neoantigen. JCI Insight 2016, 1, e88328. [Google Scholar] [CrossRef]
  277. Guo, M.N.; Jalil, A.; Liu, J.Y.; Miao, R.Y.; Tran, T.A.; Guan, J. Tongue swelling as a manifestation of tongue metastasis from pulmonary sarcomatoid carcinoma: A case report. World J. Clin. Oncol. 2021, 12, 282–289. [Google Scholar] [CrossRef]
  278. Balachandran, V.P.; Beatty, G.L.; Dougan, S.K. Broadening the Impact of Immunotherapy to Pancreatic Cancer: Challenges and Opportunities. Gastroenterology 2019, 156, 2056–2072. [Google Scholar] [CrossRef]
Figure 1. The tumor microenvironment (TME) in pancreatic cancer (PC): The TME in PC plays a critical role in tumor progression through various mechanisms. The desmoplastic stroma, comprising cancer-associated fibroblasts and the extracellular matrix, facilitates cancer growth and progression both directly and indirectly. Additionally, immunosuppressor cells, such as Treg and tumor-associates macrophages (TAM), inhibit CD8+ T cells, which are pivotal in the anti-tumor immune response, thereby creating immunosuppressive TME.
Figure 1. The tumor microenvironment (TME) in pancreatic cancer (PC): The TME in PC plays a critical role in tumor progression through various mechanisms. The desmoplastic stroma, comprising cancer-associated fibroblasts and the extracellular matrix, facilitates cancer growth and progression both directly and indirectly. Additionally, immunosuppressor cells, such as Treg and tumor-associates macrophages (TAM), inhibit CD8+ T cells, which are pivotal in the anti-tumor immune response, thereby creating immunosuppressive TME.
Medicina 61 01776 g001
Figure 2. The immune system in pancreatic cancer. Various immune cells, including macrophages (M1 and M2), monocytes, dendritic cells, natural killer cells (NK cells), regulatory T cells (Treg), and neutrophils, modulate PC.
Figure 2. The immune system in pancreatic cancer. Various immune cells, including macrophages (M1 and M2), monocytes, dendritic cells, natural killer cells (NK cells), regulatory T cells (Treg), and neutrophils, modulate PC.
Medicina 61 01776 g002
Figure 3. Immunotherapy for pancreatic tumors. Various therapeutic approaches for the treatment of PC are shown. The blockade of Immune checkpoints using anti-PD-1, anti-PD-L1, and/or anti-CTLA-4 agents facilitates the activation of T cells, representing a promising approach for the immunotherapy of pancreatic tumors. Ipilimumab and tremelimumab are fully humanized mAbs targeting anti-CTLA-4 IgG1 and IgG2, respectively, and are approved for clinical application. Therapeutic cancer vaccines, which present immunogenic cancer antigens to the immune system, can induce in vivo activation of cytotoxic T lymphocytes specific to cancer antigens, thereby eliciting an anti-cancer immune response. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is expressed by pancreatic tumor cells engineered to produce GVAX, a whole tumor cell vaccine. PDAC is characterized by a dense stromal/desmoplastic response, involving a multitude of diverse cells, including fibroblasts, immune cells, PSCs, extracellular matrix, and numerous soluble proteins, including growth factors and cytokines. Stromal synthesis may account for 90% of the overall tumor volume. The most clinically advanced form of adoptive cell transfer (ACT) is chimeric antigen receptor (CAR) T-cell therapy, wherein the T cells of clinical participants are genetically modified to express CAR on their surface.
Figure 3. Immunotherapy for pancreatic tumors. Various therapeutic approaches for the treatment of PC are shown. The blockade of Immune checkpoints using anti-PD-1, anti-PD-L1, and/or anti-CTLA-4 agents facilitates the activation of T cells, representing a promising approach for the immunotherapy of pancreatic tumors. Ipilimumab and tremelimumab are fully humanized mAbs targeting anti-CTLA-4 IgG1 and IgG2, respectively, and are approved for clinical application. Therapeutic cancer vaccines, which present immunogenic cancer antigens to the immune system, can induce in vivo activation of cytotoxic T lymphocytes specific to cancer antigens, thereby eliciting an anti-cancer immune response. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is expressed by pancreatic tumor cells engineered to produce GVAX, a whole tumor cell vaccine. PDAC is characterized by a dense stromal/desmoplastic response, involving a multitude of diverse cells, including fibroblasts, immune cells, PSCs, extracellular matrix, and numerous soluble proteins, including growth factors and cytokines. Stromal synthesis may account for 90% of the overall tumor volume. The most clinically advanced form of adoptive cell transfer (ACT) is chimeric antigen receptor (CAR) T-cell therapy, wherein the T cells of clinical participants are genetically modified to express CAR on their surface.
Medicina 61 01776 g003
Figure 4. DCs as a cancer vaccine targeting PC cells. DCs can be loaded with tumor-specific antigens and exosomal antigens. In addition, the fusion of tumor cell lysates with DCs, as well as WT1- or MUC-1-targeted DCs, can be produced. Following the administration of the DC vaccine, the released tumor-specific antigens are presented in the bloodstream by APCs to helper T cells, thereby initiating cell-mediated immunity against pancreatic tumor cells. DCs directed against MUC-1 have the capability to bind directly to the MUC-1 receptors present on the tumor cells, thus eliciting an immune response.
Figure 4. DCs as a cancer vaccine targeting PC cells. DCs can be loaded with tumor-specific antigens and exosomal antigens. In addition, the fusion of tumor cell lysates with DCs, as well as WT1- or MUC-1-targeted DCs, can be produced. Following the administration of the DC vaccine, the released tumor-specific antigens are presented in the bloodstream by APCs to helper T cells, thereby initiating cell-mediated immunity against pancreatic tumor cells. DCs directed against MUC-1 have the capability to bind directly to the MUC-1 receptors present on the tumor cells, thus eliciting an immune response.
Medicina 61 01776 g004
Figure 5. Immunosuppression. CD73 is expressed in various cell types within the TME and serves as a significant immunosuppressive agent by generating extracellular adenosine. Adenosine, through its interaction with A2AR and A2BR receptors on diverse immune cells, impairs the function of immune cells that defend against tumors (such as effector T cells, NK cells, DCs, and B cells), while enhancing the activity of immune cells that suppress the immune response (such as Treg cells, TAMs, MDSCs, and CAFs).
Figure 5. Immunosuppression. CD73 is expressed in various cell types within the TME and serves as a significant immunosuppressive agent by generating extracellular adenosine. Adenosine, through its interaction with A2AR and A2BR receptors on diverse immune cells, impairs the function of immune cells that defend against tumors (such as effector T cells, NK cells, DCs, and B cells), while enhancing the activity of immune cells that suppress the immune response (such as Treg cells, TAMs, MDSCs, and CAFs).
Medicina 61 01776 g005
Table 1. Immunotherapies for pancreatic cancer clinical trials.
Table 1. Immunotherapies for pancreatic cancer clinical trials.
PhaseNCTSettingsInterventionsOutcomesReference
IINCT03190265Second-line or laterNivolumab +GVAX + CRS-207 +
Ipilimumab + Cyclophosphamide
vs. Nivolumab + CRS-207 + Ipilimumab
The study did not meet its primary endpoint of improvement in overall survival.[98]
I/IINCT02305186NeoadjuvantChemoradiotherapy (with Capecitabine)Adding pembrolizumab to neoadjuvant chemoradiotherapy was safe. However, no convincing effect on CD8+ TILs was observed.[140]
IINCT03727880Neoadjuvant/AdjuvantPembrolizumab + Defactinib vs.
Pembrolizumab
Pembrolizumab combined with defactinib was associated with lower fibroblast infiltration, higher anti-tumor M1 macrophage expression and increased CD8+ T-cell infiltration.[141]
IINCT03161379NeoadjuvantNivolumab + Cyclophosphamide + GVAX + SBRTObserved median OS, pCR and R0 resection rates were comparable to contemporary studies administering neoadjuvant mFOLFIRINOX and SBRT.[142]
IINCT03563248NeoadjuvantFOLFIRINOX → SBRT → Surgery
vs.
FOLFIRINOX + Losartan → SBRT + Losartan → Surgery
Downstaging of locally advanced pancreatic ductal adenocarcinoma.[143]
IINCT04940286NeoadjuvantOleclumab + Durvalumab +
Gemcitabina + Nab-Paclitaxel
Standard neoadjuvant therapy has the potential to improve outcomes in PDAC.[144]
IINCT05093231First linePembrolizumab + OlaparibPARP-inhibitor combinations, vaccines, and CAR-T-cells therapy provide some encouraging results.[145]
IINCT02648282First linePembrolizumab + Cyclophosphamide + GVAX + SBRTPhase II study of 54 pts w LAPC treated w CY/GVAX/pembro and SBRT. Primary endpoint of DMFS > 13.6 mos not reached, however 44% of pts underwent surgical resection of whom 42% had grade 1 path response rate.[146]
IIbNCT02907099First linePembrolizumab + CXCR4 antagonist BL-8040The increased T cell infiltration, CXCR4 antagonism was in fact associated with enrichment of CD206hiIA/IElo macrophage subtypes and modestly dampened efficacy[147]
IIINCT03977272First lineAnti-PD-1 antibody 200 mg + mFOLFIRINOX vs. mFOLFIRINOXSintilimab to mFFX improved ORR in advanced PDAC patients significantly, however no superior OS and PFS were observed.[148]
IINCT04377048First lineNivolumab + Gemcitabine + Tegafur-Gimeracil-OteracilAdding-on nivolumab was associated with improved OS in patients with advanced PDAC.[149]
IINCT04543071First lineCemiplimab, Motixafortide,
Gemcitabine, Nab-Paclitaxel
Preliminary results from this pilot study of MCGN in mPDAC were promising, with a durable PR rate of 55% and disease control rate (DCR) of 82%, compared to historic PRs and DCRs of 23% and 48% reported with gemcitabine and nab-paclitaxel (GN), respectively.[150]
IINCT04177810First lineCemiplimab + PlerixaforMobilization of myeloid cells by CXCR4 antagonism results in the recruitment of additional myeloid cells from circulation and that alternative chemokine signaling pathways.[151]
IINCT04493060First lineDostarlimab + NiraparibPARPi plus anti-PD1 checkpoint inhibition was not sufficiently active as later line therapy in metastatic pancreatic cancer for the majority of patients with HRD mutations.[152]
I/IINCT04827953First lineZalifrelimab + Gemcitabine + Nab-Paclitaxe + NLM-001Reduced hypoxia and cancer cell contents in all pt and reduction in CAF, Tregs and macrophages in one pt.[153]
IINCT05014776Second-line or laterPembrolizumab + Ipilimumab + Tadalafil + CRS-207PDE5 inhibition combined with vaccine-based immunotherapy promotes pro-inflammatory states of myeloid cells, activation of T cells, and enhanced myeloid/T cell crosstalk to yield antitumor efficacy against immune-resistant PDAC.[154]
I/IINCT04247165Locally advancedNivolumab + Gemcitabine + SBRT + Nab-Paclitaxel + IpilimumabThe combination was associated with good local control, low adverse event rate, and good QoL.[155]
Table 2. TME and immunosuppression of pancreatic cancer.
Table 2. TME and immunosuppression of pancreatic cancer.
Target CellsRecruitment & ActivationStromal CellsCell Derived ComponentsImmune ResponsesReferences
Tregs, CD8+ T, TAMs, NK cells, tumor cellsDifferentiated from fibroblasts, MSCs, adipocytes and PSCsCAFsCCL, IL-6, TGF-β, CXCL, MCP-1, IDO, PGE2.Recruitment of Tregs and MDSCs, suppression of T cell and NK cell activity, along with heightened PD-L1 expression.[242,243,244]
NK cells, CD8+ T, CD4+, and TregsVEGF, CXCL, CSF, IL, TGF-β, TNF-α, IFN-γ and PEG-2.MDSCsArginase, iNOS, TGF-β, ROS, IDO, COX2, IL-6 and IL-10Inhibition of lymphocyte function, recruitment of Tregs, and expression of immunosuppressive checkpoint molecules.[245,246,247,248]
Tumor cells, CD8+ T, TregsCXCL, CCL, TLR4, VEGF, IL-4, IL-13TAMsTNF I, FN-γ, iNOS, MHCII, ARG1, IL-10, PGE2, EGF, EGFR, TGF-β IL-10, CD163 and CD204Causing Treg differentiation, preventing T cell activity, generating inhibitory cytokines, and raising the expression of CTLA-4 and PD-1.[249,250,251]
CD8+ T, Th cells, NK cells, and APCsCXCR3, CCL9/10/11, CXCL10, CCR4-CCL17/22 and CCR8-CCL1Treg cellsIL-10, PD-L1, TCF-β, CTLA4, MADCAM-1, VCAM-1, granzyme B and perforinImmunosuppressive cytokine secretion, NK cell apoptosis induction and immune cell function inhibition.[252,253]
Tregs, CAFs, and TAMsCXCL, TGF-β, IFN-β and GM-CSFTANsIL-13, CCL17, CCL2, ARG1, elastase and MMP9Advancing the polarization of TAMs, Treg recruitment, and the up-regulation of ARG1 and PD-1.[7,254]
Immune cells, tumor cells, TAMs, MDSCsinterleukin and TGF-β,PSCsIL-10, CXCL12, MCP-1, VEGF, fibronectin and type I collagenEncouraging the differentiation and migration of MDSCs and TAMs, resulting in an imbalance of Th1/Th2 cytokines[255,256]
T cells, Tregs, Th17.Recruitment is inhibited by PGE2DCsCD80, CD40, CD70, CD86, MHC-I, MHC-II, IFN-γ, IL-12, IL-15, MGL2 and PD-L2.Impairment of DC activation, maturation and the display of antigens; Encouraging the multiplication of Treg cells while hindering immunity mediated by CD8+ T cells; controlling the equilibrium between Th17 and Treg cells.[257,258]
T cells, DCs, macrophages.CD47, HLA-G, CCL27/CCR10, CCL5/CCR5, CX3CL1/CX3CR1, ECMNK cellsGM-CSF, IFN-γ, TNF-α, IL-3, perforin and granzymeMDSCs and Treg cells prevent NK cell toxicity through TGF-β and inhibitory signals.[259,260]
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

Thankachan, S.V.; Jayaraman, V.; Datta, L.; Apthi, S.; Zaman, B.F.; Gurunathan, R.; Suresh, A.; Chandrakesan, P.; Vinayagam, R.; Kang, S.G.; et al. Tumor Microenvironment: Recent Advances in Immunotherapies of Pancreatic Cancer. Medicina 2025, 61, 1776. https://doi.org/10.3390/medicina61101776

AMA Style

Thankachan SV, Jayaraman V, Datta L, Apthi S, Zaman BF, Gurunathan R, Suresh A, Chandrakesan P, Vinayagam R, Kang SG, et al. Tumor Microenvironment: Recent Advances in Immunotherapies of Pancreatic Cancer. Medicina. 2025; 61(10):1776. https://doi.org/10.3390/medicina61101776

Chicago/Turabian Style

Thankachan, Sharon Varghese, Vijayalakshmi Jayaraman, Liza Datta, Soniga Apthi, Binish Fatima Zaman, Raghav Gurunathan, Anuppama Suresh, Parthasarathy Chandrakesan, Ramachandran Vinayagam, Sang Gu Kang, and et al. 2025. "Tumor Microenvironment: Recent Advances in Immunotherapies of Pancreatic Cancer" Medicina 61, no. 10: 1776. https://doi.org/10.3390/medicina61101776

APA Style

Thankachan, S. V., Jayaraman, V., Datta, L., Apthi, S., Zaman, B. F., Gurunathan, R., Suresh, A., Chandrakesan, P., Vinayagam, R., Kang, S. G., Palaniyandi, K., & Gnanasampanthapandian, D. (2025). Tumor Microenvironment: Recent Advances in Immunotherapies of Pancreatic Cancer. Medicina, 61(10), 1776. https://doi.org/10.3390/medicina61101776

Article Metrics

Back to TopTop