- 1Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- 2Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
- 3First College for Clinical Medicine, Xuzhou Medical University, Jiangsu, Xuzhou, China
- 4Department of Gastroenterology, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
Pancreatic cancer is projected to become the second leading cause of cancer−related death by 2030. Conventional interventions including surgery, radiotherapy, and chemotherapy provide only modest survival benefits, underscoring an urgent need for more effective therapies. Although immunotherapy has revolutionized the management of several solid tumors, its clinical benefit in pancreatic cancer has so far been disappointing. Mounting evidence indicates that a highly immunosuppressive tumor microenvironment (TME), dominated by tumor−associated macrophages (TAMs), myeloid−derived suppressor cells (MDSCs), and regulatory T cells (Tregs), drives immune evasion, tumor progression, metastasis, and chemoresistance through complex cytokine and chemokine networks. This review summarizes current knowledge of these immunosuppressive mechanisms and provides emerging strategies aimed at re−educating or depleting these cellular constituents to enhance the efficacy of immunotherapy in pancreatic cancer.
1 Introduction
Pancreatic cancer is a highly malignant digestive system tumor with subtle and non-specific clinical symptoms, making early diagnosis difficult. By 2030, it is expected to become the second leading cause of cancer-related deaths globally (1). Traditional treatments like surgery, radiation, and chemotherapy are limited in efficacy (2, 3), contributing to poor prognosis (4, 5). Immunotherapy has shown promise in treating various cancers, but clinical trials for pancreatic cancer have not met expectations. One key challenge is the immunosuppressive tumor microenvironment (TME), which plays a critical role in the tumor’s initiation, development, and prognosis. The TME of pancreatic cancer is characterized by immune cell infiltration (6, 7), primarily of immunosuppressive cells such as pancreatic stellate cells, regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) (8), and tumor-associated macrophages (TAMs) (9). These cells secrete immunosuppressive molecules that inhibit the function of anti-tumor immune cells, promote immune evasion, and enhance tumor progression and metastasis (1, 10, 11). This review summarizes the mechanisms of the immunosuppressive components in the pancreatic cancer TME, aiming to provide insights into its immunotherapy.
2 The role of immune cells in the pancreatic cancer TME
Pancreatic cancer evades immunity via MHC I downregulation, inhibiting CD8+ T cell activation (12–14). Tumor-specific neoantigens may fail to trigger immune responses due to the TME’s immunosuppressive effects (15, 16). Additionally, TGF-β and IDO secreted by cancer cells further impair immune function (17, 18), with Tregs, MDSCs, and TAMs contributing to immune suppression in early stages (19–21).
2.1 The role of TAMs in pancreatic cancer
2.1.1 TAMs promote tumor inflammation and immune evasion
TAMs secrete cytokines and chemokines, such as CCL18, which upregulates VCAM-1 in pancreatic cancer via the CCL18/PITPNM3/NF-κB/VCAM-1 pathway, promoting tumor progression (22). They also release IL-10, IL-12, and CCL13, mediating Th2 responses and suppressing T-cell immunity (23). In mouse models, TAM-produced IL-6 and TNF drive inflammation, while IL-6/STAT3 inhibition reduces inflammatory cell infiltration (24). TAMs activate TLR-6/TLR-2 via Versican, expressing inflammation-related genes (25). CTCF promotes pancreatic cancer progression through FLG-AS1-mediated epigenetic mechanisms and macrophage polarization (26). TAMs facilitate immune evasion by secreting IL-10, inducing T cell apoptosis via CD120a/b (27, 28), altering tumor cell phenotypes, and overexpressing B7-H3 via EGFR/MAPK, inhibiting CD8+ T cells (29). Arginase I expression depletes L-arginine, suppressing T cell receptors (30), while the hypoxic, low-glucose TME polarizes macrophages to M2-like phenotypes, further impairing T cell function (31).
2.1.2 TAMs promote tumor metastasis and chemoresistance
Tumor-associated macrophages (TAMs) play multifaceted roles in tumor progression through diverse molecular mechanisms. These immune cells facilitate epithelial-mesenchymal transition (EMT) and enhance metastatic potential via TLR4/IL-10 signaling, while simultaneously inducing matrix metalloproteinases (MMP-2/9) through MIP-3α to promote pancreatic ductal adenocarcinoma (PDAC) invasion (32, 33). Under hypoxic conditions, TAMs activate the PI3Kγ/PTEN pathway and upregulate HIF-1/2α, leading to increased production of pro-angiogenic factors (VEGF, TNF-α, IL-1β) and metastasis-promoting mediators (34–36). TAMs produce EGF and VEGF-A, aiding tumor cell circulation entry. β-catenin-driven TAMs enhance metastasis via OSM/STAT3/LOXL2 (37). The TYROBP-mediated M2 polarization further exacerbates these pro-tumoral effects (38). TAMs express HIF-1α, upregulating VEGF, TNF-α, IL-1β, IL-8, PDGF, bFGF, thymidine phosphorylase, and MMPs, promoting angiogenesis (39). VEGF-A recruits VEGFR2+ macrophages, forming TAMs (40). In PDAC, hypoxia increases HIF-1/2 in TAMs, upregulating TGF-β and NRF2 to induce VEGF-A (41). Vasohibin-1 is regulated by TGF-β/BMP signaling between TAMs and tumor cells (42). Notably, TAMs significantly contribute to therapeutic resistance in PDAC through multiple pathways: (1) promoting dense stromal formation and IGF/IGF1R activation (43); (2) enhancing EMT-mediated drug evasion (44); (3) driving gemcitabine resistance via TGF-β1/Gfi-1 signaling, which can be attenuated by simvastatin (45); and (4) fostering immunotherapy resistance through CREB3L1-mediated TAM reprogramming within the tumor microenvironment (46).
2.2 The role of tumor-associated neutrophils in pancreatic cancer
2.2.1 TANs regulate tumor immunity in pancreatic cancer
TANs regulate immune responses in PDAC through chemokines and cytokines (47), impairing CD8+ T cell infiltration and function. Nectin2+ and OLR1+ TAN phenotypes are associated with T-cell exhaustion. ER stress regulates TAN protumor activities (48). IL-17-induced neutrophil extracellular traps (NETs) contribute to resistance to immune checkpoint inhibitors (ICIs). CXCR2 and its ligands, such as CXCL5, are crucial for TAN recruitment in pancreatic cancer (49). Immunotargeting neutrophils can restore anti-tumor immunity in pancreatic cancer, improving therapeutic outcomes by addressing immune evasion mechanisms.
2.2.2 TANs regulate pancreatic cancer proliferation and metastasis
TANs in the TME can polarize into N1 and N2 phenotypes. Interferon-β promotes N1 polarization, enhancing anti-tumor immunity, while TGF-β and G-CSF induce N2 polarization, supporting tumor growth (50–52). In pancreatic cancer, TANs secrete a proliferation-inducing ligand (APRIL) (53), which was indicated to plays a role in promoting the progression of pancreatic cancer (54). The interaction of TIMP1 with its receptor CD63 activates the ERK pathway, enhancing NETs formation and tumor proliferation (55). PADI4, a key enzyme driving NET formation, accelerates pancreatic cancer growth. In PADI4 knockout mice, tumor growth is slower, and deoxyribonuclease treatment reduces cancer growth by inhibiting NETs (56).
TME fibrosis and collagen deposition facilitate metastasis, with discoidin domain receptor 1 (DDR1) signaling via NF-κB inducing CXCL5 production, recruiting TANs, and enhancing NET formation (57, 58). NETs trap cancer cells, shield them from immune attack, and induce EMT via PADI4 and elastase translocation, promoting metastasis (59). NETs also enhance liver metastasis by recruiting CAFs and hematopoietic stem cells (60). Consistent with this, dense TAN−derived NET lattices have been visualized within hepatic sinusoids before overt metastatic seeding; these structures trap circulating pancreatic cancer cells, facilitate their extravasation, and attract immunosuppressive macrophages, thereby functionally linking NET formation to both immune suppression and metastatic colonization (61, 62). TANs secrete CCL5, promoting cancer cell migration and CD8+ T-cell dysfunction via Nectin2 upregulation (48). Angiogenesis is critical for tumor progression. CXCL5/CXCR2 blockade inhibits tumor growth and angiogenesis via activation of the protein kinase B (Akt), extracellular signal-regulated kinase (ERK) pathways. CXCL8 and CXCL12 synergistically enhance endothelial cell migration and proliferation, with MMP-2 activation further promoting angiogenesis (63). Targeting angiogenesis-related factors is a promising research direction.
2.2.3 TANs influence chemoresistance in pancreatic cancer
TANs contribute to chemoresistance, with growth arrest-specific protein 6 (Gas6) from neutrophils promoting cancer cell regeneration via the Gas6/AXL pathway (64). G-CSF enhances neutrophil recruitment and resistance to anti-VEGF therapy, while MEK inhibition reduces G-CSF production and synergizes with anti-VEGF drugs (65). N2 TANs interfere with antigen-presenting cell (APC) maturation, leading to resistance in CD40-targeted therapies (66). Targeting TANs may improve chemotherapy efficacy in pancreatic cancer. IL-17 induced NETs, which play a key role in the resistance to ICIs in pancreatic cancer (67).
2.3 Tumor-infiltrating lymphocytes in pancreatic cancer
TILs, including CD4+ Th cells, CD8+ CTLs, and Tregs, are pivotal in the TME. High CD8+ CTL infiltration correlates with improved survival, as these cells induce tumor cell apoptosis via MHC I-dependent perforin, granzyme, TNF, and IFN-γ release (68, 69), though IL-18 receptor signaling impedes their migration (70). Conversely, Tregs suppress antitumor immunity; lipid synthesis inhibition in Tregs enhances immune responses (71). Moncada et al. (72) linked pancreatic cancer cell states to TME composition, revealing clinical implications. Foxp3, a Treg regulator (73), drives immunosuppression via TGF-β secretion, dendritic cell suppression, and CD8+ T-cell inhibition (74–76). Treg levels rise from precancerous lesions to adenocarcinoma, associating with metastasis and advanced staging (77). In tumors, Tregs co-infiltrate with CD4+/CD8+ T cells but suppress CTL activity, promoting immune evasion (78, 79). Tregs interact with (1) CD20+ B cells (worse prognosis), (2) CD3+CD56+ NKT cells, and (3) CD68+CD163+ macrophages, influencing immune polarization (80). B regulatory cells (Bregs) are elevated in PDAC and linked to progression (81). PDAC cells and Bregs mutually activate via IL-18, with Bregs expressing PD-L1/IL-35 to suppress CD8+ T-cell proliferation and IFN-γ production (81–85). Dual IL-18/PD-L1 blockade reduces tumor growth in models, highlighting Breg-cancer crosstalk as a therapeutic target (81).
2.4 MDSCs in pancreatic cancer
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells that play a critical role in tumor-mediated immune suppression. Although scarcely present in normal pancreatic tissue, these cells accumulate significantly in pathological conditions including pancreatic intraepithelial neoplasia and chronic pancreatitis (86, 87). In advanced pancreatic cancer, tumor-derived factors and inflammatory mediators promote MDSC recruitment and activation, leading to their substantial expansion in the bone marrow, peripheral circulation, and tumor microenvironment (88–90). The immunosuppressive functions of MDSCs are mediated through multiple interconnected mechanisms. These cells generate reactive oxygen species (ROS) in response to cytokines such as TGF-β, IL-10, and IL-6, creating an oxidative environment that impairs immune cell function within the TME. Furthermore, MDSCs express high levels of arginase and nitric oxide synthase, which deplete essential amino acids and disrupt critical signaling pathways including JAK3 and STAT5, ultimately leading to T cell dysfunction and apoptosis. The production of peroxynitrite by MDSCs causes nitration of T cell receptor (TCR) and CD8 molecules, thereby compromising antigen recognition capacity (91).
MDSCs also promote immune tolerance through indirect mechanisms. Under IFN-γ stimulation, they secrete IL-10 and TGF-β to drive regulatory T cell (Treg) differentiation (92) Additionally, MDSCs upregulate PD-L1 expression to directly inhibit T cell activity while simultaneously reducing L-selectin expression, which impairs T cell homing and activation (91). Preclinical studies demonstrate that MDSC depletion in pancreatic cancer models enhances T cell infiltration, suppresses tumor progression, and improves survival outcomes, highlighting these cells as promising therapeutic targets (88). Besides, KRAS mutations occur in 90% of PDAC cases (93), driving tumorigenesis and progression (94). These alterations shape an immunosuppressive TME by expanding MDSCs and depleting dendritic cells, undermining antitumor immunity (95). Combining KRAS inhibition with I/O therapies may thus overcome resistance.
2.5 Inflammatory mediators in pancreatic cancer
The TME is integral to tumorigenesis, development, and metastasis, comprising cancer cells, stromal cells, cytokines, and inflammatory mediators (96–98). Inflammation is closely linked to cancer, with conditions like obesity and diabetes being risk factors for PDAC, supported by the concept of “parainflammation” (99). Tumor cells secrete chemokines to recruit inflammatory cells, as demonstrated by Makoto Sano et al. (100), showing that the CXCR2-CXCL5 axis accelerates invasion and migration in PDAC. CXCR2-dependent regulation modulates the formation, angiogenesis, and metastasis of pancreatic cancer, with CXCR2 also promoting immune evasion and cancer development through autocrine effects, while recent experiments show that cell-autonomous CXCL5 maintains tolerance loops and stromal inflammation by inducing TNF derived from neutrophils in cancer cells (101, 102). Cytokines such as TGF-β, IL-6, IL-10, and TNF-α play critical roles in tumor progression and metastasis. TGF-β enhances invasion via EMT, IL-6 activates JAK2-STAT3 to promote angiogenesis, and TNF-α induces metastasis by activating fibroblasts and promoting VEGF production in PDAC (99) (Figure 1).
2.6 Exosomes in pancreatic cancer
Exosomes secreted by both eukaryotic and prokaryotic cells contain RNA, DNA, proteins, lipids, and sugars (103–105). In pancreatic cancer, exosomes transfer signaling molecules that promote tumor proliferation, differentiation, and metastasis (106). For example, miR-3960 inhibits TFAP2A in exosomes, counteracting their promotive effects (107). Exosomes, enriched with miRNAs like miR-10b, miR-550, and miR-1246, serve as biomarkers for early diagnosis (108). Exosomes also deliver siRNA targeting oncogenes, like KrasG12D, more effectively than liposomes (109). Exosomes, linked to tumor metastasis, are secreted in the hypoxic microenvironment and enriched with miRNA-301a-3p, phosphatases, and the angiotensin II/PI3Kγ signaling pathway, which induce HIF1-α or HIF2-α to stimulate M2 macrophage polarization, thereby promoting tumor cell metastasis (34, 110). Exosomes derived from hypoxic pancreatic neuroendocrine tumors (PNETs) contain CEACAM5, which facilitates the polarization of TAMs towards the M2 phenotype, thereby enhancing tumor metastasis through the activation of MMP9 (111). Additionally, hypoxic tumor-derived exosomal miR-4488 induces M2 polarization in macrophages, promoting liver metastasis of pancreatic neuroendocrine tumors via the RTN3/FABP5 axis, which drives fatty acid oxidation (112). Exosomes can also influence metastasis and invasiveness via signaling pathways like circ-PDE8A (113). Thus, exosomes have significant clinical value, warranting further exploration in pancreatic and other cancers.
3 Therapies targeting immune suppressive cells
3.1 Targeting TAMs in treatment
Tumor aggressiveness is influenced by TAM location and quantity. Inhibiting the CCL2/CCR2 axis reduces TAMs TAM recruitment, promotes M1 polarization, and suppresses M2 phenotypes (114, 115). Clinical evidence highlights carlumab (anti-CCL2) for pancreatic cancer (116) and PF-04136309 (CCR2 antagonist) in preclinical/clinical settings (117, 118). CCR2 or CSF-1R targeting augments chemotherapy, curbs metastasis, and amplifies T-cell activity (119). Ultrasound-mediated CSF1/CSF1R blockade depletes macrophages, showing therapeutic potential (120). Clodronate liposomes deplete TAMs, increasing IFN-γ+ CD8+ T-cell infiltration in PDAC (121). Macrophage repolarization (M2→M1) via LPS, IFN-γ, TLR4 agonists, or paclitaxel/Nab-paclitaxel enhances immunity (122–124). NF-κB inhibition reprograms TAMs, potentially through IFN-γ/CCL2, improving anti-tumor responses (125). CD40 agonists with gemcitabine remodel PDAC’s immune landscape, activating T cells (126). Histamine-rich glycoproteins induce M1 polarization, normalize vasculature, and restore CD8+ T-cell function via PI3Kγ inhibition (126, 127). IL-27, produced by activated macrophages, activates JAK-STAT, shifting TAMs from M2 to M1, inhibiting tumor growth, and enhancing gemcitabine efficacy (128). Natural molecules like sphingosine (129) and Urolithin A (130) also modulate M2 polarization in PDAC. Targeting PI3Kγ inhibitors offer new therapies, which enhances macrophage efferocytosis in pancreatic cancer, supporting tumor control when combined with radiotherapy (131). Besides, the phase Ib/II study NCT03767582, which tests the dual CCR2/CCR5 antagonist BMS−813160 together with nivolumab (± GVAX) in locally−advanced PDAC show enhanced intratumoral CD8+CD137+ T-cells and manageable toxicity, while preclinical data confirmed CCR2 blockade synergizes with anti-PD-1 by increasing CD8+ T-cell infiltration and reducing T-regs in PDAC (132).
3.2 Targeting TANs in treatment
Current immune therapies targeting TANs focus on inhibiting TAN recruitment via cytokine/chemokine axes. CXCR2 deficiency reduces pancreatic cancer vascular density (101), while blocking CXCR2-CXCL8 enhances PD-1 efficacy (133). Lorlatinib inhibits G-CSF, reduces TAN recruitment, and boosts CD8+ T/NK cell cytotoxicity (134). HMGB1 from NETs promotes malignancy, despite thrombomodulin degrades HMGB1 (135). Neutralizing IL-1β inhibits EGFR/ERK activation and EMT (136), while NET targeting mitigates hypercoagulability and thrombosis (137). Inhibiting specific TAN phenotypes: P2RX1-negative neutrophils in liver metastases correlate with PD-L1 expression; their inhibition activates CD8+ T cell anti-tumor immunity, suppressing pancreatic cancer progression.
3.3 Adoptive TILs cell therapy
Since the 1980s, TILs adoptive cell therapy has evolved, involving extraction, in vitro culture, and reinfusion into patients. Rosenberg et al. (138) reported 34%-56% efficacy in melanoma, while its potential in pancreatic cancer remains unexplored but promising. Sakellariou-Thompson et al. (139) demonstrated that CD8+ TILs from pancreatic cancer can grow with 4-1BB agonists, supporting clinical feasibility. Targeting immunosuppressive cells, such as CCR4+ Tregs in melanoma, shows promise, with CCR4 antibodies depleting Tregs in vivo and in vitro (140). Mogamulizumab, an anti-CCR4 antibody, is in clinical trials. Bacterial therapy, like Salmonella A1-R, enhances CD8+ TILs in pancreatic cancer models, suggesting anti-tumor immunity activation (141, 142). Chemotherapy combined with innate immune agonists improves T cell priming for ICIs (143) (144–146). TNFR2 blockade in PDAC targets Tregs, reducing immunosuppression and T cell exhaustion (147). eIF4G1 overexpression in PDAC correlates with poor prognosis; its inhibition reduces pro-tumor cytokines, promotes M2-TAM polarization (148), and enhances CD8+ T cell recruitment (149–151), offering a therapeutic strategy (152).
3.4 Anti-CTLA-4/PD-1 therapy
Anti-CTLA-4 and anti-PD-1 antibodies are ICIs that activate CD8+ T cell responses. CTLA-4, identified as a T cell checkpoint factor by KRUMMEL et al. (153), but ICIs show limited efficacy in pancreatic cancer due to resistance (154). PD-1 treatment’s effectiveness remains controversial, though PDL-1 overexpression may predict response. Combined anti-PD-L1 and anti-CTLA-4 therapy showed promise in pancreatic neuroendocrine tumors (154). Deng et al. (155) found glucocorticoid receptor (GR) inhibition downregulates PD-L1 and upregulates MHC-I, enhancing immune therapy sensitivity. Other immunosuppressive pathways (TIM3, TIGIT, LAG3, VISTA, CD73) are highly expressed in PDAC (156), suggesting potential therapeutic targets. Oncolytic virotherapy (OVs) using modified viruses (e.g., adenoviruses, herpes simplex viruses) combined with ICIs shows promise, particularly in head and neck squamous cell carcinoma (157). Hyperthermia, as a sensitizer, enhances immune activation and, when combined with gemcitabine, reduces invasion and metastasis in pancreatic cancer cells by promoting apoptosis via reactive oxygen species (158).
3.5 Other immune suppressive cells targeted therapies
Exosome-based dual delivery systems, such as iEXO-OXA, enhance immune responses in orthotopic PDAC mice by inducing immunogenic cell death (ICD) and reversing immune suppression. These exosomes improve drug accumulation in tumors while minimizing systemic distribution, promoting innate and adaptive anti-PDAC immunity by enhancing ICD, dendritic cell maturation, and cytotoxic T lymphocyte infiltration (159). GVAX, a GM-CSF gene-transfected pancreatic tumor cell vaccine, combined with low-dose cyclophosphamide, induces anti-tumor immunity, including Treg depletion and tertiary lymphoid structure formation, improving immune cell infiltration in the TME. The combination with nivolumab and urelumab shows promising efficacy in resectable pancreatic cancer (160–162).
4 Conclusion
Pancreatic cancer remains a formidable challenge due to its immunosuppressive TME, which facilitates tumor progression, metastasis, and resistance to conventional therapies. The intricate interplay between immune cells, stromal components, and inflammatory mediators creates a hostile environment that limits the efficacy of current treatments. Immunosuppressive cells, as key players in this process, promote immune evasion, angiogenesis, and chemoresistance through diverse mechanisms, including cytokine secretion, NET formation, and stromal remodeling. Novel treatment approaches, including TAM/TAN-directed interventions, adoptive transfer of tumor-infiltrating lymphocytes, blockade of immune checkpoints, and engineered exosome platforms, present viable solutions to circumvent these limitations. However, the complexity of the TME necessitates a multifaceted approach, combining these therapies with conventional treatments to enhance anti-tumor immunity and improve patient outcomes. Future research should focus on elucidating the molecular mechanisms underlying immune suppression in pancreatic cancer and developing innovative, targeted therapies to reprogram the TME and restore effective immune surveillance.
Author contributions
DP: Writing – original draft. XL: Writing – original draft. XQ: Writing – original draft, Writing – review & editing. QW: Writing – original draft, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This research was supported by the Huai’an City Municipal Science and Technology Plan (No. HAB202210).
Acknowledgments
We appreciate the support from the Wenzhou Central Hospital and Xuzhou Medical University for this study.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Fan JQ, Wang MF, Chen HL, Shang D, Das JK, and Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer. (2020) 19:32. doi: 10.1186/s12943-020-01151-3
2. Li Z, Zhou H, Xia Z, Xia T, Du G, Franziska SD, et al. HMGA1 augments palbociclib efficacy via PI3K/mTOR signaling in intrahepatic cholangiocarcinoma. Biomark Res. (2023) 11:33. doi: 10.1186/s40364-023-00473-w
3. Zhai X, Xia Z, Du G, Zhang X, Xia T, Ma D, et al. LRP1B suppresses HCC progression through the NCSTN/PI3K/AKT signaling axis and affects doxorubicin resistance. Genes Dis. (2023) 10:2082–96. doi: 10.1016/j.gendis.2022.10.021
4. Jemal A, Siegel R, Xu J, and Ward E. Cancer statistics, 2010. CA Cancer J Clin. (2010) 60:277–300. doi: 10.3322/caac.20073
5. Zhang X, Zhang P, Cong A, Feng Y, Chi H, Xia Z, et al. Unraveling molecular networks in thymic epithelial tumors: deciphering the unique signatures. Front Immunol. (2023) 14:1264325. doi: 10.3389/fimmu.2023.1264325
6. Xu S, Liang J, Shen T, Zhang D, and Lu Z. Causal links between immune cells and asthma: Insights from a Mendelian Randomization analysis. J Asthma. (2025) 62:346–53. doi: 10.1080/02770903.2024.2403740
7. Xu S and Lu Z. Exploring FNDC4 as a biomarker for prognosis and immunotherapy response in lung adenocarcinoma. Asian J Surg. (2024). doi: 10.1016/j.asjsur.2024.09.054
8. Deng Y, Shi M, Yi L, Naveed Khan M, Xia Z, and Li X. Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment. Heliyon. (2024) 10:e37060. doi: 10.1016/j.heliyon.2024.e37060
9. Vitorakis N, Gargalionis AN, Papavassiliou KA, Adamopoulos C, and Papavassiliou AG. Precision targeting strategies in pancreatic cancer: the role of tumor microenvironment. Cancers (Basel). (2024) 16:2876. doi: 10.3390/cancers16162876
10. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2018) 68:394–424. doi: 10.3322/caac.21492
11. Liu X, Xi X, Xu S, Chu H, Hu P, Li D, et al. Targeting T cell exhaustion: emerging strategies in non-small cell lung cancer. Front Immunol. (2024) 15:1507501. doi: 10.3389/fimmu.2024.1507501
12. Chang JH, Jiang Y, and Pillarisetty VG. Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Med (Baltimore). (2016) 95:e5541. doi: 10.1097/MD.0000000000005541
13. Ryschich E, Notzel T, Hinz U, Autschbach F, Ferguson J, Simon I, et al. Control of T-cell-mediated immune response by HLA class I in human pancreatic carcinoma. Clin Cancer Res. (2005) 11:498–504. doi: 10.1158/1078-0432.498.11.2
14. Xie H, Xi X, Lei T, Liu H, and Xia Z. CD8(+) T cell exhaustion in the tumor microenvironment of breast cancer. Front Immunol. (2024) 15:1507283. doi: 10.3389/fimmu.2024.1507283
15. Schumacher TN and Schreiber RD. Neoantigens in cancer immunotherapy. Science. (2015) 348:69–74. doi: 10.1126/science.aaa4971
16. Bailey P, Chang DK, Forget MA, Lucas FA, Alvarez HA, Haymaker C, et al. Exploiting the neoantigen landscape for immunotherapy of pancreatic ductal adenocarcinoma. Sci Rep. (2016) 6:35848. doi: 10.1038/srep35848
17. Moo-Young TA, Larson JW, Belt BA, Tan MC, Hawkins WG, Eberlein TJ, et al. Tumor-derived TGF-beta mediates conversion of CD4+Foxp3+ regulatory T cells in a murine model of pancreas cancer. J Immunother. (2009) 32:12–21. doi: 10.1097/CJI.0b013e318189f13c
18. Azimnasab-Sorkhabi P, Soltani-Asl M, Yoshinaga TT, Zaidan Dagli ML, Massoco CO, and Kfoury Junior JR. Indoleamine-2,3 dioxygenase: a fate-changer of the tumor microenvironment. Mol Biol Rep. (2023) 50:6133–45. doi: 10.1007/s11033-023-08469-3
19. Hashimoto A and Hashimoto S. Plasticity and tumor microenvironment in pancreatic cancer: genetic, metabolic, and immune perspectives. Cancers (Basel). (2024) 16:4094. doi: 10.3390/cancers16234094
20. Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. (2003) 9:1269–74. doi: 10.1038/nm934
21. Falcomata C, Barthel S, Schneider G, Rad R, Schmidt-Supprian M, and Saur D. Context-specific determinants of the immunosuppressive tumor microenvironment in pancreatic cancer. Cancer Discov. (2023) 13:278–97. doi: 10.1158/2159-8290.CD-22-0876
22. Ye H, Zhou Q, Zheng S, Li G, Lin Q, Wei L, et al. Tumor-associated macrophages promote progression and the Warburg effect via CCL18/NF-kB/VCAM-1 pathway in pancreatic ductal adenocarcinoma. Cell Death Dis. (2018) 9:453. doi: 10.1038/s41419-018-0486-0
23. Yang L and Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J Hematol Oncol. (2017) 10:58. doi: 10.1186/s13045-017-0430-2
24. Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Kloppel G, et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. (2011) 19:456–69. doi: 10.1016/j.ccr.2011.03.009
25. Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. (2009) 457:102–6. doi: 10.1038/nature07623
26. Liu Y, Liu P, Duan S, Lin J, Qi W, Yu Z, et al. CTCF enhances pancreatic cancer progression via FLG-AS1-dependent epigenetic regulation and macrophage polarization. Cell Death Differ. (2024) 32:745–62. doi: 10.1038/s41418-024-01423-1
27. D’Alincourt Salazar M, Manuel ER, Tsai W, D’Apuzzo M, Goldstein L, Blazar BR, et al. Evaluation of innate and adaptive immunity contributing to the antitumor effects of PD1 blockade in an orthotopic murine model of pancreatic cancer. Oncoimmunology. (2016) 5:e1160184. doi: 10.1080/2162402X.2016.1160184
28. Saio M, Radoja S, Marino M, and Frey AB. Tumor-infiltrating macrophages induce apoptosis in activated CD8(+) T cells by a mechanism requiring cell contact and mediated by both the cell-associated form of TNF and nitric oxide. J Immunol. (2001) 167:5583–93. doi: 10.4049/jimmunol.167.10.5583
29. Zhang Y, Velez-Delgado A, Mathew E, Li D, Mendez FM, Flannagan K, et al. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut. (2017) 66:124–36. doi: 10.1136/gutjnl-2016-312078
30. Kusmartsev S and Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. (2005) 174:4880–91. doi: 10.4049/jimmunol.174.8.4880
31. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumor-associated macrophages by tumor-derived lactic acid. Nature. (2014) 513:559–63. doi: 10.1038/nature13490
32. Kimsey TF, Campbell AS, Albo D, Wilson M, and Wang TN. Co-localization of macrophage inflammatory protein-3alpha (Mip-3alpha) and its receptor, CCR6, promotes pancreatic cancer cell invasion. Cancer J. (2004) 10:374–80. doi: 10.1097/00130404-200411000-00007
33. Liu CY, Xu JY, Shi XY, Huang W, Ruan TY, Xie P, et al. M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab Invest. (2013) 93:844–54. doi: 10.1038/labinvest.2013.69
34. Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, et al. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kgamma to Promote Pancreatic Cancer Metastasis. Cancer Res. (2018) 78:4586–98. doi: 10.1158/0008-5472.CAN-17-3841
35. Sensi B, Angelico R, Toti L, Conte L, Coppola A, Tisone G, et al. Mechanism, potential, and concerns of immunotherapy for hepatocellular carcinoma and liver transplantation. Curr Mol Pharmacol. (2024) 17:e18761429310703. doi: 10.2174/0118761429310703240823045808
36. Chen J, Lin A, and Luo P. Advancing pharmaceutical research: A comprehensive review of cutting-edge tools and technologies. Current Pharmaceutical Analysis. (2024) 21:1–19. doi: 10.1016/j.cpan.2024.11.001
37. Zhang Y, Zhu X, Chen L, Gao T, Chen G, Zhu J, et al. beta-Catenin mediated TAM phenotype promotes pancreatic cancer metastasis via the OSM/STAT3/LOXL2 axis. Neoplasia. (2025) 60:101096. doi: 10.1016/j.neo.2024.101096
38. Zhong D, Liao Y, Chen W, Huang X, Liu J, and Wang Z. TYROBP promotes the spread of pancreatic cancer by causing M2 TAM polarization. J Gastroenterol Hepatol. (2024) 39:2926–39. doi: 10.1111/jgh.v39.12
39. Qian BZ and Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. (2010) 141:39–51. doi: 10.1016/j.cell.2010.03.014
40. Dineen SP, Lynn KD, Holloway SE, Miller AF, Sullivan JP, Shames DS, et al. Vascular endothelial growth factor receptor 2 mediates macrophage infiltration into orthotopic pancreatic tumors in mice. Cancer Res. (2008) 68:4340–6. doi: 10.1158/0008-5472.CAN-07-6705
41. Feng R, Morine Y, Ikemoto T, Imura S, Iwahashi S, Saito Y, et al. Nrf2 activation drive macrophages polarization and cancer cell epithelial-mesenchymal transition during interaction. Cell Commun Signal. (2018) 16:54. doi: 10.1186/s12964-018-0262-x
42. Shen Z, Seppanen H, Kauttu T, Vainionpaa S, Ye Y, Wang S, et al. Vasohibin-1 expression is regulated by transforming growth factor-beta/bone morphogenic protein signaling pathway between tumor-associated macrophages and pancreatic cancer cells. J Interferon Cytokine Res. (2013) 33:428–33. doi: 10.1089/jir.2012.0046
43. Ireland L, Santos A, Ahmed MS, Rainer C, Nielsen SR, Quaranta V, et al. Chemoresistance in pancreatic cancer is driven by stroma-derived insulin-like growth factors. Cancer Res. (2016) 76:6851–63. doi: 10.1158/0008-5472.CAN-16-1201
44. Kuwada K, Kagawa S, Yoshida R, Sakamoto S, Ito A, Watanabe M, 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. doi: 10.1186/s13046-018-0981-2
45. Xian G, Zhao J, Qin C, Zhang Z, Lin Y, and Su Z. Simvastatin attenuates macrophage-mediated gemcitabine resistance of pancreatic ductal adenocarcinoma by regulating the TGF-beta1/Gfi-1 axis. Cancer Lett. (2017) 385:65–74. doi: 10.1016/j.canlet.2016.11.006
46. Xu H, Xue S, Sun Y, Ma J, Li S, Wang Y, et al. CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment. J Immunother Cancer. (2025) 13:e010029. doi: 10.1136/jitc-2024-010029
47. Tamassia N, Bianchetto-Aguilera F, Arruda-Silva F, Gardiman E, Gasperini S, Calzetti F, et al. Cytokine production by human neutrophils: Revisiting the “dark side of the moon. Eur J Clin Invest. (2018) 48 Suppl 2:e12952. doi: 10.1111/eci.2018.48.issue-S2
48. Luo H, Ikenaga N, Nakata K, Higashijima N, Zhong P, Kubo A, et al. Tumor-associated neutrophils upregulate Nectin2 expression, creating the immunosuppressive microenvironment in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. (2024) 43:258. doi: 10.1186/s13046-024-03178-6
49. Chao T, Furth EE, and Vonderheide RH. CXCR2-dependent accumulation of tumor-associated neutrophils regulates T-cell immunity in pancreatic ductal adenocarcinoma. Cancer Immunol Res. (2016) 4:968–82. doi: 10.1158/2326-6066.CIR-16-0188
50. Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Kockritz-Blickwede M, et al. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. (2016) 138:1982–93. doi: 10.1002/ijc.v138.8
51. Dou A and Fang J. Heterogeneous myeloid cells in tumors. Cancers (Basel). (2021) 13:3772. doi: 10.3390/cancers13153772
52. Ohms M, Moller S, and Laskay T. An attempt to polarize human neutrophils toward N1 and N2 phenotypes in vitro. Front Immunol. (2020) 11:532. doi: 10.3389/fimmu.2020.00532
53. Wang G, Wang F, Ding W, Wang J, Jing R, Li H, et al. APRIL induces tumorigenesis and metastasis of colorectal cancer cells via activation of the PI3K/Akt pathway. PLoS One. (2013) 8:e55298. doi: 10.1371/journal.pone.0055298
54. Han L, Zhang W, Song F, Guo Y, Guo K, and Zhou W. Soluble a-proliferation-inducing ligand (sAPRIL), a novel serum biomarker predicting the recurrence and metastasis of pancreatic adenocarcinoma after surgery. Mol Med Rep. (2014) 10:1978–84. doi: 10.3892/mmr.2014.2443
55. Schoeps B, Eckfeld C, Prokopchuk O, Bottcher J, Haussler D, Steiger K, et al. TIMP1 triggers neutrophil extracellular trap formation in pancreatic cancer. Cancer Res. (2021) 81:3568–79. doi: 10.1158/0008-5472.CAN-20-4125
56. Miller-Ocuin JL, Liang X, Boone BA, Doerfler WR, Singhi AD, Tang D, et al. 3rd: DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology. (2019) 8:e1605822. doi: 10.1080/2162402X.2019.1605822
57. Korc M. Pancreatic cancer-associated stroma production. Am J Surg. (2007) 194:S84–86. doi: 10.1016/j.amjsurg.2007.05.004
58. Deng J, Kang Y, Cheng CC, Li X, Dai B, Katz MH, et al. DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight. (2021) 6:e146133. doi: 10.1172/jci.insight.146133
59. Kajioka H, Kagawa S, Ito A, Yoshimoto M, Sakamoto S, Kikuchi S, et al. Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Lett. (2021) 497:1–13. doi: 10.1016/j.canlet.2020.10.015
60. Takesue S, Ohuchida K, Shinkawa T, Otsubo Y, Matsumoto S, Sagara A, et al. Neutrophil extracellular traps promote liver micrometastasis in pancreatic ductal adenocarcinoma via the activation of cancer-associated fibroblasts. Int J Oncol. (2020) 56:596–605. doi: 10.3892/ijo.2019.4951
61. Arvanitakis K, Mitroulis I, and Germanidis GJC. Tumor-associated neutrophils in hepatocellular carcinoma pathogenesis. Prognosis Ther. (2021) 13:2899. doi: 10.3390/cancers13122899
62. Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. (2018) 18:134–47. doi: 10.1038/nri.2017.105
63. Matsuo Y, Ochi N, Sawai H, Yasuda A, Takahashi H, Funahashi H, et al. CXCL8/IL-8 and CXCL12/SDF-1alpha co-operatively promote invasiveness and angiogenesis in pancreatic cancer. Int J Cancer. (2009) 124:853–61. doi: 10.1002/ijc.v124:4
64. Bellomo G, Rainer C, Quaranta V, Astuti Y, Raymant M, Boyd E, et al. Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signaling axis. Gut. (2022) 71:2284–99. doi: 10.1136/gutjnl-2021-325272
65. Phan VT, Wu X, Cheng JH, Sheng RX, Chung AS, Zhuang G, et al. Oncogenic RAS pathway activation promotes resistance to anti-VEGF therapy through G-CSF-induced neutrophil recruitment. Proc Natl Acad Sci U S A. (2013) 110:6079–84. doi: 10.1073/pnas.1303302110
66. Siolas D, Vucic E, Kurz E, Hajdu C, and Bar-Sagi D. Gain-of-function p53(R172H) mutation drives accumulation of neutrophils in pancreatic tumors, promoting resistance to immunotherapy. Cell Rep. (2021) 36:109578. doi: 10.1016/j.celrep.2021.109578
67. Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, et al. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. (2020) 217:e20190354. doi: 10.1084/jem.20190354
68. Griesmann H, Drexel C, Milosevic N, Sipos B, Rosendahl J, Gress TM, et al. Pharmacological macrophage inhibition decreases metastasis formation in a genetic model of pancreatic cancer. Gut. (2017) 66:1278–85. doi: 10.1136/gutjnl-2015-310049
69. Lopez JA, Brennan AJ, Whisstock JC, Voskoboinik I, and Trapani JA. Protecting a serial killer: pathways for perforin trafficking and self-defense ensure sequential target cell death. Trends Immunol. (2012) 33:406–12. doi: 10.1016/j.it.2012.04.001
70. Nasiri E, Student M, Roth K, Siti Utami N, Huber M, Buchholz M, et al. IL18 receptor signaling inhibits intratumoral CD8(+) T-cell migration in a murine pancreatic cancer model. Cells. (2023) 12:456. doi: 10.3390/cells12030456
71. Lim SA, Wei J, Nguyen TM, Shi H, Su W, Palacios G, et al. Lipid signaling enforces functional specialization of T(reg) cells in tumors. Nature. (2021) 591:306–11. doi: 10.1038/s41586-021-03235-6
72. Moncada R, Barkley D, Wagner F, Chiodin M, Devlin JC, Baron M, et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat Biotechnol. (2020) 38:333–42. doi: 10.1038/s41587-019-0392-8
73. Chen Z, Lin F, Gao Y, Li Z, Zhang J, Xing Y, et al. FOXP3 and RORγt: transcriptional regulation of Treg and Th17. Int Immunopharmacol. (2011) 11:536–42. doi: 10.1016/j.intimp.2010.11.008
74. Nishikawa H and Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol. (2014) 27:1–7. doi: 10.1016/j.coi.2013.12.005
75. Homma Y, Taniguchi K, Nakazawa M, Matsuyama R, Mori R, Takeda K, et al. Changes in the immune cell population and cell proliferation in peripheral blood after gemcitabine-based chemotherapy for pancreatic cancer. Clin Transl Oncol. (2014) 16:330–5. doi: 10.1007/s12094-013-1079-0
76. Vignali DA, Collison LW, and Workman CJ. How regulatory T cells work. Nat Rev Immunol. (2008) 8:523–32. doi: 10.1038/nri2343
77. Cysneiros M, Cirqueira MB, Barbosa LF, Chaves de Oliveira E, Morais LK, Wastowski IJ, et al. Immune cells and checkpoints in pancreatic adenocarcinoma: Association with clinical and pathological characteristics. PloS One. (2024) 19:e0305648. doi: 10.1371/journal.pone.0305648
78. Cinier J, Hubert M, Besson L, Di Roio A, Rodriguez C, Lombardi V, et al. Recruitment and expansion of tregs cells in the tumor environment-how to target them? Cancers (Basel). (2021) 13:1850. doi: 10.3390/cancers13122899
79. Tang Y, Xu X, Guo S, Zhang C, Tang Y, Tian Y, et al. An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma. PloS One. (2014) 9:e91551. doi: 10.1371/journal.pone.0091551
80. Bayati F, Mohammadi M, Valadi M, Jamshidi S, Foma AM, and Sharif-Paghaleh E. The therapeutic potential of regulatory T cells: challenges and opportunities. Front Immunol. (2020) 11:585819. doi: 10.3389/fimmu.2020.585819
81. Zhao Y, Shen M, Feng Y, He R, Xu X, Xie Y, et al. Regulatory B cells induced by pancreatic cancer cell-derived interleukin-18 promote immune tolerance via the PD-1/PD-L1 pathway. Oncotarget. (2018) 9:14803–14. doi: 10.18632/oncotarget.22976
82. Tong DN, Guan J, Sun JH, Zhao CY, Chen SG, Zhang ZY, et al. Characterization of B cell-mediated PD-1/PD-L1 interaction in pancreatic cancer patients. Clin Exp Pharmacol Physiol. (2020) 47:1342–9. doi: 10.1111/1440-1681.13317
83. Mirlekar B, Michaud D, Lee SJ, Kren NP, Harris C, Greene K, et al. B cell-derived IL35 drives STAT3-dependent CD8(+) T-cell exclusion in pancreatic cancer. Cancer Immunol Res. (2020) 8:292–308. doi: 10.1158/2326-6066.CIR-19-0349
84. Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, and Fallon PG. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun. (2015) 6:5997. doi: 10.1038/ncomms6997
85. Wang Z, Zhao Y, and Zhang L. Emerging trends and hot topics in the application of multi-omics in drug discovery: A bibliometric and visualized study. Current Pharmaceutical Analysis. (2024) 21:20–32. doi: 10.1016/j.cpan.2024.12.001
86. Bronte V, Apolloni E, Cabrelle A, Ronca R, Serafini P, Zamboni P, et al. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood. (2000) 96:3838–46. doi: 10.1182/blood.V96.12.3838
87. Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, and Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. (2007) 67:9518–27. doi: 10.1158/0008-5472.CAN-07-0175
88. Porembka MR, Mitchem JB, Belt BA, Hsieh CS, Lee HM, Herndon J, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother. (2012) 61:1373–85. doi: 10.1007/s00262-011-1178-0
89. Khaled YS, Ammori BJ, and Elkord E. Increased levels of granulocytic myeloid-derived suppressor cells in peripheral blood and tumor tissue of pancreatic cancer patients. J Immunol Res. (2014) 2014:879897. doi: 10.1155/2014/879897
90. Lin A, Jiang A, Huang L, Li Y, Zhang C, Zhu L, et al. From chaos to order: optimizing fecal microbiota transplantation for enhanced immune checkpoint inhibitors efficacy. Gut Microbes. (2025) 17:2452277. doi: 10.1080/19490976.2025.2452277
91. Gabrilovich DI and Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. (2009) 9:162–74. doi: 10.1038/nri2506
92. Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. (2006) 66:1123–31. doi: 10.1158/0008-5472.CAN-05-1299
93. Singhi AD, George B, Greenbowe JR, Chung J, Suh J, Maitra A, et al. Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers. Gastroenterology. (2019) 156:2242–2253 e2244. doi: 10.1053/j.gastro.2019.02.037
94. Bannoura SF, Uddin MH, Nagasaka M, Fazili F, Al-Hallak MN, Philip PA, et al. Targeting KRAS in pancreatic cancer: new drugs on the horizon. Cancer Metastasis Rev. (2021) 40:819–35. doi: 10.1007/s10555-021-09990-2
95. Norgard RJ, Budhani P, O’Brien SA, Xia Y, Egan JN, Flynn B, et al. Reshaping the tumor microenvironment of KRASG12D pancreatic ductal adenocarcinoma with combined SOS1 and MEK inhibition for improved immunotherapy response. Cancer Res Commun. (2024) 4:1548–60. doi: 10.1158/2767-9764.CRC-24-0172
96. Yan J, Ye G, Jin Y, Miao M, Li Q, and Zhou H. Identification of novel prognostic circRNA biomarkers in circRNA-miRNA-mRNA regulatory network in gastric cancer and immune infiltration analysis. BMC Genomics. (2023) 24:323. doi: 10.1186/s12864-023-09421-2
97. Yan J, Yu X, Li Q, Miao M, and Shao Y. Machine learning to establish three sphingolipid metabolism genes signature to characterize the immune landscape and prognosis of patients with gastric cancer. BMC Genomics. (2024) 25:319. doi: 10.1186/s12864-024-10243-z
98. Gu J, Wang Y, Zhang H, Gu H, and Zhu H. SIGLEC1 has the potential to be an immune-related prognostic indicator in colon adenocarcinoma: a study based on transcriptomic data and Mendelian randomization analysis. Discov Oncol. (2025) 16:324. doi: 10.1007/s12672-025-02093-2
99. Padoan A, Plebani M, and Basso D. Inflammation and pancreatic cancer: focus on metabolism, cytokines, and immunity. Int J Mol Sci. (2019) 20:676. doi: 10.3390/ijms20030676
100. Sano M, Ijichi H, Takahashi R, Miyabayashi K, Fujiwara H, Yamada T, et al. Blocking CXCLs-CXCR2 axis in tumor-stromal interactions contributes to survival in a mouse model of pancreatic ductal adenocarcinoma through reduced cell invasion/migration and a shift of immune-inflammatory microenvironment. Oncogenesis. (2019) 8:8. doi: 10.1038/s41389-018-0117-8
101. Purohit A, Saxena S, Varney M, Prajapati DR, Kozel JA, Lazenby A, et al. Host cxcr2-dependent regulation of pancreatic cancer growth, angiogenesis, and metastasis. Am J Pathol. (2021) 191:759–71. doi: 10.1016/j.ajpath.2021.01.002
102. Bianchi A, De Castro Silva I, Deshpande NU, Singh S, Mehra S, Garrido VT, et al. Cell-autonomous cxcl1 sustains tolerogenic circuitries and stromal inflammation via neutrophil-derived TNF in pancreatic cancer. Cancer Discov. (2023) 13:1428–53. doi: 10.1158/2159-8290.CD-22-1046
103. Xiong J, Chi H, Yang G, Zhao S, Zhang J, Tran LJ, et al. Revolutionizing anti-tumor therapy: unleashing the potential of B cell-derived exosomes. Front Immunol. (2023) 14:1188760. doi: 10.3389/fimmu.2023.1188760
104. Gong X, Chi H, Strohmer DF, Teichmann AT, Xia Z, and Wang Q. Exosomes: A potential tool for immunotherapy of ovarian cancer. Front Immunol. (2022) 13:1089410. doi: 10.3389/fimmu.2022.1089410
105. Lou Y, Yan J, Liu Q, Miao M, and Shao Y. Biological functions and molecular mechanisms of exosome-derived circular RNAs and their clinical implications in digestive Malignancies: the vintage in the bottle. Ann Med. (2024) 56:2420861. doi: 10.1080/07853890.2024.2420861
106. Zhang X, Sai B, Wang F, Wang L, Wang Y, Zheng L, et al. Hypoxic BMSC-derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3-induced EMT. Mol Cancer. (2019) 18:40. doi: 10.1186/s12943-019-0959-5
107. Wu J. Pancreatic cancer-derived exosomes promote the proliferation, invasion, and metastasis of pancreatic cancer by the miR-3960/TFAP2A axis. J Oncol. (2022) 2022:3590326. doi: 10.1155/2022/3590326
108. Uddin MH, Al-Hallak MN, Philip PA, Mohammad RM, Viola N, Wagner KU, et al. Exosomal microRNA in pancreatic cancer diagnosis, prognosis, and treatment: from bench to bedside. Cancers (Basel). (2021) 13:2777. doi: 10.3390/cancers13112777
109. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. (2017) 546:498–503. doi: 10.1038/nature22341
110. Wang J, Cao Z, Zhang XM, Nakamura M, Sun M, Hartman J, et al. Novel mechanism of macrophage-mediated metastasis revealed in a zebrafish model of tumor development. Cancer Res. (2015) 75:306–15. doi: 10.1158/0008-5472.CAN-14-2819
111. Ye M, Lu F, Gu D, Xue B, Xu L, Hu C, et al. Hypoxia exosome derived CEACAM5 promotes tumor-associated macrophages M2 polarization to accelerate pancreatic neuroendocrine tumors metastasis via MMP9. FASEB J. (2024) 38:e23762. doi: 10.1096/fj.202302489RRR
112. Lu F, Ye M, Shen Y, Xu Y, Hu C, Chen J, et al. Hypoxic tumor-derived exosomal miR-4488 induces macrophage M2 polarization to promote liver metastasis of pancreatic neuroendocrine neoplasm through RTN3/FABP5 mediated fatty acid oxidation. Int J Biol Sci. (2024) 20:3201–18. doi: 10.7150/ijbs.96831
113. Li Z, Yanfang W, Li J, Jiang P, Peng T, Chen K, et al. Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. (2018) 432:237–50. doi: 10.1016/j.canlet.2018.04.035
114. Sierra-Filardi E, Nieto C, Dominguez-Soto A, Barroso R, Sanchez-Mateos P, Puig-Kroger A, et al. CCL2 shapes macrophage polarization by GM-CSF and M-CSF: identification of CCL2/CCR2-dependent gene expression profile. J Immunol. (2014) 192:3858–67. doi: 10.4049/jimmunol.1302821
115. Zhai X, Zhang H, Xia Z, Liu M, Du G, Jiang Z, et al. Oxytocin alleviates liver fibrosis via hepatic macrophages. JHEP Rep. (2024) 6:101032. doi: 10.1016/j.jhepr.2024.101032
116. Brana I, Calles A, LoRusso PM, Yee LK, Puchalski TA, Seetharam S, et al. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study. Target Oncol. (2015) 10:111–23. doi: 10.1007/s11523-014-0320-2
117. Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res. (2013) 19:3404–15. doi: 10.1158/1078-0432.CCR-13-0525
118. Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, et al. Targeting tumor-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-center, open-label, dose-finding, non-randomized, phase 1b trial. Lancet Oncol. (2016) 17:651–62. doi: 10.1016/S1470-2045(16)00078-4
119. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. (2013) 73:1128–41. doi: 10.1158/0008-5472.CAN-12-2731
120. Wang Q, Wang J, Xu K, and Luo Z. Targeting the CSF1/CSF1R signaling pathway: an innovative strategy for ultrasound combined with macrophage exhaustion in pancreatic cancer therapy. Front Immunol. (2024) 15:1481247. doi: 10.3389/fimmu.2024.1481247
121. Yang X, Lin J, Wang G, and Xu D. Targeting proliferating tumor-infiltrating macrophages facilitates spatial redistribution of CD8(+) T cells in pancreatic cancer. Cancers (Basel). (2022) 14:1474. doi: 10.3390/cancers14061474
122. Prakash H, Nadella V, Singh S, and Schmitz-Winnenthal H. CD14/TLR4 priming potentially recalibrates and exerts anti-tumor efficacy in tumor associated macrophages in a mouse model of pancreatic carcinoma. Sci Rep. (2016) 6:31490. doi: 10.1038/srep31490
123. Zimmer SM, Liu J, Clayton JL, Stephens DS, and Snyder JP. Paclitaxel binding to human and murine MD-2. J Biol Chem. (2008) 283:27916–26. doi: 10.1074/jbc.M802826200
124. Okada KI, Hirono S, Kawai M, Miyazawa M, Shimizu A, Kitahata Y, et al. Phase I study of nab-paclitaxel plus gemcitabine as neoadjuvant therapy for borderline resectable pancreatic cancer. Anticancer Res. (2017) 37:853–8. doi: 10.21873/anticanres.11389
125. Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, Thompson RG, et al. Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med. (2008) 205:1261–8. doi: 10.1084/jem.20080108
126. Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. (2011) 331:1612–6. doi: 10.1126/science.1198443
127. Kaneda MM, Cappello P, Nguyen AV, Ralainirina N, Hardamon CR, Foubert P, et al. Macrophage PI3Kgamma drives pancreatic ductal adenocarcinoma progression. Cancer Discov. (2016) 6:870–85. doi: 10.1158/2159-8290.CD-15-1346
128. Yao L, Wang M, Niu Z, Liu Q, Gao X, Zhou L, et al. Interleukin-27 inhibits Malignant behaviors of pancreatic cancer cells by targeting M2 polarized tumor associated macrophages. Cytokine. (2017) 89:194–200. doi: 10.1016/j.cyto.2015.12.003
129. Wang B, Zheng X, Liu J, Zhang Z, Qiu C, Yang L, et al. Osthole inhibits pancreatic cancer progression by directly exerting negative effects on cancer cells and attenuating tumor-infiltrating M2 macrophages. J Pharmacol Sci. (2018) 137:290–8. doi: 10.1016/j.jphs.2018.07.007
130. Totiger TM, Srinivasan S, Jala VR, Lamichhane P, Dosch AR, Gaidarski AA 3rd, et al. : urolithin A, a novel natural compound to target PI3K/AKT/mTOR pathway in pancreatic cancer. Mol Cancer Ther. (2019) 18:301–11. doi: 10.1158/1535-7163.MCT-18-0464
131. Russell SN, Demetriou C, Valenzano G, Evans A, Go S, Stanly T, et al. Induction of macrophage efferocytosis in pancreatic cancer via PI3Kgamma inhibition and radiotherapy promotes tumor control. Gut. (2025) 74:825–39. doi: 10.1136/gutjnl-2024-333492
132. Christenson E, Lim SJ, Wang H, Ferguson A, Parkinson R, Cetasaan Y, et al. Nivolumab and a CCR2/CCR5 dual antagonist (BMS-813160) with or without GVAX for locally advanced pancreatic ductal adenocarcinomas: Results of phase I study. Am Soc Clin Oncol. (2023) 10:1827. doi: 10.1200/JCO.2023.41.4_suppl.730
133. Zhang M, Huang L, Ding G, Huang H, Cao G, Sun X, et al. Interferon gamma inhibits CXCL8-CXCR2 axis mediated tumor-associated macrophages tumor trafficking and enhances anti-PD1 efficacy in pancreatic cancer. J Immunother Cancer. (2020) 8:e000308. doi: 10.1136/jitc-2019-000308
134. Nielsen SR, Strobech JE, Horton ER, Jackstadt R, Laitala A, Bravo MC, et al. Suppression of tumor-associated neutrophils by lorlatinib attenuates pancreatic cancer growth and improves treatment with immune checkpoint blockade. Nat Commun. (2021) 12:3414. doi: 10.1038/s41467-021-23731-7
135. Bausch D, Pausch T, Krauss T, Hopt UT, Fernandez-del-Castillo C, Warshaw AL, et al. Neutrophil granulocyte derived MMP-9 is a VEGF independent functional component of the angiogenic switch in pancreatic ductal adenocarcinoma. Angiogenesis. (2011) 14:235–43. doi: 10.1007/s10456-011-9207-3
136. Jin W, Yin H, Li H, Yu XJ, Xu HX, and Liu L. Neutrophil extracellular DNA traps promote pancreatic cancer cells migration and invasion by activating EGFR/ERK pathway. J Cell Mol Med. (2021) 25:5443–56. doi: 10.1111/jcmm.v25.12
137. Abdol Razak N, Elaskalani O, and Metharom P. Pancreatic cancer-induced neutrophil extracellular traps: A potential contributor to cancer-associated thrombosis. Int J Mol Sci. (2017) 18:487. doi: 10.3390/ijms18030487
138. Rosenberg SA and Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. (2015) 348:62–8. doi: 10.1126/science.aaa4967
139. Sakellariou-Thompson D, Forget MA, Creasy C, Bernard V, Zhao L, Kim YU, et al. 4-1BB agonist focuses CD8(+) tumor-infiltrating T-cell growth into a distinct repertoire capable of tumor recognition in pancreatic cancer. Clin Cancer Res. (2017) 23:7263–75. doi: 10.1158/1078-0432.CCR-17-0831
140. Sugiyama D, Nishikawa H, Maeda Y, Nishioka M, Tanemura A, Katayama I, et al. Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evoking antitumor immune responses in humans. Proc Natl Acad Sci U S A. (2013) 110:17945–50. doi: 10.1073/pnas.1316796110
141. Hiroshima Y, Zhang Y, Murakami T, Maawy A, Miwa S, Yamamoto M, et al. Efficacy of tumor-targeting Salmonella typhimurium A1-R in combination with anti-angiogenesis therapy on a pancreatic cancer patient-derived orthotopic xenograft (PDOX) and cell line mouse models. Oncotarget. (2014) 5:12346–57. doi: 10.18632/oncotarget.v5i23
142. Murakami T, Hiroshima Y, Zhang Y, Zhao M, Kiyuna T, Hwang HK, et al. Tumor-targeting salmonella typhimurium A1-R promotes tumoricidal CD8(+) T cell tumor infiltration and arrests growth and metastasis in a syngeneic pancreatic-cancer orthotopic mouse model. J Cell Biochem. (2018) 119:634–9. doi: 10.1002/jcb.v119.1
143. Zhang X, Zhuge J, Liu J, Xia Z, Wang H, Gao Q, et al. Prognostic signatures of sphingolipids: Understanding the immune landscape and predictive role in immunotherapy response and outcomes of hepatocellular carcinoma. Front Immunol. (2023) 14:1153423. doi: 10.3389/fimmu.2023.1153423
144. Niu N, Li K, Wang J, Funes V, Espinoza B, Li P, et al. Chemotherapy in synergy with innate immune agonists enhances T cell priming for checkpoint inhibitor treatment in pancreatic cancer. Biomark Res. (2025) 13:21. doi: 10.1186/s40364-024-00721-7
145. Xia Z, Chen S, He M, Li B, Deng Y, Yi L, et al. Editorial: Targeting metabolism to activate T cells and enhance the efficacy of checkpoint blockade immunotherapy in solid tumors. Front Immunol. (2023) 14:1247178. doi: 10.3389/fimmu.2023.1247178
146. Zhang J, Peng G, Chi H, Yang J, Xie X, Song G, et al. CD8 + T-cell marker genes reveal different immune subtypes of oral lichen planus by integrating single-cell RNA-seq and bulk RNA-sequencing. BMC Health. (2023) 23:464. doi: 10.1186/s12903-023-03138-0
147. Debesset A, Pilon C, Meunier S, Cuelenaere-Bonizec O, Richer W, Thiolat A, et al. TNFR2 blockade promotes antitumoral immune response in PDAC by targeting activated Treg and reducing T cell exhaustion. J Immunother Cancer. (2024) 12:e008898. doi: 10.1136/jitc-2024-008898
148. Bhatia R, Bhyravbhatla N, Kisling A, Li X, Batra SK, and Kumar S. Cytokines chattering in pancreatic ductal adenocarcinoma tumor microenvironment. Semin Cancer Biol. (2022) 86:499–510. doi: 10.1016/j.semcancer.2022.03.021
149. Roberto M, Arrivi G, Di Civita MA, Barchiesi G, Pilozzi E, Marchetti P, et al. The role of CXCL12 axis in pancreatic cancer: New biomarkers and potential targets. Front Oncol. (2023) 13:1154581. doi: 10.3389/fonc.2023.1154581
150. Reschke R and Gajewski TF. CXCL9 and CXCL10 bring the heat to tumors. Sci Immunol. (2022) 7:eabq6509. doi: 10.1126/sciimmunol.abq6509
151. Wang Y, Wang J, Liu J, and Zhu H. Immune-related diagnostic markers for benign prostatic hyperplasia and their potential as drug targets. Front Immunol. (2024) 15:1516362. doi: 10.3389/fimmu.2024.1516362
152. He L, Zhang X, Shi F, Zhang H, Chen Y, Sun K, et al. Reprograming immunosuppressive microenvironment by eIF4G1 targeting to eradicate pancreatic ductal adenocarcinoma. Cell Rep Med. (2024) 5:101731. doi: 10.1016/j.xcrm.2024.101731
153. Krummel MF and Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. (1995) 182:459–65. doi: 10.1084/jem.182.2.459
154. Chick RC and Pawlik TM. Updates in immunotherapy for pancreatic cancer. J Clin Med. (2024) 13:6419. doi: 10.3390/jcm13216419
155. Deng Y, Xia X, Zhao Y, Zhao Z, Martinez C, Yin W, et al. Glucocorticoid receptor regulates PD-L1 and MHC-I in pancreatic cancer cells to promote immune evasion and immunotherapy resistance. Nat Commun. (2021) 12:7041. doi: 10.1038/s41467-021-27349-7
156. Balli D, Rech AJ, Stanger BZ, and Vonderheide RH. Immune cytolytic activity stratifies molecular subsets of human pancreatic cancer. Clin Cancer Res. (2017) 23:3129–38. doi: 10.1158/1078-0432.CCR-16-2128
157. Dong H, Li M, Yang C, Wei W, He X, Cheng G, et al. Combination therapy with oncolytic viruses and immune checkpoint inhibitors in head and neck squamous cell carcinomas: an approach of complementary advantages. Cancer Cell Int. (2023) 23:1. doi: 10.1186/s12935-022-02846-x
158. Maurici CE, Colenbier R, Wylleman B, Brancato L, van Zwol E, Van den Bossche J, et al. Hyperthermia enhances efficacy of chemotherapeutic agents in pancreatic cancer cell lines. Biomolecules. (2022) 12:6519. doi: 10.3390/biom12050651
159. Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. (2021) 268:120546. doi: 10.1016/j.biomaterials.2020.120546
160. Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res. (2014) 2:616–31. doi: 10.1158/2326-6066.CIR-14-0027
161. Zheng L, Ding D, Edil BH, Judkins C, Durham JN, Thomas DL 2nd, et al. Vaccine-induced intratumoral lymphoid aggregates correlate with survival following treatment with a neoadjuvant and adjuvant vaccine in patients with resectable pancreatic adenocarcinoma. Clin Cancer Res. (2021) 27:1278–86. doi: 10.1158/1078-0432.CCR-20-2974
162. Heumann T, Judkins C, Li K, Lim SJ, Hoare J, Parkinson R, et al. A platform trial of neoadjuvant and adjuvant antitumor vaccination alone or in combination with PD-1 antagonist and CD137 agonist antibodies in patients with resectable pancreatic adenocarcinoma. Nat Commun. (2023) 14:3650. doi: 10.1038/s41467-023-39196-9
Keywords: pancreatic cancer, tumor microenvironment, regulatory T cells, immune suppression, PD-1
Citation: Pan D, Li X, Qiao X and Wang Q (2025) Immunosuppressive tumor microenvironment in pancreatic cancer: mechanisms and therapeutic targets. Front. Immunol. 16:1582305. doi: 10.3389/fimmu.2025.1582305
Received: 24 February 2025; Accepted: 17 April 2025;
Published: 15 May 2025.
Edited by:
Yongfu Shao, Ningbo University, ChinaReviewed by:
Donglin Zhang, Ludwig Maximilian University of Munich, GermanyCopyright © 2025 Pan, Li, Qiao and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Xiao Qiao, anNoYXFpYW94aWFvQDE2My5jb20=; Qiqi Wang, cWlxaXdhbmcwMTEzQHllYWgubmV0