Next Article in Journal
A Systematic Analysis of Expression and Function of RAS GTPase-Activating Proteins (RASGAPs) in Urological Cancers: A Mini-Review
Next Article in Special Issue
The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies
Previous Article in Journal
Emerging Trends in Global Cancer Epidemiology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now?

by
Marta Fudalej
1,2,
Kamila Krupa
3,
Anna Badowska-Kozakiewicz
1 and
Andrzej Deptała
1,*
1
Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
2
Department of Oncology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
3
Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1484; https://doi.org/10.3390/cancers17091484
Submission received: 28 March 2025 / Revised: 22 April 2025 / Accepted: 26 April 2025 / Published: 28 April 2025

Simple Summary

The inflammatory process is one of the key mediators of pancreatic cancer (PC) development, yet at the same time, PC is one of the most immune-resistant tumors. Patients rarely benefit from monotherapy with immune checkpoint inhibitors; nevertheless, the latest biological findings on the complexity of the pancreatic tumor microenvironment might be translated into designing new clinical studies that combine various approaches to overcome single-agent immunotherapy resistance. On the other hand, focusing on inflammation may lead to the development of new inflammation-based prognostic markers for patients. This review aims to describe the current state of knowledge regarding the complex relationships between systemic and local inflammation, an immune response, immunosuppression, and therapeutic options in PC.

Abstract

Pancreatic cancer (PC) is one of the most commonly diagnosed and deadliest neoplasms in the modern world. Over the past few years, the incidence of PC has risen with only a slight improvement in overall survival. Moreover, the improvement in survival is primarily driven by diagnoses in the localized stage of the disease, rather than by new treatment methods. The inflammatory process is a key mediator of PC development, yet PC is also one of the most immune-resistant tumors. Patients rarely benefit from monotherapy with immune checkpoint inhibitors; nevertheless, the latest biological findings on the complexity of the pancreatic tumor microenvironment might be translated into designing new clinical studies that combine various approaches to overcome single-agent immunotherapy resistance. On the other hand, focusing on inflammation may lead to the development of new inflammation-based prognostic markers for patients. This review aims to describe the current state of knowledge regarding the complex relationships between systemic and local inflammation, immune response, immunosuppression, and therapeutic options in PC.

1. Introduction

Pancreatic cancer (PC) is one of the most commonly diagnosed and most deadly neoplasms in the modern world [1]. Over the past few years, the incidence of PC has risen with only a slight improvement in overall survival (OS), reaching 13% in 2024 [2]. A decade ago, the 5-year OS rate was only 6%; nevertheless, the improvement in survival is driven mainly by diagnoses at the localized stage of the disease, rather than by new treatment methods [3]. In the face of the complexity of patients’ conditions, multidisciplinary teams (comprising doctors of various specialties, dietitians, psychologists, and physiotherapists) should be established to care for them [4]. According to the newest National Comprehensive Cancer Network (NCCN) guidelines, treatment methods encompass surgery, systemic therapies, radiotherapy, and targeted therapy in some indications [5] The tumor is assessed as resectable, with no infiltration of major mesenteric blood vessels, i.e., the celiac axis, common hepatic artery, superior mesenteric artery, superior mesenteric vein, and portal vein. Systemic therapy is utilized in all stages of PC. The value of neoadjuvant treatment is still evolving, with no consensus about its influence on increased cure rates [6]. Preferred first-line regimens for adjuvant chemotherapy encompass modified FOLFIRINOX (mFOLFIRINOX) or gemcitabine with capecitabine as an alternative option [7,8]. In the locally advanced or metastatic stage of the disease, guidelines recommend mFOLFIRINOX or gemcitabine with albumin-bound paclitaxel [9,10]. Radiotherapy (RT) is not a first-line treatment for PC—it is only used in specific indications, such as sterilizing vessel margins or enhancing the likelihood of a margin-negative resection. Additionally, RT can be used to palliate pain and bleeding, or to alleviate local obstructive symptoms [11]. To enhance treatment possibilities, every patient should be tested for potentially actionable somatic findings such as fusions (ALK, NRG1, NTRK, ROS1, FGFR2, and RET), mutations (BRAF, BRCA1/2, KRAS, and PALB2), amplifications (HER2), microsatellite instability (MSI), mismatch repair deficiency (dMMR), or tumor mutational burden (TMB) [5]. BRCA-positive patients will benefit from platinum-based chemotherapy and maintenance with a Poly(ADP-ribose) Polymerase (PARP) inhibitor, such as Olaparib [12]. A programmed death receptor 1 (PD-1) inhibitor, Pembrolizumab, can be proposed as a second or later-line treatment for patients with MSI-high (MSI-H) or dMMR pancreatic tumors [13]. In patients with neurotrophic tropomyosin receptor kinase (NTRK) fusion, selective tyrosine kinase inhibitors such as larotrectinib or entrectinib are recommended; however, they remain inaccessible in many countries [10]. Nowadays, numerous studies concentrate on kinase inhibitors for molecularly defined subgroups of patients with PC. Protein kinase inhibitors are developed to suppress redundant signaling pathways and prevent resistance. Some ongoing studies encompass dual-targeting proteolysis-targeting chimeras (PROTACs) or nortopsentin analogs [14,15,16].
Immunotherapy has revolutionized the treatment of various malignancies and provided a deeper understanding of tumors’ biology. Several immunotherapies have been successfully applied to multiple cancers since the testing of PD-L1 and therapies targeting PD-1 or programmed cell death ligand 1 (PD-L1) inhibitors have become a standard of care in lung cancer and renal cell carcinoma [17]. On the one hand, PC is one of the most immune-resistant tumors; on the other, the inflammatory process is one of the key mediators of PC development [18,19]. Moreover, recent studies have demonstrated that systemic inflammation and immune response markers are correlated with the prognosis and treatment response of PC patients [20,21].
This review aims to describe the current state of knowledge regarding the complex relationships between systemic and local inflammation, the immune response, immunosuppression, and therapeutic options in PC.

2. Inflammation and Immunosuppression

In the 19th century, Virchow observed immune cells in neoplastic tissue and developed the concept of the role of chronic inflammation in cancer development. Since then, chronic inflammation has been linked to various stages of tumorigenesis, encompassing cellular transformation, promotion, survival, proliferation, invasion, angiogenesis, and metastasis. Many environmental causes of cancer and risk factors are associated with some form of chronic inflammation [22]. In a typical setting, inflammation is generally self-limiting, as the production of anti-inflammatory cytokines closely follows that of pro-inflammatory cytokines. The persistence of initiating factors or a failure of mechanisms required to resolve the inflammatory response leads to chronic inflammation [23]. Systemic diseases characterized by low-grade chronic inflammation, such as metabolic syndrome and diabetes mellitus, enhance the risk of cancer, particularly PC. Inflammatory cells simultaneously orchestrate inflammation, enhancing tumor development and governing the immune response against emerging tumor cells. Moreover, they might support the escape of tumor cells from immune control by favoring more immunosuppressive phenotypes and modifying their antigenic fingerprint. Tumor cell metabolism, primarily glucose metabolism, is both affected by and affects inflammatory cells [24]. Chronic inflammation can generate an immunosuppressive microenvironment that allows tumor formation and progression. The pro-inflammatory mediators facilitate crosstalk between different cells, creating a tumor-supporting, immunosuppressive microenvironment [25]. The pancreatic tumor microenvironment (TME) exhibits dynamic changes between inflammation and the immune response, primarily manifesting as inflammation-induced immunosuppression [26]. The immune response, controlling the identification and elimination of tumor cells, can be suppressed by chronic inflammation. Moreover, cytokines produced by inflammatory cells can inhibit the function of immune cells. It further simplifies the process by which cancer cells can proliferate and metastasize [27]. Although inflammation at the tumor site usually promotes cancer progression, it can be remodeled to acquire anti-tumor properties [28].

3. Inflammatory Markers

As previously mentioned, inflammation and the host’s ineffective immune response are primary hallmarks of PC development and progression. Many studies have reported that various pretreatment inflammatory markers may be prognostic factors in malignant tumors, including PC; nevertheless, which combination of these factors may be the most valuable remains undefined [20]. Moreover, they may be used to monitor the effectiveness of chemotherapy.
Some inflammatory markers include various settings of lymphocytes, neutrophils, platelets, and monocytes. One of them—the neutrophil-to-lymphocyte ratio (NLR)—was analyzed in several studies. The cut-off value of the NLR varied a lot, from two to five. Based on a meta-analysis of 43 cohort studies, including 8252 patients with PC, patients with a low NLR had a longer OS, disease-free survival (DFS), significantly smaller tumors, and lower grades, stages, and CA19-9 levels [29]. The underlying mechanism of the association between a high NLR and poor prognosis in patients with PC remains unclear. Neutrophils may promote the proliferation and metastasis of PC through multiple mechanisms, including mediating the epithelial–mesenchymal transition (EMT), inducing angiogenesis by secreting various cytokines, and creating an immunosuppressive microenvironment for PC [30,31]. In terms of lymphocytes, the relationship is far more complex, as there are controversies regarding the changes in T cells in PC patients and the contrasting responses of different T cell subtypes [29]. Nevertheless, a decreased lymphocyte count may reflect an insufficient immunological response to the tumor, as a reduction in lymphocytes might weaken the anti-tumor response and immune surveillance [24].
Another marker, the platelet-to-lymphocyte ratio (PLR), was assessed in the meta-analysis of 17 cohort studies, including 3182 patients with PC. The authors concluded that PLR could be a prognostic factor, as a low PLR was associated with a longer OS and progression-free survival (PFS) [32]. Similar to the NLR, the exact mechanism remains elusive. Platelets promote tumor growth, angiogenesis, metastasis, and cancer-associated thrombosis [33]. They also promote tumor cell adhesion to the microvascular endothelium and form a defense barrier around circulating tumor cells, helping them escape immune surveillance [34].
Hu et al. (2018) conducted a meta-analysis of the lymphocyte-to-monocyte ratio (LMR) as a prognostic indicator for patients with PC. The analysis included 11 cohorts comprising 2557 patients with PC. A low LMR was associated with an unfavorable OS, shorter DFS, and recurrence-free survival (RFS). A low LMR also correlated with higher CA19-9 and TNM stages [35]. In 2024, Li et al. examined whether LMR could serve as an indicator for predicting the prognosis of patients only with resectable PC. They confirmed the substantial predictive value of the LMR for OS; however, the LMR did not exhibit predictive significance for RFS [36]. Monocytes infiltrate tumors and differentiate into TAMs involved in tumor progression. As a result, an increased level of monocytes may express a higher tumor burden [37]. Furthermore, monocytes can inhibit antigen- and mitogen-induced lymphocyte proliferation, impair lymphocyte-dependent anti-tumor defenses, and suppress anti-tumor immunity [38]. Whether the circulating level of monocytes serves as a surrogate for the increased production of tissue macrophages remains an open question for further study.
The systemic immune–inflammation index (SII) is a combined biomarker based on the counts of lymphocytes, platelets, and neutrophils. It was first identified as a prognostic indicator for clinical outcomes of hepatocellular carcinoma (HCC) patients. Two literature review-based meta-analyses of the relationship between the SII and the prognosis of PC patients have shown that patients with higher SII scores have a worse prognosis [39,40]. Another parameter, the systemic inflammation response index (SIRI), is calculated using counts of neutrophils, lymphocytes, and monocytes. In the meta-analysis by Shen et al. (2024) [41] comprising seven studies and 1160 patients, a higher SIRI was associated with worse OS and PFS. Furthermore, prospective studies are needed to confirm whether the SII or SIRI is superior to the NLR or PLR alone in predicting survival in patients with PC.
Neumann et al. (2023) [42] developed a new score, termed the Inflammatory Benchmark Index (IBI). It combines the sum of neutrophils, monocytes, lymphocytes, and platelets, all divided by CRP. The IBI was highly significant in univariate and multivariate analyses of prognostic factors for OS in PC, independent of the tumor stage and PS. Interestingly, in this marker, unlike all the ones discussed before, lymphocytes are grouped together with monocytes, platelets, and neutrophils in the same numerator. This parameter takes CRP into account. In her PhD dissertation, Fudalej observed that among patients with PC and diabetes mellitus, a CRP level ≤ 5 mg/L was the strongest predictor of survival. Moreover, a lower level of another parameter, including CRP—the CRP-to-lymphocyte ratio (CLR)—was significantly associated with longer OS in the group of patients with PC and diabetes mellitus or hypertension [21,43].
The CRP/albumin ratio (CAR) is supposed to represent the potential inflammatory status of the body, as albumins can be involved in the inflammatory response. It may be associated with patients’ nutritional and septic status [44]. Hypoalbuminemia, as an exponent of malnutrition and cachexia, can exacerbate cancer-associated inflammation [45]. Most studies have focused on the CAR and analyzed its preoperative levels in patients with PC who are resectable. In one of them, the CAR was statistically superior to other prognostic markers, including the CLR, NLR, PLR, the modified Glasgow Prognostic Score (mGPS), and the Prognostic Index (PNI) [45]. In another study, a high CAR at postoperative day 14 was related to a high BMI, a large amount of intraoperative bleeding, and the presence of complications, all resulting in shorter OS and RFS [46]. Other authors have analyzed the CRP-to-prealbumin ratio, as some studies suggest that prealbumin may be a more sensitive marker than albumin for assessing the nutritional status [47]. In the study by Kwon et al. (2023), the CRP-to-prealbumin ratio was found to have a significant association with early recurrence after curative resection in patients with resectable PC [48]. Many studies have confirmed that the CAR may be a valuable predictor of recurrence or OS; however, as presented in Table 1, the cut-off values of the CAR vary considerably across different studies.
The CALLY index, derived from CRP, the serum albumin level, and total lymphocyte count, reflects the body’s immune, nutritional, and inflammatory status. It was initially established as a predictive marker in patients undergoing transarterial chemoembolization for HCC [49]. In the investigation of 3511 cancer-afflicted adults, the CALLY index demonstrated a linear and negative association with all-cause mortality, as well as mortality caused by cancer and cardiac conditions [50]. A recent study investigated the utility of the CALLY index in patients undergoing curative surgical treatment for PC. It proved that a low CALLY score reflected a higher risk of complications and mortality due to a weaker immune function, poorer nutritional status, and higher systemic inflammation [51]. In the study by Kawahara et al. (2024), a low CALLY index was significantly associated with a larger tumor size, metastatic lymph nodes, a worse pathological TNM stage, older age, and a low administration rate of adjuvant chemotherapy [52]. Another study examining the preoperative value of the CALLY index in PC patients reported its superior prognostic ability for DFS and OS compared to other scores, including the Geriatric Nutritional Risk Index (GNRI), SII, Controlling Nutritional Status (CONUT score), NLR, and PLR [53]. The CALLY index can be used as an objective indicator reflecting the tumor’s biological malignancy and the patient’s condition. The authors suggest that the CALLY index may be valuable for guiding surgical decision making. Patients with a low CALY index may benefit from initial treatment with neoadjuvant chemotherapy, accompanied by intensive nutritional status enhancement and a reduction in inflammation [53].
As discussed above, various markers or indexes may be implemented to predict the outcomes of patients with PC; nevertheless, there are still more questions than answers. Firstly, most of the presented studies are retrospective, encompassing different stages of PC and other treatment methods. Non-specific processes, such as trauma, infections, or immune-related diseases, may affect cell counts. The same parameter had no uniform threshold; the cut-off values varied significantly between studies. We believe that conducting a well-planned prospective study with a large sample, comparing different indexes, will provide a clear answer about the best new inflammation-based prognostic marker for patients with PC.
Table 1. Analysis of studies examining various parameters for predicting PC prognosis.
Table 1. Analysis of studies examining various parameters for predicting PC prognosis.
ParameterHow CountedCut-Off ValueNo of
Patients
Time
of Measurement
Association Between High Parameters and OutcomesRef.
CARCRP (mg/L) ÷
albumin (g/dL)
0.18595Pretreatment
Post-chemotherapy
Shorter OS[54]
0.06163PreoperativeShorter OS and DFS[45]
0.3414214 postoperative daysShorter OS and RFS[46]
3.85302PretreatmentShorter OS and PFS[55]
0.09143PreoperativeShorter OS[56]
0.03113PreoperativeShorter OS and RFS[44]
0.41294PretreatmentShorter OS[42]
CRP/
pre-albumin
CRP (mg/dl) ÷ prealbumin (mg/dL)1.320PreoperativeShorter RFS[48]
CALLY[albumin (g/L) × lymphocyte count] ÷ [CRP (mg/L) × 104]1.03121PreoperativeLower rates of postoperative complications, longer OS[51]
3.00307Longer OS and DFS[53]
1.90461Longer OS and RFS[52]
SII[platelet count × neutrophil count] ÷ lymphocyte count400–9002132 (meta-analysis), 1749 (meta-analysis)PretreatmentShorter OS, DFS, and PFS[39,40]
SIRI[neutrophil count × monocyte count] ÷ lymphocyte count0.69–2.351160 (meta-analysis)PretreatmentShorter OS and PFS[41]
LMRlymphocyte count ÷ monocyte count2.05–4.622557 (meta-analysis)PretreatmentLonger OS, DFS, RFS[35]
1.60–5.004019 (meta-analysis)PreoperativeLonger OS[36]
NLRneutrophil count ÷ lymphocyte count2.00–5.008252 (meta-analysis)PretreatmentShorter OS and DFS[29]
PLRplatelet count ÷ lymphocyte count126–3003182 (meta-analysis)PretreatmentShorter OS and PFS[32]
IBI[lymphocyte count + monocyte count + neutrophil count + platelet count] ÷ CRP (mg/L)301294PretreatmentLonger OS[42]
Abbreviations: CAR—CRP-to-albumin ratio, OS—overall survival, DFS—disease-free survival, PFS—progression-free survival, RFS—recurrence-free survival, CALLY—CRP-albumin-lymphocyte index, SII—immune–inflammation index, SIRI—systemic inflammation response index, LMR—lymphocyte-to-monocyte ratio, NLR—neutrophil-to-lymphocyte ratio, PLR—platelet-to-lymphocyte ratio, IBI—inflammatory benchmark index.

4. Immunotherapy

Physiologically, when tumor cells invade healthy tissue, the immune system should recognize and eliminate them. Unfortunately, cancerous cells can evade the immune system through different immune escape mechanisms. The main mechanisms encompass (1) recruiting suppressor immune cells (myeloid-derived suppressor cells (MDSCs), regulatory T-cells (Tregs), and cytokines) to form a suppressive immune microenvironment; (2) down-regulating surface antigen expression resulting in immunogenicity decrease; (3) suppressing T-cell activity by up-regulating the immune checkpoint on the surface; and (4) releasing acidic and toxic metabolites which inhibit the activity of immune cells in the TME [57]. Immunotherapy aims to boost the natural defenses of the immunological system to eliminate malignant cells. This kind of treatment is a breakthrough and has revolutionized the field of oncology [58]. The immune responses explicitly targeting cancer cells, triggered by immunotherapy, differ from those stimulated by tumor-directed therapies. Moreover, the response to immunotherapy can endure for a prolonged period even after treatment is discontinued [59].

4.1. Current Immunotherapy Options in PC

MSI-H or dMMR is detected in approximately 1% of patients with PC. It is typically associated with Lynch syndrome, but may also occur sporadically due to promoter methylation or somatic mutations [60]. In 2017, based on five uncontrolled, multicohort, multicenter, and single-arm clinical trials, the U.S. Food and Drug Administration (FDA) granted accelerated approval to pembrolizumab for adult and pediatric patients with unresectable or metastatic MSI-H or dMMR solid tumors that have progressed after previous treatment and for whom there are no satisfactory alternative options. In 2023, the approval was converted to full, supported by findings from the multicenter, non-randomized, open-label, multicohort, phase 2 KEYNOTE-158, KEYNOTE-164, and KEYNOTE-051 trials. Pembrolizumab is an anti-PD-1 receptor antibody that blocks its interaction with PD-L1 and PD-L2 [61,62]. According to the latest NCCN guidelines, pembrolizumab is recommended for patients with advanced MSI-H/dMMR PC for both first-line and subsequent treatment, provided no prior immunotherapy has been implemented [5]. ESMO recommends pembrolizumab as a second or later-line treatment in this cohort [10]. NCCN also includes Dostarlimab-gxly as a subsequent treatment option (with no prior immunotherapy) for patients with MSI-H or dMMR locally advanced, metastatic, or recurrent PC and any performance status (PS) [63] and nivolumab/ipilimumab as a subsequent therapy option for patients with a high tumor mutational burden (TMB-H) and with good or intermediate PS [64], however, with category 2B of recommendation strength.

4.2. Why Is Single-Agent Immunotherapy Not Effective in PC?

PC patients rarely benefit from treatment with immune checkpoint inhibitors (ICI) due to the properties of the highly immunosuppressive TME; pancreatic tumors are often considered “cold tumors”. The low expression of the major histocompatibility complex (MHC) class I molecules on PC cells inhibits T-cell activation, as they are activated by an interaction with antigens presented by MHC I molecules on antigen-presenting cells [65]. Up-regulated proteins, facilitating immunosuppresion and tumor progression, encompass Tregs, tumor-associated macrophages (TAMs), MDSCs, cancer-associated fibroblasts (CAFs), and pancreatic stellate cells (PSCs) (Figure 1) [66].
PSCs are one of the key components of the TME. They release various cytokines and chemokines, such as IL-6, C-X-C motif chemokine ligand (CXCL)12, or CXCL10. CXCL12 sequesters CD8 T-cells and hinders their antitumor activity, while CXCL10 contributes to the recruitment of Tregs. Tregs induce immune suppression through various mechanisms, such as secreting immunosuppressive cytokines or inhibiting dendritic cell maturation. What is more, permanently expressed on Tregs, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) plays a significant role in suppressing antigen-presenting cells [65]. On the other hand, IL-6 promotes the migration of MDSCs to the TME. MDSCs further suppress T-cell proliferation by depleting L-arginine and L-tryptophan. Also, the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) by MDSCs leads to a decrease in the reactivity of MHC I molecules with cytotoxic T-cells [67]. PC presents a dense layer of cancer-associated fibroblasts (CAFs) surrounding malignant cells. CAFs actively crosstalk with cancer cells within the TME and contribute to drug resistance. One of the hallmarks of the physical and immunological barriers of PC is a prominent desmoplastic reaction. The dense and fibrous connective tissue comprising cellular and non-cellular components prevents immune cells from infiltrating the tumor [68]. Down-regulated cells include previously mentioned CD8+ cytotoxic T lymphocytes, natural killer (NK) cells, dendritic cells (DCs), and some phenotypes of B-cells [69]. The poor infiltration of effector T cells leads to the insufficient action of immune checkpoint inhibitors. The hypoxic state of the TME also leads to resistance to immunotherapy, as hypoxia-induced HIF-1α can promote PD-L1 expression in cancer cells and suppress the immune effect. The TME under hypoxia provides survival conditions for tumor cells and obstructs the effect of anti-tumor drugs by hindering drug delivery [70]. Last but not least, PC is characterized by a low mutation burden in most cases; thus, fewer tumor-specific neoantigens are available to trigger an immune response, making checkpoint inhibitors even less effective [71].

4.3. Clinical Trials

As shown in Figure 2, various approaches are employed to overcome resistance to immunotherapy. One of them is combining immunostimulatory strategies. The DURIPANC trial combines an ICI Durvalumab with a Toll-like receptor (TLR-3) agonist (NCT05927142). TLR-3 agonists are hypothesized to induce the production of pro-inflammatory cytokines, including type I interferon (IFN-I), increase dendritic cell maturation, and the cross-priming of naive cytotoxic CD8+ T cells while eliminating the attraction of Tregs [72]. Preliminary data from this study demonstrated early disease control and safety in patients with late-stage PC [73]. Another study combines Pembrolizumab with dipeptidyl peptidase (DPP) and fibroblast activation protein (FAP) inhibitor—Talabostat (NCT05558982). Talabostat is intended to upregulate immune responses against cancer by increasing cytokine and chemokine production, thereby enhancing tumor-specific T-cell immunity. Moreover, high-affinity interactions involving FAP stimulate the anti-tumor activity of neutrophils, macrophages, and natural killer cells [74]. In the preliminary findings, BXCL701 (talabostat) plus pembrolizumab was well-tolerated and showed early signs of potential clinical activity in patients with metastatic PC refractory to chemotherapy [75]. Other approaches combine multi-kinase inhibitors, such as anlotinib, cabozantinib, or levantinib, with anti-PD-1 immunotherapy and chemotherapy (NCT06051851, NCT05052723, and NCT04887805). Multi-kinase inhibitors work synergistically with ICI by depleting the TME. They reduce immunosuppressive MDSCs and Tregs, enhance anti-tumor macrophage activity, and inhibit tumor growth and dissemination [76,77]. Some studies design immune-based therapies against KRAS or BRCA mutations (NCT06015724 and NCT05093231). By reversing some KRAS-initiated immunosuppressive changes, KRAS inhibitors make tumors more sensitive to immunotherapy [78]. Several ongoing clinical trials that combine immunotherapy with other therapeutic strategies are presented in Table 2.

4.4. Vaccine-Based Immunotherapy

Therapeutic cancer vaccines aim to generate long-lived, tumor-specific, functional T-cells that destroy cancer cells [79]. Vaccination is mainly considered in PC in the adjuvant setting due to the minimized tumor disease burden and TME immunosuppression, which may, in effect, improve clinical efficacy [18]. Types of cancer vaccines in PC include whole-cell, dendritic cell, peptide-based, mRNA-based, and viral-vector vaccines [80].
One of the most intensively studied whole cancer cell vaccines is GVAX (an allogenic, human granulocyte macrophage-colony-stimulating factor (GM-CSF)-secreting whole-cell PC vaccine) [59]. GVAX, when administered with cyclophosphamide (Cy) to deplete Tregs, has been shown to induce T-cell infiltration in TME. Moreover, vaccine therapy induced PD-L1 expression in both tumor epithelial cells and myeloid cells within the TME. These observations resulted in the design of studies that combined GVAX with ICI [81,82]. One of them—a platform trial of neoadjuvant and adjuvant antitumor vaccination with a PD-1 antagonist (nivolumab) and CD137 agonist (urelumab)—proved that this triplet arm is safe, increased intratumoral activated, cytotoxic T cells, and demonstrated a potentially promising efficacy signal in resectable pancreatic ductal adenocarcinoma (PDAC). Nevertheless, there was no DFS benefit to adding a PD-1a antagonist compared to GVAX alone. The triple regimen was associated with a marginally significant improvement in DFS compared to GVAX alone. The authors emphasized that this study was underpowered to achieve statistical significance, and further studies involving larger patient groups are required [83]. Another ongoing Phase 1 and 2 trial focuses on the CD137 agonist AGEN2373, combined with balstilimab (anti-PD-1), in combination with GVAX (Arm 1) or mKRASvx (Arm 2) (NCT06782932).
DC vaccines promote the activation of cytotoxic CD8+ T cells by presenting antigens to CD4+ and CD8+ T cells and secreting cytokines such as IL-15, IL-12, IFN-γ, and TNF [84]. Although studies on monotherapy with DC vaccines have shown no improvement in survival, DC vaccines have induced cytotoxic T cells and enhanced cytotoxic cytokine signaling, thereby suggesting that they may strengthen immunity, but require alternative combination therapeutics to improve outcomes [85]. This ongoing study examines the DC vaccine loaded with personalized peptides in combination with adjuvant chemotherapy (mFOLFIRINOX or gemcitabine/capecitabine), followed by nivolumab (NCT04627246). Another active study focuses on delivering autologous DC loaded with pancreatic adenocarcinoma lysate plus mRNA as an adjuvant therapy following the completion of standard chemotherapy (NCT04157127).
Peptide-based vaccines can induce immunity against specific antigenic epitopes derived from the vaccinated proteins or peptides expressed in cancer cells [80]. Neoantigens or tumor-specific antigens are applied to stimulate an immune response. They might be identified via the genetic testing of samples from patients’ biopsies to deliver the most suitable personalized vaccine [86]. Peptide-based vaccines present tumor-specific antigen peptides to MHC molecules to elicit the production of specific and long-term memory T-cells against cancer. MHC class I cells stimulate CD8+ cytotoxic T cell responses, while a small proportion of MHC class II cells stimulate CD4+ helper cells. CD4+ T-cells support the anticancer effect of CD8+ T-cells through enhancing the antigen presentation and releasing cytokines such as INF-γ and IL-2. [87]. One of the key peptides targeted in PC is mutant KRAS, as activating KRAS mutations are detected in over 90% of PDAC cases and implicated in tumor initiation and progression [88]. Recently, the AMPLIFY-201 Phase 1 study on patients with PC who are positive for minimal residual mKRAS disease (ctDNA and/or a serum tumor antigen) after locoregional treatment showed promising results. It examined the ELI-002 2P cancer vaccine, which enhances lymph node delivery and the immune response. The vaccine consists of the amphiphile (Amph) modification of G12D and G12R mutant KRAS (mKRAS) peptides (Amph-Peptides-2P) together with the CpG oligonucleotide adjuvant (Amph-CpG-7909). The therapy was well-tolerated, induced significant T-cell responses, and resulted in biomarker clearance and improved RFS [89]. These positive findings on the 2-peptide formulation of ELI-002 led to the design of another ongoing Phase 1 and 2 study on ELI-002 7P, which includes seven Amphipathic-modified KRAS and NRAS peptides: G12D, G12R, G12V, G12A, G12C, G12S, and G13D (Amph-Peptides 7P) (NCT05726864). Another promising, still recruiting study investigates the combination of the KRAS peptide vaccine with Ipilimumab and Nivolumab for patients with resected PDAC after neoadjuvant and/or adjuvant chemotherapy and/or radiation. The designed vaccine consists of polyinosinic–polycytidylic acid synthetic long peptides corresponding to six common mKRAS subtypes: G12D, G12R, G12V, G12A, G12C, and G13D (NCT04117087) [90].
mRNA vaccines function by delivering a single-stranded molecule encoding viral proteins or neoantigens. The molecule is incorporated into the cytoplasm of antigen-presenting cells and further translated into neoantigen proteins. Subsequently, mature antigen-presenting cells are transported to lymph nodes, generating a remarkable humoral and cellular immune response [91]. One of the most widely studied mRNA cancer vaccines is autogene cevumeran, an individualized mRNA neoantigen vaccine containing up to 20 primary histocompatibility complex class I (MHC I) and MHC class II (MHC II) restricted neoantigens in lipoplex nanoparticles, delivered intravenously. The study by Rojas et al. (2023) reported preliminary evidence that adjuvant autogene cevumeran, in combination with atezolizumab and mFOLFIRINOX, induced substantial T-cell activity in patients with surgically resected PDAC that correlated with delayed recurrence [92]. Based on these findings, a phase II, open-label, multicenter, randomized trial was designed to compare adjuvant autogene cevumeran with atezolizumab and mFOLFIRINOX versus mFOLFIRINOX alone (NCT05968326).
Viral-vector vaccines contain modified viruses that deliver cancer-specific genes or antigens, stimulating the immune system [93]. One is CRS-207, a live, attenuated, double-deleted Listeria monocytogenes (LADD) engineered to stimulate an immune response to mesothelin. The results from the ECLIPSE study showed that the combination of Cy/GVAX and CRS-207 did not improve survival compared to chemotherapy [94]. In further trials, adding nivolumab to Cy/GVAX plus CRS-207 did not improve the OS; however, objective responses and immunologic changes in the TME were evident [95]. The ongoing Phase 2 clinical trial is evaluating the combination of CRS-207 with pembrolizumab, ipilimumab, and tadalafil in patients with metastatic PDAC (NCT05014776). The pre-clinical study by Gross et al. (2024) demonstrated that phosphodiesterase type 5 (PDE5) inhibition with tadalafil, combined with vaccine-based immunotherapy, promoted pro-inflammatory states in myeloid cells, activated T cells, and enhanced crosstalk between myeloid cells and T cells [96].
Most vaccine therapy clinical trials have not demonstrated dramatic improvements in survival. However, most of these studies demonstrate signals of increased cancer antigen-specific T-cell responses after vaccination and potent ICI enhancement [85].

5. Conclusions

The inflammatory process is a key mediator of PC development, yet PC is also one of the most immune-resistant tumors. A plethora of various aspects relating to the involvement of inflammatory and anti-inflammatory processes are being investigated; nevertheless, none of them have turned out to be a panacea for PC. Patients with PC rarely benefit from monotherapy with immune checkpoint inhibitors, as indications for the use of anti-PD-1 antibodies are limited to only 1% of PC cases that exhibit dMMR or MSI-H. Nevertheless, the latest biological findings on the complexity of the TME may be translated into designing new clinical studies that combine various approaches to overcome resistance to single-agent immunotherapy. Some of them include combining immunostimulatory strategies or using specifically developed cancer vaccines. Another aspect is that focusing on inflammation may lead to new inflammation-based prognostic markers for patients. Establishing new indices or markers from the blood samples tested before the first course of chemotherapy may result in more accurate treatment choices.

Author Contributions

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

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
PCPancreatic cancer
OSOverall survival
NCCNNational Comprehensive Cancer Network
mFOLFIRINOXModified FOLFIRINOX
RTRadiotherapy
MSIMicrosatellite instability
MSI-HHigh microsatellite instability
dMMRMismatch repair deficiency
TMBTumor mutational burden
PARPPoly(ADP-ribose) Polymerase
NTRKNeurotrophic tropomyosin receptor kinase
PD-L1Programmed cell death ligand 1
TMETumor microenvironment
MDSCsMyeloid-derived suppressor cells
TregsRegulatory T-cells
FDAU.S. Food and Drug Administration (FDA)
ESMOEuropean Society for Medical Oncology
PSPerformance status
TMB-HHigh tumor mutational burden
ICIImmune checkpoint inhibitor
TAMsTumor-associated macrophages
TLRToll-like receptor
IFN-IType I Interferon
DPPDipeptidyl peptidase
FAP Fibroblast activation protein
NLRNeutrophil-to-lymphocyte ratio
DFSDisease-free survival
EMTEpithelial-to-mesenchymal transition
PLRPlatelet-to-lymphocyte ratio
PFSProgression-free survival
LMRLymphocyte-to-monocyte ratio
RFSRecurrence-free survival
SIIImmune–inflammation index
SIRISystemic inflammation response index
IBI Inflammatory benchmark index
CARCRP/albumin ratio
mGPSModified Glasgow prognostic score
PNIPrognostic index
CALLY indexCRP-albumin-lymphocyte index
GNRI Geriatric nutritional risk index
CONUT statusControlling nutritional status
PDACPancreatic ductal adenocarcinoma
PDE5Phosphodiesterase type 5
CXCLC-X-C motif chemokine ligand
MHCMajor histocompatibility complex
CAFCancer-associated fibroblasts
PSCPancreatic stellate cell
ROSReactive oxygen species
RNSReactive nitrogen species
NKNatural killer
DCDendritic cells

References

  1. Wang, S.; Zheng, R.; Li, J.; Zeng, H.; Li, L.; Chen, R.; Sun, K.; Han, B.; Bray, F.; Wei, W.; et al. Global, regional, and national lifetime risks of developing and dying from gastrointestinal cancers in 185 countries: A population-based systematic analysis of GLOBOCAN. Lancet Gastroenterol. Hepatol. 2024, 9, 229–237. [Google Scholar] [CrossRef] [PubMed]
  2. American Cancer Society. Cancer Facts & Figures 2025. Atlanta: American Cancer Society. 2025. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/2025-cancer-facts-figures.html (accessed on 12 March 2025).
  3. Stoffel, E.M.; Brand, R.E.; Goggins, M. Pancreatic Cancer: Changing Epidemiology and New Approaches to Risk Assessment, Early Detection, and Prevention. Gastroenterology 2023, 164, 752–765. [Google Scholar] [CrossRef] [PubMed]
  4. Yao, W.; Chen, X.; Fan, B.; Zeng, L.; Zhou, Z.; Mao, Z.; Shen, Q. Multidisciplinary team diagnosis and treatment of pancreatic cancer: Current landscape and future prospects. Front. Oncol. 2023, 13, 1077605. [Google Scholar] [CrossRef]
  5. National Comprehensive Cancer Network. Pancreatic Adenocarcinoma (Version 2.2025). Available online: https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf (accessed on 20 February 2025).
  6. Kwaśniewska, D.; Fudalej, M.; Nurzyński, P.; Badowska-Kozakiewicz, A.; Czerw, A.; Cipora, E.; Sygit, K.; Bandurska, E.; Deptała, A. How A Patient with Resectable or Borderline Resectable Pancreatic Cancer should Be Treated-A Comprehensive Review. Cancers 2023, 15, 4275. [Google Scholar] [CrossRef]
  7. Neoptolemos, J.P.; Stocken, D.D.; Bassi, C.; Ghaneh, P.; Cunningham, D.; Goldstein, D.; Padbury, R.; Moore, M.J.; Gallinger, S.; Mariette, C.; et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: A randomized controlled trial. JAMA 2010, 304, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  8. Neoptolemos, J.P.; Palmer, D.H.; Ghaneh, P.; Psarelli, E.E.; Valle, J.W.; Halloran, C.M.; Faluyi, O.; O’Reilly, D.A.; Cunningham, D.; Wadsley, J.; et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet 2017, 389, 1011–1024. [Google Scholar] [CrossRef]
  9. Wainberg, Z.A.; Melisi, D.; Macarulla, T.; Pazo Cid, R.; Chandana, S.R.; De La Fouchardière, C.; Dean, A.; Kiss, I.; Lee, W.J.; Goetze, T.O.; et al. NALIRIFOX versus nab-paclitaxel and gemcitabine in treatment-naive patients with metastatic pancreatic ductal adenocarcinoma (NAPOLI 3): A randomised, open-label, phase 3 trial. Lancet 2023, 402, 1272–1281. [Google Scholar] [CrossRef]
  10. Conroy, T.; Pfeiffer, P.; Vilgrain, V.; Lamarca, A.; Seufferlein, T.; O’Reilly, E.M.; Hackert, T.; Golan, T.; Prager, G.; Haustermans, K.; et al. Pancreatic cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 987–1002. [Google Scholar] [CrossRef]
  11. Huguet, F.; Dabout, V.; Rivin Del Campo, E.; Gaujoux, S.; Bachet, J.B. The role of radiotherapy in locally advanced pancreatic cancer. Br. J. Radiol. 2021, 94, 20210044. [Google Scholar] [CrossRef]
  12. Fudalej, M.; Kwaśniewska, D.; Nurzyński, P.; Badowska-Kozakiewicz, A.; Mękal, D.; Czerw, A.; Sygit, K.; Deptała, A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers 2023, 15, 2327. [Google Scholar] [CrossRef]
  13. Storandt, M.H.; Tran, N.; Martin, N.; Jatoi, A. Pembrolizumab near the end of life in patients with metastatic pancreatic cancer: A multi-site consecutive series to examine survival and patient treatment burden. Cancer Immunol. Immunother. 2023, 72, 2515–2520. [Google Scholar] [CrossRef] [PubMed]
  14. Carbone, D.; Pecoraro, C.; Panzeca, G.; Xu, G.; Roeten, M.S.F.; Cascioferro, S.; Giovannetti, E.; Diana, P.; Parrino, B. 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Nortopsentin Derivatives against Pancreatic Ductal Adenocarcinoma: Synthesis, Cytotoxic Activity, and Inhibition of CDK1. Mar. Drugs 2023, 21, 412. [Google Scholar] [CrossRef] [PubMed]
  15. Neerasa, J.; Kim, B.; Chung, H. Novel dual-targeting PROTAC degraders of GSK-3β and CDK5: A promising approach for pancreatic cancer treatment. Bioorganic Med. Chem. 2025, 120, 118085. [Google Scholar] [CrossRef]
  16. Rudloff, U. Emerging kinase inhibitors for the treatment of pancreatic ductal adenocarcinoma. Expert. Opin. Emerg. Drugs 2022, 27, 345–368. [Google Scholar] [CrossRef] [PubMed]
  17. Rosellini, M.; Marchetti, A.; Mollica, V.; Rizzo, A.; Santoni, M.; Massari, F. Prognostic and predictive biomarkers for immunotherapy in advanced renal cell carcinoma. Nat. Rev. Urol. 2023, 20, 133–157. [Google Scholar] [CrossRef]
  18. 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]
  19. Shadhu, K.; Xi, C. Inflammation and pancreatic cancer: An updated review. Saudi J. Gastroenterol. 2019, 25, 3–13. [Google Scholar] [CrossRef]
  20. Fan, Z.; Luo, G.; Gong, Y.; Xu, H.; Qian, Y.; Deng, S.; Huang, Q.; Yang, C.; Cheng, H.; Jin, K.; et al. Prognostic Value of the C-Reactive Protein/Lymphocyte Ratio in Pancreatic Cancer. Ann. Surg. Oncol. 2020, 27, 4017–4025. [Google Scholar] [CrossRef]
  21. Fudalej, M.; Cichowska, I.; Badowska-Kozakiewicz, A.; Deptała, A. The prevalence and impact of overweight and hypertension among patients with pancreatic cancer. Oncol. Clin. Pract. 2024, 21, 15–27. [Google Scholar] [CrossRef]
  22. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126. [Google Scholar] [CrossRef]
  23. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  24. Padoan, A.; Plebani, M.; Basso, D. Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int. J. Mol. Sci. 2019, 20, 676. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, D.; DuBois, R.N. Immunosuppression associated with chronic inflammation in the tumor microenvironment. Carcinogenesis 2015, 36, 1085–1093. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Y.; Wang, J.; Wang, H.; Zhang, S.; Wei, Y.; Liu, S. The Interplay Between Inflammation and Stromal Components in Pancreatic Cancer. Front. Immunol. 2022, 13, 850093. [Google Scholar] [CrossRef] [PubMed]
  27. Nigam, M.; Mishra, A.P.; Deb, V.K.; Dimri, D.B.; Tiwari, V.; Bungau, S.G.; Bungau, A.F.; Radu, A.-F. Evaluation of the association of chronic inflammation and cancer: Insights and implications. Biomed. Pharmacother. 2023, 164, 115015. [Google Scholar] [CrossRef]
  28. Pęczek, P.; Gajda, M.; Rutkowski, K.; Fudalej, M.; Deptała, A.; Badowska-Kozakiewicz, A.M. Cancer-associated inflammation: Pathophysiology and clinical significance. J. Cancer Res. Clin. Oncol. 2023, 149, 2657–2672. [Google Scholar] [CrossRef]
  29. Zhou, Y.; Wei, Q.; Fan, J.; Cheng, S.; Ding, W.; Hua, Z. Prognostic role of the neutrophil-to-lymphocyte ratio in pancreatic cancer: A meta-analysis containing 8252 patients. Clin. Chim. Acta 2018, 479, 181–189. [Google Scholar] [CrossRef]
  30. Gaida, M.M.; Steffen, T.G.; Günther, F.; Tschaharganeh, D.F.; Felix, K.; Bergmann, F.; Schirmacher, P.; Hänsch, G.M. Polymorphonuclear neutrophils promote dyshesion of tumor cells and elastase-mediated degradation of E-cadherin in pancreatic tumors. Eur. J. Immunol. 2012, 42, 3369–3380. [Google Scholar] [CrossRef]
  31. Bausch, D.; Pausch, T.; Krauss, T.; Hopt, U.T.; Fernandez-del-Castillo, C.; Warshaw, A.L.; Thayer, S.P.; Keck, T. Neutrophil granulocyte derived MMP-9 is a VEGF independent functional component of the angiogenic switch in pancreatic ductal adenocarcinoma. Angiogenesis 2011, 14, 235–243. [Google Scholar] [CrossRef]
  32. Zhou, Y.; Cheng, S.; Fathy, A.H.; Qian, H.; Zhao, Y. Prognostic value of platelet-to-lymphocyte ratio in pancreatic cancer: A comprehensive meta-analysis of 17 cohort studies. Onco Targets Ther. 2018, 11, 1899–1908. [Google Scholar] [CrossRef]
  33. Menter, D.G.; Tucker, S.C.; Kopetz, S.; Sood, A.K.; Crissman, J.D.; Honn, K.V. Platelets and cancer: A casual or causal relationship: Revisited. Cancer Metastasis Rev. 2014, 33, 231–269. [Google Scholar] [CrossRef] [PubMed]
  34. Boone, B.A.; Zenati, M.S.; Rieser, C.; Hamad, A.; Al-Abbas, A.; Zureikat, A.H.; Hogg, M.E.; Neal, M.D.; Zeh, H.J., 3rd. Risk of Venous Thromboembolism for Patients with Pancreatic Ductal Adenocarcinoma Undergoing Preoperative Chemotherapy Followed by Surgical Resection. Ann. Surg. Oncol. 2019, 26, 1503–1511. [Google Scholar] [CrossRef] [PubMed]
  35. Hu, R.-j.; Ma, J.-y.; Hu, G. Lymphocyte-to-monocyte ratio in pancreatic cancer: Prognostic significance and meta-analysis. Clin. Chim. Acta 2018, 481, 142–146. [Google Scholar] [CrossRef]
  36. Li, H.; Peng, S.; An, R.; Du, N.; Wu, H.; Zhen, X.; Gao, Y.; Li, Z.; Min, J. The prognostic role of lymphocyte-to-monocyte ratio in patients with resectable pancreatic cancer: A systematic review and meta-analysis. PeerJ 2024, 12, e17585. [Google Scholar] [CrossRef] [PubMed]
  37. Goto, W.; Kashiwagi, S.; Asano, Y.; Takada, K.; Takahashi, K.; Hatano, T.; Takashima, T.; Tomita, S.; Motomura, H.; Hirakawa, K.; et al. Predictive value of lymphocyte-to-monocyte ratio in the preoperative setting for progression of patients with breast cancer. BMC Cancer 2018, 18, 1137. [Google Scholar] [CrossRef]
  38. Pollard, J.W. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer 2004, 4, 71–78. [Google Scholar] [CrossRef]
  39. Li, X.; Lin, H.; Ouyang, R.; Yang, Y.; Peng, J. Prognostic significance of the systemic immune-inflammation index in pancreatic carcinoma patients: A meta-analysis. Biosci. Rep. 2021, 41, BSR20204401. [Google Scholar] [CrossRef] [PubMed]
  40. Li, M.; Li, Z.; Wang, Z.; Yue, C.; Hu, W.; Lu, H. Prognostic value of systemic immune-inflammation index in patients with pancreatic cancer: A meta-analysis. Clin. Exp. Med. 2022, 22, 637–646. [Google Scholar] [CrossRef]
  41. Shen, H.; Zuo, F. Prognostic role of systemic inflammation response index (SIRI) in patients with pancreatic cancer: A meta-analysis. Front. Oncol. 2024, 14, 1465279. [Google Scholar] [CrossRef]
  42. Neumann, C.C.M.; Schneider, F.; Hilfenhaus, G.; Vecchione, L.; Felsenstein, M.; Ihlow, J.; Geisel, D.; Sander, S.; Pratschke, J.; Stintzing, S.; et al. Inflammation-Based Prognostic Scores in Pancreatic Cancer Patients-A Single-Center Analysis of 1294 Patients within the Last Decade. Cancers 2023, 15, 2367. [Google Scholar] [CrossRef]
  43. Badowska-Kozakiewicz, A.; Fudalej, M.; Kwaśniewska, D.; Durlik, M.; Nasierowska-Guttmejer, A.; Mormul, A.; Włoszek, E.; Czerw, A.; Banaś, T.; Deptała, A. Diabetes Mellitus and Pancreatic Ductal Adenocarcinoma—Prevalence, Clinicopathological Variables, and Clinical Outcomes. Cancers 2022, 14, 2840. [Google Scholar] [CrossRef] [PubMed]
  44. Haruki, K.; Shiba, H.; Shirai, Y.; Horiuchi, T.; Iwase, R.; Fujiwara, Y.; Furukawa, K.; Misawa, T.; Yanaga, K. The C-reactive Protein to Albumin Ratio Predicts Long-Term Outcomes in Patients with Pancreatic Cancer After Pancreatic Resection. World J. Surg. 2016, 40, 2254–2260. [Google Scholar] [CrossRef] [PubMed]
  45. Arakawa, Y.; Miyazaki, K.; Yoshikawa, M.; Yamada, S.; Saito, Y.; Ikemoto, T.; Imura, S.; Morine, Y.; Shimada, M. Value of the CRP-albumin ratio in patients with resectable pancreatic cancer. J. Med. Investig. 2021, 68, 244–255. [Google Scholar] [CrossRef] [PubMed]
  46. Arima, K.; Yamashita, Y.-i.; Hashimoto, D.; Nakagawa, S.; Umezaki, N.; Yamao, T.; Tsukamoto, M.; Kitano, Y.; Yamamura, K.; Miyata, T.; et al. Clinical usefulness of postoperative C-reactive protein/albumin ratio in pancreatic ductal adenocarcinoma. Am. J. Surg. 2018, 216, 111–115. [Google Scholar] [CrossRef]
  47. Unal, D.; Orhan, O.; Eroglu, C.; Kaplan, B. Prealbumin is a more sensitive marker than albumin to assess the nutritional status in patients undergoing radiotherapy for head and neck cancer. Contemp. Oncol./Współczesna Onkol. 2013, 17, 276–280. [Google Scholar] [CrossRef]
  48. Kwon, C.H.; Seo, H.I.; Kim, D.U.; Han, S.Y.; Kim, S.; Lee, N.K.; Hong, S.B.; Ahn, J.H.; Park, Y.M.; Noh, B.G. Clinical significance of C-reactive protein-to-prealbumin ratio in predicting early recurrence in resectable pancreatic cancer. Korean J. Clin. Oncol. 2023, 19, 11–17. [Google Scholar] [CrossRef]
  49. Müller, L.; Hahn, F.; Mähringer-Kunz, A.; Stoehr, F.; Gairing, S.J.; Michel, M.; Foerster, F.; Weinmann, A.; Galle, P.R.; Mittler, J.; et al. Immunonutritive Scoring for Patients with Hepatocellular Carcinoma Undergoing Transarterial Chemoembolization: Evaluation of the CALLY Index. Cancers 2021, 13, 5018. [Google Scholar] [CrossRef]
  50. Zhu, D.; Lin, Y.-D.; Yao, Y.-Z.; Qi, X.-J.; Qian, K.; Lin, L.-Z. Negative association of C-reactive protein-albumin-lymphocyte index (CALLY index) with all-cause and cause-specific mortality in patients with cancer: Results from NHANES 1999–2018. BMC Cancer 2024, 24, 1499. [Google Scholar] [CrossRef]
  51. Angın, Y.S.; Şendil, A.M.; Zengin, A.; Ceylan, C.; Bal, C.; Kılıç, M.; Ulaş, M. Pancreatic Cancer and the Prognostic Value of the CALLY Index: A Comparative Analysis. Indian J. Surg. 2025. [Google Scholar] [CrossRef]
  52. Kawahara, S.; Aoyama, T.; Murakawa, M.; Kanemoto, R.; Matsushita, N.; Hashimoto, I.; Kamiya, M.; Maezawa, Y.; Kobayashi, S.; Ueno, M.; et al. Clinical usefulness of C-reactive protein-albumin-lymphocyte (CALLY) index as a prognostic biomarker in patients undergoing surgical resection of pancreatic cancer. Langenbecks Arch. Surg. 2024, 409, 317. [Google Scholar] [CrossRef]
  53. Matsui, S.; Kato, Y.; Ohgi, K.; Ashida, R.; Yamada, M.; Otsuka, S.; Uesaka, K.; Sugiura, T. Prognostic impact of the CALLY index in patients with resectable pancreatic cancer. Surg. Oncol. Insight 2025, 2, 100119. [Google Scholar] [CrossRef]
  54. Fan, Z.; Fan, K.; Gong, Y.; Huang, Q.; Yang, C.; Cheng, H.; Jin, K.; Ni, Q.; Yu, X.; Luo, G.; et al. The CRP/Albumin Ratio Predicts Survival And Monitors Chemotherapeutic Effectiveness In Patients With Advanced Pancreatic Cancer. Cancer Manag. Res. 2019, 11, 8781–8788. [Google Scholar] [CrossRef]
  55. Kim, H.J.; Lee, S.Y.; Kim, D.S.; Kang, E.J.; Kim, J.S.; Choi, Y.J.; Oh, S.C.; Seo, J.H.; Kim, J.S. Inflammatory markers as prognostic indicators in pancreatic cancer patients who underwent gemcitabine-based palliative chemotherapy. Korean J. Intern. Med. 2020, 35, 171–184. [Google Scholar] [CrossRef]
  56. Ikuta, S.; Aihara, T.; Yamanaka, N. Preoperative C-reactive protein to albumin ratio is a predictor of survival after pancreatic resection for pancreatic ductal adenocarcinoma. Asia Pac. J. Clin. Oncol. 2019, 15, e109–e114. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, C.; Yang, M.; Zhang, D.; Chen, M.; Zhu, D. Clinical cancer immunotherapy: Current progress and prospects. Front. Immunol. 2022, 13, 961805. [Google Scholar] [CrossRef]
  58. 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] [PubMed]
  59. Farhangnia, P.; Khorramdelazad, H.; Nickho, H.; Delbandi, A.-A. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J. Hematol. Oncol. 2024, 17, 40. [Google Scholar] [CrossRef]
  60. Sakakida, T.; Ishikawa, T.; Doi, T.; Morita, R.; Kataoka, S.; Miyake, H.; Yamaguchi, K.; Moriguchi, M.; Sogame, Y.; Yasuda, H.; et al. Genomic profile and clinical features of MSI-H and TMB-high pancreatic cancers: Real-world data from C-CAT database. J. Gastroenterol. 2024, 59, 145–156. [Google Scholar] [CrossRef]
  61. Maio, M.; Ascierto, P.A.; Manzyuk, L.; Motola-Kuba, D.; Penel, N.; Cassier, P.A.; Bariani, G.M.; De Jesus Acosta, A.; Doi, T.; Longo, F.; et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: Updated analysis from the phase II KEYNOTE-158 study. Ann. Oncol. 2022, 33, 929–938. [Google Scholar] [CrossRef]
  62. Le, D.T.; Diaz, L.A., Jr.; Kim, T.W.; Van Cutsem, E.; Geva, R.; Jäger, D.; Hara, H.; Burge, M.; O’Neil, B.H.; Kavan, P.; et al. Pembrolizumab for previously treated, microsatellite instability-high/mismatch repair-deficient advanced colorectal cancer: Final analysis of KEYNOTE-164. Eur. J. Cancer 2023, 186, 185–195. [Google Scholar] [CrossRef]
  63. André, T.; Berton, D.; Curigliano, G.; Sabatier, R.; Tinker, A.V.; Oaknin, A.; Ellard, S.; de Braud, F.; Arkenau, H.T.; Trigo, J.; et al. Antitumor Activity and Safety of Dostarlimab Monotherapy in Patients With Mismatch Repair Deficient Solid Tumors: A Nonrandomized Controlled Trial. JAMA Netw. Open 2023, 6, e2341165. [Google Scholar] [CrossRef]
  64. Schenker, M.; Burotto, M.; Richardet, M.; Ciuleanu, T.; Goncalves, A.; Steeghs, N.; Schöffski, P.; Ascierto, P.A.; Maio, M.; Lugowska, I.; et al. Abstract CT022: CheckMate 848: A randomized, open-label, phase 2 study of nivolumab in combination with ipilimumab or nivolumab monotherapy in patients with advanced or metastatic solid tumors of high tumor mutational burden. Cancer Res. 2022, 82, CT022. [Google Scholar] [CrossRef]
  65. Murakami, T.; Hiroshima, Y.; Matsuyama, R.; Homma, Y.; Hoffman, R.M.; Endo, I. Role of the tumor microenvironment in pancreatic cancer. Ann. Gastroenterol. Surg. 2019, 3, 130–137. [Google Scholar] [CrossRef] [PubMed]
  66. Bilotta, M.T.; Antignani, A.; Fitzgerald, D.J. Managing the TME to improve the efficacy of cancer therapy. Front. Immunol. 2022, 13, 954992. [Google Scholar] [CrossRef] [PubMed]
  67. Hartupee, C.; Nagalo, B.M.; Chabu, C.Y.; Tesfay, M.Z.; Coleman-Barnett, J.; West, J.T.; Moaven, O. Pancreatic cancer tumor microenvironment is a major therapeutic barrier and target. Front. Immunol. 2024, 15, 1287459. [Google Scholar] [CrossRef]
  68. Ullman, N.A.; Burchard, P.R.; Dunne, R.F.; Linehan, D.C. Immunologic Strategies in Pancreatic Cancer: Making Cold Tumors Hot. J. Clin. Oncol. 2022, 40, 2789–2805. [Google Scholar] [CrossRef]
  69. 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]
  70. 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]
  71. Imamura, T.; Ashida, R.; Ohshima, K.; Uesaka, K.; Sugiura, T.; Ohgi, K.; Yamada, M.; Otsuka, S.; Hatakeyama, K.; Nagashima, T.; et al. Characterization of pancreatic cancer with ultra-low tumor mutational burden. Sci. Rep. 2023, 13, 4359. [Google Scholar] [CrossRef]
  72. Haddaoui, H.E.; van Eijck, C.W.; Doukas, M.; van den Bosch, T.P.; van Koetsveld, P.M.; Hofland, L.J.; Mustafa, D.A.; van Eijck, C.H. Rintatolimod: A potential treatment in patients with pancreatic cancer expressing Toll-like receptor 3. Am. J. Cancer Res. 2023, 13, 2657–2669. [Google Scholar]
  73. AIM ImmunoTech Reports Positive Preliminary Data in Phase 1b/2 Study of Ampligen and Imfinzi as a Combination Therapy for Late-Stage Pancreatic Cancer. Available online: https://aimimmuno.com/aim-immunotech-reports-positive-preliminary-data-in-phase-1b-2-study-of-ampligen-and-imfinzi-as-a-combination-therapy-for-late-stage-pancreatic-cancer/ (accessed on 11 March 2025).
  74. Gehris, J.; Ervin, C.; Hawkins, C.; Womack, S.; Churillo, A.M.; Doyle, J.; Sinusas, A.J.; Spinale, F.G. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem. Pharmacol. 2024, 219, 115914. [Google Scholar] [CrossRef]
  75. Weinberg, B.A.; Lekan, A.; Fitzgerald, A.; Malchiodi, Z.; Gutierrez, M.; Tesfaye, A.A.; Tan, M.T.; Noel, M.S.; He, A.R.; Mukherji, R.; et al. Phase II trial of BXCL701 and pembrolizumab in patients with metastatic pancreatic ductal adenocarcinoma (EXPEL-PANC): Preliminary findings. J. Clin. Oncol. 2024, 42, LBA4132. [Google Scholar] [CrossRef]
  76. Adhikary, P.; Chakrabarti, J.; Resmi, M.P.; Wang, J.; Sohal, D.; Shroff, R.T.; Ahmad, S.A.; Zavros, Y. Abstract 233: Cabozantinib changes the landscape of the pancreatic tumor microenvironment and improves the efficacy of pembrolizumab. Cancer Res. 2024, 84, 233. [Google Scholar] [CrossRef]
  77. Su, J.; Zhang, J.; Wu, Y.; Ni, C.; Ding, Y.; Cai, Z.; Xu, M.; Lai, M.; Wang, J.; Lin, S.; et al. Cabozantinib in combination with immune checkpoint inhibitors for renal cell carcinoma: A systematic review and meta-analysis. Front. Pharmacol. 2024, 15, 1322473. [Google Scholar] [CrossRef] [PubMed]
  78. Dilly, J.; Hoffman, M.T.; Abbassi, L.; Li, Z.; Paradiso, F.; Parent, B.D.; Hennessey, C.J.; Jordan, A.C.; Morgado, M.; Dasgupta, S.; et al. Mechanisms of Resistance to Oncogenic KRAS Inhibition in Pancreatic Cancer. Cancer Discov. 2024, 14, 2135–2161. [Google Scholar] [CrossRef] [PubMed]
  79. Sethna, Z.; Guasp, P.; Reiche, C.; Milighetti, M.; Ceglia, N.; Patterson, E.; Lihm, J.; Payne, G.; Lyudovyk, O.; Rojas, L.A.; et al. RNA neoantigen vaccines prime long-lived CD8+ T cells in pancreatic cancer. Nature 2025, 639, 1042–1051. [Google Scholar] [CrossRef]
  80. Igarashi, Y.; Sasada, T. Cancer Vaccines: Toward the Next Breakthrough in Cancer Immunotherapy. J. Immunol. Res. 2020, 2020, 5825401. [Google Scholar] [CrossRef]
  81. 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]
  82. Tsujikawa, T.; Kumar, S.; Borkar, R.N.; Azimi, V.; Thibault, G.; Chang, Y.H.; Balter, A.; Kawashima, R.; Choe, G.; Sauer, D.; et al. Quantitative Multiplex Immunohistochemistry Reveals Myeloid-Inflamed Tumor-Immune Complexity Associated with Poor Prognosis. Cell Rep. 2017, 19, 203–217. [Google Scholar] [CrossRef]
  83. Heumann, T.; Judkins, C.; Li, K.; Lim, S.J.; Hoare, J.; Parkinson, R.; Cao, H.; Zhang, T.; Gai, J.; Celiker, B.; 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. [Google Scholar] [CrossRef]
  84. Cao, J.; Jin, Y.; Li, W.; Zhang, B.; He, Y.; Liu, H.; Xia, N.; Wei, H.; Yan, J. DNA vaccines targeting the encoded antigens to dendritic cells induce potent antitumor immunity in mice. BMC Immunol. 2013, 14, 39. [Google Scholar] [CrossRef] [PubMed]
  85. 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]
  86. Asimgil, H.; Ertetik, U.; Çevik, N.C.; Ekizce, M.; Doğruöz, A.; Gökalp, M.; Arık-Sever, E.; Istvanffy, R.; Friess, H.; Ceyhan, G.O.; et al. Targeting the undruggable oncogenic KRAS: The dawn of hope. JCI Insight 2022, 7, e153688. [Google Scholar] [CrossRef]
  87. Zhang, Y.; Ma, J.A.; Zhang, H.X.; Jiang, Y.N.; Luo, W.H. Cancer vaccines: Targeting KRAS-driven cancers. Expert. Rev. Vaccines 2020, 19, 163–173. [Google Scholar] [CrossRef]
  88. Bannoura, S.F.; Uddin, M.H.; Nagasaka, M.; Fazili, F.; Al-Hallak, M.N.; Philip, P.A.; El-Rayes, B.; Azmi, A.S. Targeting KRAS in pancreatic cancer: New drugs on the horizon. Cancer Metastasis Rev. 2021, 40, 819–835. [Google Scholar] [CrossRef] [PubMed]
  89. Pant, S.; Wainberg, Z.A.; Weekes, C.D.; Furqan, M.; Kasi, P.M.; Devoe, C.E.; Leal, A.D.; Chung, V.; Basturk, O.; VanWyk, H.; et al. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: The phase 1 AMPLIFY-201 trial. Nat. Med. 2024, 30, 531–542. [Google Scholar] [CrossRef] [PubMed]
  90. Haldar, S.D.; Heumann, T.R.; Berg, M.; Ferguson, A.; Lim, S.J.; Wang, H.; Nauroth, J.; Laheru, D.; Jaffee, E.M.; Azad, N.S.; et al. A phase I study of a mutant KRAS-targeted long peptide vaccine combined with ipilimumab/nivolumab in resected pancreatic cancer and MMR-proficient metastatic colorectal cancer. J. Clin. Oncol. 2023, 41, TPS814. [Google Scholar] [CrossRef]
  91. Ni, L. Advances in mRNA-Based Cancer Vaccines. Vaccines 2023, 11, 1599. [Google Scholar] [CrossRef]
  92. Rojas, L.A.; Sethna, Z.; Soares, K.C.; Olcese, C.; Pang, N.; Patterson, E.; Lihm, J.; Ceglia, N.; Guasp, P.; Chu, A.; et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature 2023, 618, 144–150. [Google Scholar] [CrossRef]
  93. Wang, S.; Liang, B.; Wang, W.; Li, L.; Feng, N.; Zhao, Y.; Wang, T.; Yan, F.; Yang, S.; Xia, X. Viral vectored vaccines: Design, development, preventive and therapeutic applications in human diseases. Signal Transduct. Target. Ther. 2023, 8, 149. [Google Scholar] [CrossRef]
  94. 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] [PubMed]
  95. 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] [PubMed]
  96. 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] [PubMed]
Figure 1. The graphical summary of up- and down-regulated cells in the pancreatic cancer tumor microenvironment.
Figure 1. The graphical summary of up- and down-regulated cells in the pancreatic cancer tumor microenvironment.
Cancers 17 01484 g001
Figure 2. Different strategies for enhancing immunotherapy potential in pancreatic cancer.
Figure 2. Different strategies for enhancing immunotherapy potential in pancreatic cancer.
Cancers 17 01484 g002
Table 2. Ongoing clinical trials combining immunotherapy with other agents.
Table 2. Ongoing clinical trials combining immunotherapy with other agents.
CombinationPhaseIndicationIdentifier
DARATUMUMAB (anti-CD38) + KRAS vaccine +
NIVOLUMAB (anti-PD-1)
Phase IIAdvanced PDAC—second-line treatment
* Mutant KRAS
in codon 12 (12A, C, D, R, S, V) or 13D
NCT06015724
DURVALUMAB (anti-PD-1) + RINTATOLIMOD (TLR-3 agonist)Phase I, Phase IIMetastatic PDAC—first-line treatmentNCT05927142
DURVALUMAB (anti-PD-1) + OLECLUMAB (anti-CD73)Phase IIResectable PDAC—treatment before the surgeryNCT06060405
PEMBROLIZUMAB (anti-PD-1) + TALABOSTAT
(DPP8/9 and FAP inhibitor)
Phase IIMetastatic PDAC—second-line treatmentNCT05558982
PENPULIMAB (anti-PD-1) + ANLOTINIB (multi-targeting TKI) + chemotherapy (gemcitabine + nab-paclitaxel)Phase IIMetastatic PC—first-line treatmentNCT06051851
BOTENSILIMAB
(CTLA-4 inhibitor) + chemotherapy (gemcitabine + nab-paclitaxel)
Phase IIMetastatic PDAC—second-line treatmentNCT05630183
TADALAFIL (PDE5 inhibitor) + PEMBROLIZUMAB (anti-PD-1) + IPILIMUMAB (CTLA-4 inhibitor) + CRS-207 (Listeria monocytogenes vaccine)Phase IIMetastatic PDAC—second- or later-line treatmentNCT05014776
AVELUMAB (anti-PD-L1) +
PEPINEMAB (SEMA4D inhibitor)
Phase I, Phase IIMetastatic PDAC—second-line treatmentNCT05102721
Chemotherapy + SBRT + NIVOLUMAB (anti-PD-1) +
IPILIMUMAB (CTLA-4 inhibitor)
Phase IMetastatic PDAC—first-line treatmentNCT05088889
Neoadjuvant chemotherapy (mFOLFIRINOX + PEMBROLIZUMAB (anti-PD-1) + surgery + adjuvant chemotherapy (mFOLFIRINOX) + PEMBROLIZUMABPhase IIResectable PDAC—treatment before and after the surgeryNCT05132504
CABOZANTINIB (multi-targeting TKI) + PEMBROLIZUMAB
(anti-PD-1)
Phase IIMetastatic PDAC—second- or later-line treatmentNCT05052723
LEVANTINIB (multi-targeting TKI) + PEMBROLIZUMAB (anti-PD-1)Phase IIMetastatic/Unresectable PDAC—maintenance (PR/SD after 16 weeks of 1st- or 2nd-line treatment)NCT04887805
NIRAPARIB (PARP inhibitor) + DOSTARLIMAB (anti-PD-1)Phase IIMetastatic PDAC—second- or later-line treatment
* germline or somatic mutations BRCA1/2, PALB2, BARD1,
RAD51C, or RAD51D
NCT04493060
OLAPARIB (PARP inhibitor) + PEMBROLIZUMAB (anti-PD-1)Phase IIMetastatic PDAC
* dMMR or TMB > 4 Mutations/Mb
NCT05093231
XH001 (neoantigen cancer vaccine) + IPILIMUMAB (CTLA-4 inhibitor) + chemotherapy (gemcitabine + capecitabine)Phase I, Phase IIResectable PDAC—treatment after the surgeryNCT06353646
Irreversible electroporation +
PEMBROLIZUMAB (anti-PD-1)
Phase ILocally advanced unresectable PC—treatment after chemotherapy and ablative stereotactic magnetic resonance image-guided adaptive radiation therapyNCT06378047
ANLOTINIB (multi-targeting TKI) + BENMELSTOBART (anti-PD-L1) + chemotherapy (gemcitabine + nab-paclitaxel)Phase IIMetastatic PC—first-line treatmentNCT06621095
SBRT + BOTENSILIMAB
(Fc-enhanced anti-CTLA-4) +
BALSTILIMAB (anti-PD-1)
Phase IIMetastatic PDAC—second- or later-line treatmentNCT06843551
AGEN1423 (anti-CD73-TGF-β-trap) + BOTENSILIMAB
(Fc-enhanced anti-CTLA4)
Phase IIAdvanced PDAC—second- or later-line treatmentNCT05632328
Abbreviations: PDAC—pancreatic ductal adenocarcinoma, PD-1—programmed cell death protein 1, TLR-3—toll-like receptor 3, DPP—dipeptidyl peptidases, FAP—fibroblast activation protein, TKI—tyrosine kinase inhibitor, PDE5—phosphodiesterase 5, CTLA-4—cytotoxic T-lymphocyte–associated antigen 4, SEMA4D—semaphorin 4D, PR—partial response, SD—stable disease, PARP—poly-ADP ribose polymerase, dMMR—mismatch repair deficiency, TMB—tumor mutational burden, SBRT—stereotactic body radiation therapy. *—required mutations.
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

Fudalej, M.; Krupa, K.; Badowska-Kozakiewicz, A.; Deptała, A. Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now? Cancers 2025, 17, 1484. https://doi.org/10.3390/cancers17091484

AMA Style

Fudalej M, Krupa K, Badowska-Kozakiewicz A, Deptała A. Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now? Cancers. 2025; 17(9):1484. https://doi.org/10.3390/cancers17091484

Chicago/Turabian Style

Fudalej, Marta, Kamila Krupa, Anna Badowska-Kozakiewicz, and Andrzej Deptała. 2025. "Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now?" Cancers 17, no. 9: 1484. https://doi.org/10.3390/cancers17091484

APA Style

Fudalej, M., Krupa, K., Badowska-Kozakiewicz, A., & Deptała, A. (2025). Inflammation, Immunosuppression, and Immunotherapy in Pancreatic Cancer—Where Are We Now? Cancers, 17(9), 1484. https://doi.org/10.3390/cancers17091484

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

Article Metrics

Back to TopTop