Next Article in Journal
Integrin α2 and β1 Cross-Communication with mTOR/AKT and the CDK-Cyclin Axis in Hepatocellular Carcinoma Cells
Previous Article in Journal
Current and Emerging Approaches to Study Microenvironmental Interactions and Drug Activity in Classical Hodgkin Lymphoma
Previous Article in Special Issue
Clinical Significance of Tumor-Infiltrating Conventional and Plasmacytoid Dendritic Cells in Pancreatic Ductal Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? A Focus on Tumor Immune Microenvironment, Predictive Biomarkers and Treatment Outcomes

1
Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15, 40138 Bologna, Italy
2
Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Via Giuseppe Massarenti, 9, 40138 Bologna, Italy
3
Osteoncology, Bone and Soft Tissue Sarcomas, and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
4
Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
5
Departmental Unit of Medical Oncology, ASL BA, 20142 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(10), 2429; https://doi.org/10.3390/cancers14102429
Submission received: 30 March 2022 / Revised: 6 May 2022 / Accepted: 11 May 2022 / Published: 14 May 2022

Abstract

:

Simple Summary

In pancreatic cancer, immunotherapy and targeted therapies have not brought about the therapeutic revolution that has been observed in other malignancies. Among the reasons to explain this difference is the possibly crucial role played by the pancreatic tumor microenvironment, which has unique features and is different from that of other neoplasms. The aim of this review is to provide a comprehensive overview of the distinctive tumor immune microenvironment of pancreatic cancer and to summarize existing data about the use of immunotherapy and immune biomarkers in this cancer.

Abstract

The advent of immunotherapy and targeted therapies has dramatically changed the outcomes of patients affected by many malignancies. Pancreatic cancer (PC) remains one the few tumors that is not treated with new generation therapies, as chemotherapy still represents the only effective therapeutic strategy in advanced-stage disease. Agents aiming to reactivate the host immune system against cancer cells, such as those targeting immune checkpoints, failed to demonstrate significant activity, despite the success of these treatments in other tumors. In many cases, the proportion of patients who derived benefits in early-phase trials was too small and unpredictable to justify larger studies. The population of PC patients with high microsatellite instability/mismatch repair deficiency is currently the only population that may benefit from immunotherapy; nevertheless, the prevalence of these alterations is too low to determine a real change in the treatment scenario of this tumor. The reasons for the unsuccess of immunotherapy may lie in the extremely peculiar tumor microenvironment, including distinctive immune composition and cross talk between different cells. These unique features may also explain why the biomarkers commonly used to predict immunotherapy efficacy in other tumors seem to be useless in PC. In the current paper, we provide a comprehensive and up-to-date review of immunotherapy in PC, from the analysis of the tumor immune microenvironment to immune biomarkers and treatment outcomes, with the aim to highlight that simply transferring the knowledge acquired on immunotherapy in other tumors might not be a successful strategy in patients affected by PC.

1. Introduction

Pancreatic cancer (PC) is one of the leading causes of cancer death worldwide [1]. Despite the incidence of PC being much lower than that of other malignancies, such as lung, breast, colorectal, and prostate tumors, it was the third highest cause of cancer-related death in 2021 and is projected to rise to the second position by 2030 [1,2]. As much as 52% of pancreatic tumors show distant dissemination at diagnosis, which confers a 5-year survival rate of only 3%, highlighting the remarkable need to develop more effective therapeutic strategies [1]. Currently, chemotherapy represents the mainstay of the treatment of patients affected by advanced PC [3]. Despite the success of immunotherapy and targeted therapies in many solid tumors in the last decade, these agents were not shown to provide a significant benefit to PC patients [4]. The peculiar pancreatic tumor microenvironment (TME) certainly plays an important role in the lack of success of most therapeutic strategies, including immunotherapy. Studying the tumor immune microenvironment (TIME) is therefore essential to understanding the complex interactions between all the actors involved in cancer immune escape mechanisms and treatment resistance and to learning how to exploit the immune system against PC cells. Moreover, the identification of key features that are able to predict the outcomes of immune-based strategies is necessary to identify that a small proportion of patients that would benefit from these types of treatments. In the current paper, we provide a comprehensive review of the literature, focusing on the immune contexture of PC and its impact on therapeutic strategies aiming to reactivate the immune system against cancer cells. We also analyzed the current knowledge regarding potential predictive and prognostic immune biomarkers and provide an up-to-date overview of immunotherapy in this aggressive tumor.

2. The Pancreatic Tumor Immune Microenvironment

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense and rich stroma, composed of immune cells, blood vessels, fibroblasts, and many other types of cells [5]. Interactions between stroma and cancer cells are responsible for tumor growth, proliferation, and survival, as well as for drug responsiveness and resistance; therefore, the analysis and understanding of TME and TIME are essential to developing valid therapeutic strategies. Many studies have been conducted to understand the association between TIME composition and patient prognoses, but reliable data are still lacking. Different types and subtypes of immune cells are present in TIME in varying percentages and can interact with each other in multiple ways, determining a multitude of different effects (Figure 1) [6,7]. Myeloid cells represent a major component of stroma cells and the high number of tumor-associated macrophages (TAMs) seems to inversely correlate with prognosis in PDAC patients [8,9]. TAMs can promote neo-angiogenesis, cancer cell proliferation, and metastases through the release of cytokines, proteases, and growth factors, such as vascular endothelial growth factor (VEGF) [10,11]. Moreover, TAMs are able to influence the activity of cytidine deaminase, responsible for gemcitabine metabolization, conferring resistance to this drug [12]. In TME, TAMs are differentiated into two subpopulations called “M1” and “M2”, which have opposite roles: M2 have mainly anti-inflammatory functions, while M1 exert anti-cancer effects through the release of pro-inflammatory cytokines [13,14]. Myeloid-derived suppressive cells (MDSCs), which are recruited in PDAC stroma by cancer cells through the production of the granulocyte-macrophage colony-stimulating factor (GM-CSF), play an important anti-inflammatory function in PDAC TIME [15,16]. MDSCs inhibit both innate and adaptive immune responses; in fact, they can block natural killer cells (NK cells) with a direct-contact mechanism and are able to upregulate the expression of programmed death-1 (PD-1) on their surface, favoring the suppression of T-cell activation [17,18,19]. Moreover, MDSCs produce interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), which recruit immunosuppressive regulatory T cells (Tregs) [20]. NK cells are altered in several ways in pancreatic TIME; in normal conditions, NK cells exert direct killing functions on tumor cells, which are independent from antigen stimulation and mediated by cell receptors, such as CD16, natural killer group 2 membrane D (NKG2D), DNAM-1, and natural cytotoxicity receptors (NCRs). Instead, in PDAC, they seem to be polarized towards a less-functioning and tumor-promoting phenotype, a result of a series of complex interactions between NK cells, cancer cells, and other immune cells [21,22]. The increased production of interleukin-10, interleukin-18, and TGFβ and the downregulation of activating receptors are some of the causes of the reduced NK cell function seen in PDAC [23,24]. These “polarized” NK cells are characterized by a lower production of cytotoxic granzyme B and perforin and lower expression of the chemokine receptor CXCR2, with a subsequent impaired tumor trafficking and reactivity [25,26].
Neutrophils have a controversial role in PDAC development. In TIME, tumor-associated neutrophils (TANs) are polarized in two subpopulations named “N1” and “N2” by TGFβ and IFNα, respectively [27]. N1 neutrophils have pro-inflammatory effects and stimulate the recruitment and activation of CD8+ T cells [28], while N2 neutrophils release tumor-promoting factors such as metalloproteinase (MMPs), neutrophil elastase (NE), reactive oxygen, and nitrogen species [29,30]. IL-17, mainly produced by Tregs, acts as an indirect inducer factor of neutrophil extracellular traps production (NETs) [31] and NET formation can promote liver metastasis and immune checkpoint inhibitor (ICI) resistance by blocking CD8+ T cells. Neutrophils are also responsible for the production of lipocalin-2, an adipokine that is implicated in stromal remodeling and tumor cell activation [32].
Tregs have several anti-inflammatory effects, which are primarily mediated by their ability to suppress T-cell activity and are present from the initial stages of the tumorigenesis process [33]. In fact, the number of Treg and Th-17 cells is elevated in premalignant lesions, such as intraductal papillary mucinous neoplasm (IPMNs) and pancreatic intraepithelial neoplasia (PanIN), and seems to be directly correlated with tumor stage and poor prognosis [34,35,36].
In pancreatic TIME, an increased Th2/Th1 tumor-infiltrating lymphocytes ratio has been largely documented [37,38,39]. Th2 role is mediated by the master transcription factor GATA3, which induces M2 macrophage activation and stimulate cancer cell proliferation. Tumor-promoting function is also directly explained by the increased activation of STAT3, AKT, and MAPK pathways [40]. Th2 enrichment in TIME is mediated by different stimulating agents produced by dendritic cells, B cells, TAMs, microbiota, and cancer-associated fibroblast (CAFs) [41,42,43].
Among T cells, CD8+ T lymphocytes have a direct and cytotoxic effect on cancer cells. However, tumors can induce CD8+ T exhaustion, a state characterized by shorter cell survival and impaired effector abilities. A recent study showed that cytotoxic T cells are associated with a longer survival only in patients whose tumors overexpressed targets for pyroptosis and ferroptosis, which are two mechanisms of tumor cell killing. The same study also noted that a high number of T cells overexpressing ribosome-related proteins correlates with better outcomes [44].
As previously mentioned, non-immune cells, such as stellate cells and CAFs, are present in PDAC TIME and play essential roles [45,46,47]. CAFs can be divided into three specific subpopulations. The first is located near cancer cells and has myofibroblastic and anti-tumor features, while the second one is activated by IL-1 and contributes to the generation of an immunosuppressed TIME [48,49]. The role of the third subpopulation, named antigen-presenting CAFs, has not been well established, but they seem to have an immune-suppressive role [47]. Interactions between CAFs, cancer cells, and immune cells are variable and complex. Intra-tumoral or gut commensal bacteria may indirectly increase the expression of IL-1 and consequently the activation of the second subpopulation of CAFs, which is responsible for the production of IL-6, IL-33, CXCL12, IL-8, and other molecules. These cytokines and chemokines promote tumor angiogenesis and bestow chemoresistance and resistance to T-cell killing activity [50,51,52]. Myofibroblastic CAFs are stimulated by TGFβ, which suppresses T-cell activity, polarizes macrophages to M2 subtypes, and is implicated in cell growth, epithelial-to-mesenchymal transition, and extracellular matrix production [53,54]. These functions have made TGFβ an attractive target in pancreatic cancer treatment and have led to the evaluation of its inhibitor, Galunisertib, in combination with chemotherapy in a first-line setting [55]. Other studies have been conducted using pegvorhyaluronidase alfa (PEGPH20), a molecule which degrades hyaluronic acid in TME, with contrasting results [56,57].
Finally, the contribution of microbiota in the generation of an immune-suppressed TIME in PDAC cannot be overlooked. In murine models, bacterial ablation resulted in a lower presence of M2 macrophages and a major presence of CD8+ T-cells and PD-1 expression in TME [58]. ICIs and antibiotics could therefore have a synergistic role, but the toxicities and adverse events of this combination strategy should be further studied and carefully considered.

3. Prognostic and Predictive Immune Biomarkers

The identification of reliable biomarkers able to predict the outcomes of PDAC patients treated with immunotherapy is of paramount importance. In fact, despite the disappointing results generally obtained with ICIs, a small proportion of patients seem to derive durable benefit from immunotherapy [4]. The peculiar immune features of the TIME may predict the prognosis and immune susceptibility of PC patients. The presence of a high tumor mutational burden (TMB) is correlated with a high tumor neoantigen load and is considered as a gross indicator of enhanced immunotherapy efficacy [59]. However, different thresholds have been adopted to define a TMB as “high”, varying across studies and tumor type [60,61]. Some authors have proposed that a high TMB should correspond to the highest TMB quintile in each histology [61]. Median TMB is typically very low in PDAC, ranging around 1–4 mutations/Mb in pancreatic ductal adenocarcinoma and, according to a recent systematic review of the literature, the proportion of TMB-high PDAC is only 1.1% [62,63,64]. Most studies adopted the cut-off of ≥20 mutations/Mb to define a TMB-high PDAC. Interestingly, these tumors show a higher prevalence of mucinous-colloid and medullary histology, which are both usually very rare (<2%), and have a distinctive genomic landscape, including mutations in BRAF, ERBB2, BRCA2, and POLE genes and a high presence (approximately 60%) of high microsatellite instability or mismatch repair deficiency (MSI-H/dMMR) [65]. Among eight patients affected by pancreatic ductal adenocarcinoma with high-TMB and treated with programmed death-1 (PD-1) inhibitors, one patient had stable disease (SD), five had partial responses (PR), and two patients had complete responses (CR), both with MSI-H/dMMR [65]. These data support further studies on the role of TMB as a potential biomarker for PDAC patients treated with immunotherapy.
Programmed death-ligand 1 (PD-L1) expression represents a predictive biomarker of response to ICI in many tumors. In PDAC, a positive PD-L1 expression can be found in approximately 30–40% of cases and is correlated with low tumor-infiltrating lymphocytes, particularly CD8+ cells, and a poor prognosis [66,67]. A recent study identified four PC patterns based on PD-L1 expression on tumor cells (TC) and immune cells (IC): “adaptive-1” (TC: 0, IC > 1%), “adaptive-2” (TC > 1% to <25%, IC > 1%), “constitutive” (TC ≥ 25%, IC: 0), and “combined” (TC ≥ 25%, IC > 1%) [66]. “Adaptive-1” tumors showed a T cell inflamed TIME, characterized by high CD3+, CD4+ and CD8+ cells and PD1+ T cells and low CD68+ macrophages, including the M2-polarized subpopulation, and were associated with the longest survival. Conversely, “constitutive” tumors had reduced IC, except for CD68+ TAMs, and had worst outcomes [66]. A recent meta-analysis exploring the prognostic role of immune infiltration in PDAC documented a negative prognostic impact of CD163+ M2 polarized macrophages [67]. Instead, a high infiltration of CD4+ and CD8+ lymphocytes was associated with longer disease-free survival [67]. PD-L1 expression has been shown to correlate with Cancer-Forkhead box P3, which can promote immune evasion in PDAC by recruiting Forkhead box P3+ Treg cells via CCL5 upregulation [68]. Consistently, ICIs seem to enhance the anti-tumor efficacy of the CCL5 blockade [68]. PD-L1 expression appears to also be positively influenced by mutations in RAS, MYC, and MLL1 genes [69,70,71]. Despite the preclinical rationale, data from clinical studies correlating PD-L1 expression with tumor response to ICI have been inconsistent. The presence of MSI-H/dMMR currently represents the only reliable predictive biomarker of response to ICI in PDAC [72]. Unfortunately, MSI-H/dMMR PDAC represent a very small population, with a prevalence of less than 3% [73,74]. Since this population partially overlaps with TMB-high PDAC, MSI-H/dMMR PDAC share with them several characteristics, such as a higher prevalence of medullary and mucinous/colloid histology and a peculiar genomic background, with more frequent JAK mutations and significantly less common KRAS and TP53 mutations compared to microsatellite stable PDAC [74]. In addition to its recognized oncogenic role, mutant KRAS seems to drive the formation of immunosuppressed TIME, directly preventing innate and adaptative anti-tumor immunity by modulating the levels of cell surface HLA class I and by regulating the expression of CD47 and PD-L1 [75,76,77]. Moreover, KRAS mutations induce a desmoplastic TME composed of suppressive immune cells [76]. Therefore, KRAS inhibition with novel therapies could be a new approach to render PDAC sensitive to immunotherapy [78].

Immunotherapy: Searching for the Right Key

Finding a valid strategy to exploit the host immune system against pancreatic cancer cells is challenging. Antibodies targeting CTLA-4, PD-1, and PD-L1 immune checkpoints demonstrated their efficacy in many tumors, including malignant melanoma, lung, urothelial, and renal cancers. However, the results obtained in patients affected by PDAC have been largely disappointing [4,79]. The number of cells expressing PD-1 and PD-L1 is lower in PDAC compared with tumors where immunotherapy demonstrated an established efficacy, such as malignant melanoma [80]. However, several other immune inhibitory molecules are frequently upregulated, including LAG-3, galectins, TIGIT, and V-domain Ig suppressor of T-cell activation (VISTA) [81,82,83,84]. Efforts to enhance immune infiltration in PDAC TME included targeting CXCR4, which can be inhibited to increase T-cell chemotaxis. In fact, the combination of PD-1 and CXCR4 inhibition resulted in enhanced T-cell expansion and tumor cell death in preclinical models [85]. CD40 activation may represent a strategy to reverse T-cell exhaustion, enhancing the anti-cancer effects of the TIME. Consistently, agonistic CD40 antibodies were shown to increase T-cell mediated cancer death and, in combination with chemotherapy, may rescue ICI sensitivity [86,87,88,89]. Recently, a phase I trial documented the safety profile and the potential activity of combining sotigalimab, a CD40 agonistic monoclonal antibody, with chemotherapy, with or without the PD-1 inhibitor nivolumab, in previously untreated metastatic PDAC [89]. Among 24 dose-limiting toxicity-evaluable patients, a 58% objective response rate (ORR) was reached, and there were no safety concerns. Many clinical trials tested ICI as a single-agent or combined with other agents that may enhance immunotherapy efficacy. CTLA-4 inhibition proved to be poorly effective, either alone or combined with PD-L1 inhibition in two phase II trials testing, respectively, single-agent ipilimumab and durvalumab, with or without tremelimumab, in patients with advanced PDAC [90,91]. Similarly, the addition of single-agent ICI or a dual immune blockade to standard chemotherapy with gemcitabine and nab-paclitaxel did not lead to significant activity or survival improvements [92,93]. In the neoadjuvant setting, adding pembrolizumab to chemoradiation therapy did not improve the efficacy of chemoradiation alone and did not result in significant changes to the infiltration of several immune cell subsets in the TIME [94]. An interesting phase II pilot trial tested the addition of nivolumab and paricalcitol to triple-agent chemotherapy, represented by a combination of nab-paclitaxel, cisplatin, and gemcitabine, in 10 patients with previously untreated metastatic PDAC [95]. Despite the small sample size, the encouraging objective response rate of 80% and disease control rate of 100% warranted further investigation.
Combining ICI with vaccines represents another viable strategy to enhance immunotherapy efficacy. The combination of ipilimumab and GVAX, a granulocyte-macrophage colony-stimulating factor (GM-CSF) cell-based vaccine, enhanced the T-cell repertoire and led to a numerically better overall survival (OS) compared to single-agent ipilimumab (5.7 months vs 3.6 months, p = 0.072), although not reaching statistical significance [96]. Recently, the results of a trial testing the combination of GVAX alone, combined with nivolumab or with nivolumab plus the anti-CD137 urelumab as neoadjuvant or adjuvant therapy for patients with resectable PDAC were presented at the 2022 ASCO Gastrointestinal Cancers Symposium [97]. The full combination regimen (GVAX plus nivolumab plus urelumab) was associated with an improved pathologic response, numerically longer disease-free survival and OS, not reaching statistical significance, and mild toxicity. A phase III trial testing algenpantucel-L, an allogenic vaccine made up of αGal-expressing engineered PDAC cell lines, in combination with adjuvant chemotherapy and chemoradiotherapy did not demonstrate significant efficacy improvements, despite the promising results in terms of disease-free survival (DFS) and OS reported by the phase II single-arm trial [98,99].
Among vaccination strategies, oncolytic viruses have been largely explored in PC, alone or in combination with conventional therapies. The term “oncolytic viruses” refers to natural or genetically modified viruses used as therapeutic agents in various malignancies. These viruses can act directly, causing cancer cell lysis, and also indirectly, modifying TME and promoting cancer regression [100,101,102]. Few encouraging results have been obtained in preclinical and clinical studies evaluating the role of vaccinia, reovirus, herpes simplex-1, and adenovirus as potential oncolytic viruses, probably because of the density and subsequent low penetrability of pancreatic cancer TME that limits the access of viruses [103].
Pelareorep, an isolate of a strain of reovirus, has been studied in combination with gemcitabine in a phase II trial, showing high viral replication in tumor cells and good tolerance [104]. It has been also studied in combination with pembrolizumab and chemotherapy in pretreated patients in a recent phase Ib trial, showing promising results and confirming its safety [105].
Bruton tyrosine kinase (BTK) is expressed by several immune cells, including macrophages [106]. In murine models of PDAC, BTK inhibition promoted the conversion from an M2-like to M1-like macrophage and the differentiation of CD8 T-cells and was associated with enhanced tumor shrinkage when combined with gemcitabine [43]. Acalabrutinib, a BTK inhibitor, was investigated, with or without the addition of pembrolizumab, in patients with previously treated advanced PDAC in a phase II randomized trial [107]. In the combination arm, PR and SD were 7.9% and 21.1%, respectively, vs. 0% and 14.3% in the acalabrutinib alone arm. The presence of high microsatellite instability (MSI-H) was found in approximately 1–2% of PDAC [74,108]. Similarly, a high tumor mutational burden (TMB), defined as ≥20 mutations/megabase, is extremely rare in PDAC [109]. The KEYNOTE-158 trial evaluated the efficacy of pembrolizumab in patients with MSI-H/mismatch repair-deficient advanced non-colorectal cancers [60]. Among 22 MSI-H PDAC patients who received the PD-1 inhibitor, ORR was observed in four (18.2%), including one CR, with a satisfactory median duration of response of 13.4 months. However, the overall results were poor compared to those of patients harboring other tumors, such as gastric and endometrial cancer or cholangiocarcinoma, either in terms of ORR or median PFS (2.1 months) and OS (4.0 months). Currently ongoing phase II and III trials testing immune-based strategies in patients affected by PC are summarized in Table 1.

4. Conclusions

Immunotherapy and targeted therapy did not yield practice-changing results in pancreatic cancer, probably because PDAC TME and TIME are peculiar compared to most tumors, as is the genomic landscape that accompanies this disease. The simple transfer of knowledge about the efficacy of immunotherapy and targeted therapy in other neoplasms is not sufficient to obtain promising results in pancreatic cancer and needs to be integrated with a deeper comprehension of TME cells and their interactions. More research efforts are crucial to better select patients that could benefit from immunotherapy and to develop efficient TME modification mechanisms that could make the tumor more immunosensitive. Identifying biomarkers able to predict clinical outcomes of pancreatic cancer patients treated with immunotherapy and targeted therapies, regardless of the disease setting, is of paramount importance.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. National Cancer Institute: Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Pancreatic Cancer. 2022. Available online: https://seer.cancer.gov/statfacts/html/pancreas (accessed on 5 March 2022).
  2. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting Cancer Incidence and Deaths to 2030: The Unexpected Burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Brunner, M.; Wu, Z.; Krautz, C.; Pilarsky, C.; Grützmann, R.; Weber, G.F. Current Clinical Strategies of Pancreatic Cancer Treatment and Open Molecular Questions. Int. J. Mol. Sci. 2019, 20, 4543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Henriksen, A.; Dyhl-Polk, A.; Chen, I.; Nielsen, D. Checkpoint Inhibitors in Pancreatic Cancer. Cancer Treat. Rev. 2019, 78, 17–30. [Google Scholar] [CrossRef] [PubMed]
  5. Farrow, B.; Albo, D.; Berger, D.H. The Role of the Tumor Microenvironment in the Progression of Pancreatic Cancer. J. Surg. Res. 2008, 149, 319–328. [Google Scholar] [CrossRef]
  6. Balachandran, V.P.; Beatty, G.L.; Dougan, S.K. Broadening the Impact of Immunotherapy to Pancreatic Cancer: Challenges and Opportunities. Gastroenterology 2019, 156, 2056–2072. [Google Scholar] [CrossRef]
  7. Ligorio, M.; Sil, S.; Malagon-Lopez, J.; Nieman, L.T.; Misale, S.; Di Pilato, M.; Ebright, R.Y.; Karabacak, M.N.; Kulkarni, A.S.; Liu, A.; et al. Stromal Microenvironment Shapes the Intratumoral Architecture of Pancreatic Cancer. Cell 2019, 178, 160–175.e27. [Google Scholar] [CrossRef]
  8. Gordon, S.; Taylor, P.R. Monocyte and Macrophage Heterogeneity. Nat. Rev. Immunol. 2005, 5, 953–964. [Google Scholar] [CrossRef]
  9. Di Caro, G.; Cortese, N.; Castino, G.F.; Grizzi, F.; Gavazzi, F.; Ridolfi, C.; Capretti, G.; Mineri, R.; Todoric, J.; Zerbi, A.; et al. Dual Prognostic Significance of Tumour-Associated Macrophages in Human Pancreatic Adenocarcinoma Treated or Untreated with Chemotherapy. Gut 2016, 65, 1710–1720. [Google Scholar] [CrossRef]
  10. Griesmann, H.; Drexel, C.; Milosevic, N.; Sipos, B.; Rosendahl, J.; Gress, T.M.; Michl, P. Pharmacological Macrophage Inhibition Decreases Metastasis Formation in a Genetic Model of Pancreatic Cancer. Gut 2017, 66, 1278–1285. [Google Scholar] [CrossRef]
  11. Filippini, D.; Agosto, S.D.; Delfino, P.; Simbolo, M.; Piro, G.; Rusev, B.; Veghini, L.; Cantù, C.; Lupo, F.; Ugel, S.; et al. Immunoevolution of Mouse Pancreatic Organoid Isografts from Preinvasive to Metastatic Disease. Sci. Rep. 2019, 9, 12286. [Google Scholar] [CrossRef] [Green Version]
  12. Weizman, N.; Krelin, Y.; Shabtay-Orbach, A.; Amit, M.; Binenbaum, Y.; Wong, R.J.; Gil, Z. Macrophages Mediate Gemcitabine Resistance of Pancreatic Adenocarcinoma by Upregulating Cytidine Deaminase. Oncogene 2014, 33, 3812–3819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Biswas, S.K.; Mantovani, A. Macrophage Plasticity and Interaction with Lymphocyte Subsets: Cancer as a Paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef] [PubMed]
  14. Mosser, D.M.; Edwards, J.P. Exploring the Full Spectrum of Macrophage Activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef] [PubMed]
  15. Bayne, L.J.; Beatty, G.L.; Jhala, N.; Clark, C.E.; Rhim, A.D.; Stanger, B.Z.; Vonderheide, R.H. Tumor-Derived Granulocyte-Macrophage Colony-Stimulating Factor Regulates Myeloid Inflammation and T Cell Immunity in Pancreatic Cancer. Cancer Cell 2012, 21, 822–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Pylayeva-Gupta, Y.; Lee, K.E.; Hajdu, C.H.; Miller, G.; Bar-Sagi, D. Oncogenic Kras-Induced GM-CSF Production Promotes the Development of Pancreatic Neoplasia. Cancer Cell 2012, 21, 836–847. [Google Scholar] [CrossRef] [Green Version]
  17. Ostrand-Rosenberg, S.; Sinha, P.; Beury, D.W.; Clements, V.K. Cross-Talk between Myeloid-Derived Suppressor Cells (MDSC), Macrophages, and Dendritic Cells Enhances Tumor-Induced Immune Suppression. Semin. Cancer Biol. 2012, 22, 275–281. [Google Scholar] [CrossRef] [Green Version]
  18. Sinha, P.; Clements, V.K.; Bunt, S.K.; Albelda, S.M.; Ostrand-Rosenberg, S. Cross-Talk between Myeloid-Derived Suppressor Cells and Macrophages Subverts Tumor Immunity toward a Type 2 Response. J. Immunol. 2007, 179, 977–983. [Google Scholar] [CrossRef]
  19. Pinton, L.; Solito, S.; Damuzzo, V.; Francescato, S.; Pozzuoli, A.; Berizzi, A.; Mocellin, S.; Rossi, C.R.; Bronte, V.; Mandruzzato, S. Activated T Cells Sustain Myeloid-Derived Suppressor Cell-Mediated Immune Suppression. Oncotarget 2016, 7, 1168–1184. [Google Scholar] [CrossRef] [Green Version]
  20. Huang, B.; Pan, P.Y.; Li, Q.; Sato, A.I.; Levy, D.E.; Bromberg, J.; Divino, C.M.; Chen, S.H. 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, 1123–1131. [Google Scholar] [CrossRef] [Green Version]
  21. Liu, S.; Dhar, P.; Wu, J.D. NK Cell Plasticity in Cancer. J. Clin. Med. 2019, 8, 1492. [Google Scholar] [CrossRef] [Green Version]
  22. Carrega, P.; Bonaccorsi, I.; Di Carlo, E.; Morandi, B.; Paul, P.; Rizzello, V.; Cipollone, G.; Navarra, G.; Mingari, M.C.; Moretta, L.; et al. CD56(Bright)Perforin(Low) Noncytotoxic Human NK Cells Are Abundant in Both Healthy and Neoplastic Solid Tissues and Recirculate to Secondary Lymphoid Organs via Afferent Lymph. J. Immunol. 2014, 192, 3805–3815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wahl, S.M.; Wen, J.; Moutsopoulos, N.M. The Kiss of Death: Interrupted by NK-Cell Close Encounters of Another Kind. Trends Immunol. 2006, 27, 161–164. [Google Scholar] [CrossRef] [PubMed]
  24. Ghiringhelli, F.; Ménard, C.; Terme, M.; Flament, C.; Taieb, J.; Chaput, N.; Puig, P.E.; Novault, S.; Escudier, B.; Vivier, E.; et al. CD4+CD25+ Regulatory T Cells Inhibit Natural Killer Cell Functions in a Transforming Growth Factor-Beta-Dependent Manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
  25. Funa, K.; Nilsson, B.; Jacobsson, G.; Alm, G.V. Decreased Natural Killer Cell Activity and Interferon Production by Leucocytes in Patients with Adenocarcinoma of the Pancreas. Br. J. Cancer 1984, 50, 231–233. [Google Scholar] [CrossRef] [Green Version]
  26. Lim, S.A.; Kim, J.; Jeon, S.; Shin, M.H.; Kwon, J.; Kim, T.J.; Im, K.; Han, Y.; Kwon, W.; Kim, S.W.; et al. Defective Localization with Impaired Tumor Cytotoxicity Contributes to the Immune Escape of NK Cells in Pancreatic Cancer Patients. Front. Immunol. 2019, 10, 496. [Google Scholar] [CrossRef] [Green Version]
  27. Hirth, M.; Gandla, J.; Höper, C.; Gaida, M.M.; Agarwal, N.; Simonetti, M.; Demir, A.; Xie, Y.; Weiss, C.; Michalski, C.W.; et al. CXCL10 and CCL21 Promote Migration of Pancreatic Cancer Cells Toward Sensory Neurons and Neural Remodeling in Tumors in Mice, Associated with Pain in Patients. Gastroenterology 2020, 159, 665–681.e13. [Google Scholar] [CrossRef]
  28. Mollinedo, F. Neutrophil Degranulation, Plasticity, and Cancer Metastasis. Trends Immunol. 2019, 40, 228–242. [Google Scholar] [CrossRef]
  29. Xiang, Z.J.; Hu, T.; Wang, Y.; Wang, H.; Xu, L.; Cui, N. Neutrophil-Lymphocyte Ratio (NLR) Was Associated with Prognosis and Immunomodulatory in Patients with Pancreatic Ductal Adenocarcinoma (PDAC). Biosci. Rep. 2020, 40, BSR20201190. [Google Scholar] [CrossRef]
  30. Brandau, S.; Dumitru, C.A.; Lang, S. Protumor and Antitumor Functions of Neutrophil Granulocytes. Semin. Immunopathol. 2013, 35, 163–176. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Chandra, V.; Sanchez, E.R.; Dutta, P.; Quesada, P.R.; Rakoski, A.; Zoltan, M.; Arora, N.; Baydogan, S.; Horne, W.; et al. Interleukin-17-Induced Neutrophil Extracellular Traps Mediate Resistance to Checkpoint Blockade in Pancreatic Cancer. J. Exp. Med. 2020, 217, 20190354. [Google Scholar] [CrossRef]
  32. Tao, L.; Zhang, L.; Peng, Y.; Tao, M.; Li, L.; Xiu, D.; Yuan, C.; Ma, Z.; Jiang, B. Neutrophils Assist the Metastasis of Circulating Tumor Cells in Pancreatic Ductal Adenocarcinoma: A New Hypothesis and a New Predictor for Distant Metastasis. Medicine 2016, 95, e4932. [Google Scholar] [CrossRef] [PubMed]
  33. Nasca, V.; Chiaravalli, M.; Piro, G.; Esposito, A.; Salvatore, L.; Tortora, G.; Corbo, V.; Carbone, C. Intraductal Pancreatic Mucinous Neoplasms: A Tumor-Biology Based Approach for Risk Stratification. Int. J. Mol. Sci. 2020, 21, 6386. [Google Scholar] [CrossRef] [PubMed]
  34. Vizio, B.; Novarino, A.; Giacobino, A.; Cristiano, C.; Prati, A.; Ciuffreda, L.; Montrucchio, G.; Bellone, G. Potential Plasticity of T Regulatory Cells in Pancreatic Carcinoma in Relation to Disease Progression and Outcome. Exp. Ther. Med. 2012, 4, 70–78. [Google Scholar] [CrossRef] [Green Version]
  35. Kleinewietfeld, M.; Hafler, D.A. The Plasticity of Human Treg and Th17 Cells and Its Role in Autoimmunity. Semin. Immunol. 2013, 25, 305–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. He, S.; Fei, M.; Wu, Y.; Zheng, D.; Wan, D.; Wang, L.; Li, D. Distribution and Clinical Significance of Th17 Cells in the Tumor Microenvironment and Peripheral Blood of Pancreatic Cancer Patients. Int. J. Mol. Sci. 2011, 12, 7424–7437. [Google Scholar] [CrossRef] [PubMed]
  37. Dobrzanski, M.J. Expanding Roles for CD4 T Cells and Their Subpopulations in Tumor Immunity and Therapy. Front. Oncol. 2013, 3, 63. [Google Scholar] [CrossRef] [Green Version]
  38. De Monte, L.; Reni, M.; Tassi, E.; Clavenna, D.; Papa, I.; Recalde, H.; Braga, M.; Di Carlo, V.; Doglioni, C.; Protti, M.P. Intratumor T Helper Type 2 Cell Infiltrate Correlates with Cancer-Associated Fibroblast Thymic Stromal Lymphopoietin Production and Reduced Survival in Pancreatic Cancer. J. Exp. Med. 2011, 208, 469–478. [Google Scholar] [CrossRef]
  39. Tassi, E.; Gavazzi, F.; Albarello, L.; Senyukov, V.; Longhi, R.; Dellabona, P.; Doglioni, C.; Braga, M.; Di Carlo, V.; Protti, M.P. Carcinoembryonic Antigen-Specific but Not Antiviral CD4+ T Cell Immunity Is Impaired in Pancreatic Carcinoma Patients. J. Immunol. 2008, 181, 6595–6603. [Google Scholar] [CrossRef] [Green Version]
  40. Piro, G.; Simionato, F.; Carbone, C.; Frizziero, M.; Malleo, G.; Zanini, S.; Casolino, R.; Santoro, R.; Mina, M.M.; Zecchetto, C.; et al. A Circulating T H 2 Cytokines Profile Predicts Survival in Patients with Resectable Pancreatic Adenocarcinoma. Oncoimmunology 2017, 6, e1322242. [Google Scholar] [CrossRef] [Green Version]
  41. Prokopchuk, O.; Liu, Y.; Henne-Bruns, D.; Kornmann, M. Interleukin-4 Enhances Proliferation of Human Pancreatic Cancer Cells: Evidence for Autocrine and Paracrine Actions. Br. J. Cancer 2005, 92, 921–928. [Google Scholar] [CrossRef] [Green Version]
  42. De Monte, L.; Wörmann, S.; Brunetto, E.; Heltai, S.; Magliacane, G.; Reni, M.; Paganoni, A.M.; Recalde, H.; Mondino, A.; Falconi, M.; et al. Basophil Recruitment into Tumor-Draining Lymph Nodes Correlates with Th2 Inflammation and Reduced Survival in Pancreatic Cancer Patients. Cancer Res. 2016, 76, 1792–1803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gunderson, A.J.; Kaneda, M.M.; Tsujikawa, T.; Nguyen, A.V.; Affara, N.I.; Ruffell, B.; Gorjestani, S.; Liudahl, S.M.; Truit, M.; Olson, P.; et al. Bruton Tyrosine Kinase-Dependent Immune Cell Cross-Talk Drives Pancreas Cancer. Cancer Discov. 2016, 6, 270–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Tang, R.; Liu, X.; Liang, C.; Hua, J.; Xu, J.; Wang, W.; Meng, Q.; Liu, J.; Zhang, B.; Yu, X.; et al. Deciphering the Prognostic Implications of the Components and Signatures in the Immune Microenvironment of Pancreatic Ductal Adenocarcinoma. Front. Immunol. 2021, 12, 648917. [Google Scholar] [CrossRef] [PubMed]
  45. Neesse, A.; Bauer, C.A.; Öhlund, D.; Lauth, M.; Buchholz, M.; Michl, P.; Tuveson, D.A.; Gress, T.M. Stromal Biology and Therapy in Pancreatic Cancer: Ready for Clinical Translation? Gut 2019, 68, 159–171. [Google Scholar] [CrossRef] [PubMed]
  46. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct Populations of Inflammatory Fibroblasts and Myofibroblasts in Pancreatic Cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef] [PubMed]
  47. Elyada, E.; Bolisetty, M.; Laise, P.; Flynn, W.F.; Courtois, E.T.; Burkhart, R.A.; Teinor, J.A.; Belleau, P.; Biffi, G.; Lucito, M.S.; et al. Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov. 2019, 9, 1102–1123. [Google Scholar] [CrossRef] [Green Version]
  48. Belle, J.I.; Denardo, D.G. A Single-Cell Window into Pancreas Cancer Fibroblast Heterogeneity. Cancer Discov. 2019, 9, 1001–1002. [Google Scholar] [CrossRef] [Green Version]
  49. Das, S.; Shapiro, B.; Vucic, E.A.; Vogt, S.; Bar-Sagi, D. Tumor Cell-Derived IL1β Promotes Desmoplasia and Immune Suppression in Pancreatic Cancer. Cancer Res. 2020, 80, 1088–1101. [Google Scholar] [CrossRef] [Green Version]
  50. Noel, M.; O’Reilly, E.M.; Wolpin, B.M.; Ryan, D.P.; Bullock, A.J.; Britten, C.D.; Linehan, D.C.; Belt, B.A.; Gamelin, E.C.; Ganguly, B.; et al. Phase 1b Study of a Small Molecule Antagonist of Human Chemokine (C-C Motif) Receptor 2 (PF-04136309) in Combination with Nab-Paclitaxel/Gemcitabine in First-Line Treatment of Metastatic Pancreatic Ductal Adenocarcinoma. Investig. New Drugs 2020, 38, 800–811. [Google Scholar] [CrossRef] [Green Version]
  51. Kim, M.G.; Shon, Y.; Kim, J.; Oh, Y.K. Selective Activation of Anticancer Chemotherapy by Cancer-Associated Fibroblasts in the Tumor Microenvironment. J. Natl. Cancer Inst. 2016, 109, djw186. [Google Scholar] [CrossRef] [Green Version]
  52. Albrengues, J.; Bourget, I.; Pons, C.; Butet, V.; Hofman, P.; Tartare-Deckert, S.; Feral, C.C.; Meneguzzi, G.; Gaggioli, C. LIF Mediates Proinvasive Activation of Stromal Fibroblasts in Cancer. Cell Rep. 2014, 7, 1664–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kaur, A.; Ecker, B.L.; Douglass, S.M.; Kugel, C.H.; Webster, M.R.; Almeida, F.V.; Somasundaram, R.; Hayden, J.; Ban, E.; Ahmadzadeh, H.; et al. Remodeling of the Collagen Matrix in Aging Skin Promotes Melanoma Metastasis and Affects Immune Cell Motility. Cancer Discov. 2019, 9, 64–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Massagué, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Melisi, D.; Garcia-Carbonero, R.; Macarulla, T.; Pezet, D.; Deplanque, G.; Fuchs, M.; Trojan, J.; Oettle, H.; Kozloff, M.; Cleverly, A.; et al. Galunisertib plus Gemcitabine vs. Gemcitabine for First-Line Treatment of Patients with Unresectable Pancreatic Cancer. Br. J. Cancer 2018, 119, 1208–1214. [Google Scholar] [CrossRef] [Green Version]
  56. Hingorani, S.R.; Zheng, L.; Bullock, A.J.; Seery, T.E.; Harris, W.P.; Sigal, D.S.; Braiteh, F.; Ritch, P.S.; Zalupski, M.M.; Bahary, N.; et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine versus Nab-Paclitaxel/Gemcitabine in Patients with Untreated, Metastatic Pancreatic Ductal Adenocarcinoma. J. Clin. Oncol. 2018, 36, 359–366. [Google Scholar] [CrossRef]
  57. Ramanathan, R.K.; McDonough, S.L.; Philip, P.A.; Hingorani, S.R.; Lacy, J.; Kortmansky, J.S.; Thumar, J.; Chiorean, E.G.; Shields, A.F.; Behl, D.; et al. Phase IB/II Randomized Study of FOLFIRINOX Plus Pegylated Recombinant Human Hyaluronidase Versus FOLFIRINOX Alone in Patients with Metastatic Pancreatic Adenocarcinoma: SWOG S1313. J. Clin. Oncol. 2019, 37, 1062–1069. [Google Scholar] [CrossRef]
  58. Madden, J. Infinity reports update from Phase 2 study of Saridegib plus Gemcitabine in patients with metastatic pancreatic cancer. Infin. Pharm. 2012, 2012–2014. [Google Scholar]
  59. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational Burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
  60. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H.; et al. Association of Tumour Mutational Burden with Outcomes in Patients with Advanced Solid Tumours Treated with Pembrolizumab: Prospective Biomarker Analysis of the Multicohort, Open-Label, Phase 2 KEYNOTE-158 Study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef]
  61. Samstein, R.M.; Lee, C.H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor Mutational Load Predicts Survival after Immunotherapy across Multiple Cancer Types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef]
  62. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of Mutational Processes in Human Cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lee, B.; Gibbs, P. Inflammation, Biomarkers and Immuno-Oncology Pathways in Pancreatic Cancer. J. Pers. Med. 2019, 9, 20. [Google Scholar] [CrossRef] [Green Version]
  64. Lawlor, R.T.; Mattiolo, P.; Mafficini, A.; Hong, S.M.; Piredda, M.L.; Taormina, S.V.; Malleo, G.; Marchegiani, G.; Pea, A.; Salvia, R.; et al. Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Pancreatic Cancer: Systematic Review and Still-Open Questions. Cancers 2021, 13, 3119. [Google Scholar] [CrossRef]
  65. Nomi, T.; Sho, M.; Akahori, T.; Hamada, K.; Kubo, A.; Kanehiro, H.; Nakamura, S.; Enomoto, K.; Yagita, H.; Azuma, M.; et al. Clinical Significance and Therapeutic Potential of the Programmed Death-1 Ligand/Programmed Death-1 Pathway in Human Pancreatic Cancer. Clin. Cancer Res. 2007, 13, 2151–2157. [Google Scholar] [CrossRef] [Green Version]
  66. Karamitopoulou, E.; Andreou, A.; de Mortanges, A.P.; Tinguely, M.; Gloor, B.; Perren, A. PD-1/PD-L1-Associated Immunoarchitectural Patterns Stratify Pancreatic Cancer Patients into Prognostic/Predictive Subgroups. Cancer Immunol. Res. 2021, 9, 1439–1450. [Google Scholar] [CrossRef] [PubMed]
  67. McGuigan, A.J.; Coleman, H.G.; McCain, R.S.; Kelly, P.J.; Johnston, D.I.; Taylor, M.A.; Turkington, R.C. Immune Cell Infiltrates as Prognostic Biomarkers in Pancreatic Ductal Adenocarcinoma: A Systematic Review and Meta-Analysis. J. Pathol. Clin. Res. 2021, 7, 99–112. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, X.; Li, X.; Wei, X.; Jiang, H.; Lan, C.; Yang, S.; Wang, H.; Yang, Y.; Tian, C.; Xu, Z.; et al. PD-L1 Is a Direct Target of Cancer-FOXP3 in Pancreatic Ductal Adenocarcinoma (PDAC), and Combined Immunotherapy with Antibodies against PD-L1 and CCL5 Is Effective in the Treatment of PDAC. Signal Transduct. Target. Ther. 2020, 5, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Coelho, M.A.; de Carné Trécesson, S.; Rana, S.; Zecchin, D.; Moore, C.; Molina-Arcas, M.; East, P.; Spencer-Dene, B.; Nye, E.; Barnouin, K.; et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 MRNA. Immunity 2017, 47, 1083–1099.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Casey, S.C.; Tong, L.; Li, Y.; Do, R.; Walz, S.; Fitzgerald, K.N.; Gouw, A.M.; Baylot, V.; Gütgemann, I.; Eilers, M.; et al. MYC Regulates the Antitumor Immune Response through CD47 and PD-L1. Science 2016, 352, 227–231. [Google Scholar] [CrossRef] [Green Version]
  71. Lu, C.; Paschall, A.V.; Shi, H.; Savage, N.; Waller, J.L.; Sabbatini, M.E.; Oberlies, N.H.; Pearce, C.; Liu, K. The MLL1-H3K4me3 Axis-Mediated PD-L1 Expression and Pancreatic Cancer Immune Evasion. J. Natl. Cancer Inst. 2017, 109, djw283. [Google Scholar] [CrossRef] [Green Version]
  72. Lemery, S.; Keegan, P.; Pazdur, R. First FDA Approval Agnostic of Cancer Site—When a Biomarker Defines the Indication. N. Engl. J. Med. 2017, 377, 1409–1412. [Google Scholar] [CrossRef] [PubMed]
  73. Macherla, S.; Laks, S.; Naqash, A.R.; Bulumulle, A.; Zervos, E.; Muzaffar, M. Emerging Role of Immune Checkpoint Blockade in Pancreatic Cancer. Int. J. Mol. Sci. 2018, 19, 3505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Luchini, C.; Brosens, L.A.A.; Wood, L.D.; Chatterjee, D.; Shin, J.I.; Sciammarella, C.; Fiadone, G.; Malleo, G.; Salvia, R.; Kryklyva, V.; et al. Comprehensive Characterisation of Pancreatic Ductal Adenocarcinoma with Microsatellite Instability: Histology, Molecular Pathology and Clinical Implications. Gut 2021, 70, 148–156. [Google Scholar] [CrossRef] [PubMed]
  75. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC Proteins Confer Differential Sensitivity to CTLA-4 and PD-1 Blockade in Untreated Metastatic Melanoma. Sci. Transl. Med. 2018, 10, eaar3342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Carvalho, P.D.; Guimarães, C.F.; Cardoso, A.P.; Mendonça, S.; Costa, Â.M.; Oliveira, M.J.; Velho, S. KRAS Oncogenic Signaling Extends beyond Cancer Cells to Orchestrate the Microenvironment. Cancer Res. 2018, 78, 7–14. [Google Scholar] [CrossRef] [Green Version]
  77. El-Jawhari, J.J.; El-Sherbiny, Y.M.; Scott, G.B.; Morgan, R.S.M.; Prestwich, R.; Bowles, P.A.; Blair, G.E.; Tanaka, T.; Rabbitts, T.H.; Meade, J.L.; et al. Blocking Oncogenic RAS Enhances Tumour Cell Surface MHC Class I Expression but Does Not Alter Susceptibility to Cytotoxic Lymphocytes. Mol. Immunol. 2014, 58, 160–168. [Google Scholar] [CrossRef] [Green Version]
  78. 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]
  79. Hester, R.; Mazur, P.K.; McAllister, F. Immunotherapy in Pancreatic Adenocarcinoma: Beyond “Copy/Paste”. Clin. Cancer Res. 2021, 27, 6287–6297. [Google Scholar] [CrossRef]
  80. Blando, J.; Sharma, A.; Higa, M.G.; Zhao, H.; Vence, L.; Yadav, S.S.; Kim, J.; Sepulveda, A.M.; Sharp, M.; Maitra, A.; et al. Comparison of Immune Infiltrates in Melanoma and Pancreatic Cancer Highlights VISTA as a Potential Target in Pancreatic Cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 1692–1697. [Google Scholar] [CrossRef] [Green Version]
  81. Meng, Q.; Liu, Z.; Rangelova, E.; Poiret, T.; Ambati, A.; Rane, L.; Xie, S.; Verbeke, C.; Dodoo, E.; Del Chiaro, M.; et al. Expansion of Tumor-Reactive T Cells from Patients with Pancreatic Cancer. J. Immunother. 2016, 39, 81–89. [Google Scholar] [CrossRef] [Green Version]
  82. Manero-Rupérez, N.; Martínez-Bosch, N.; Barranco, L.E.; Visa, L.; Navarro, P. The Galectin Family as Molecular Targets: Hopes for Defeating Pancreatic Cancer. Cells 2020, 9, 689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Stanietsky, N.; Simic, H.; Arapovic, J.; Toporik, A.; Levy, O.; Novik, A.; Levine, Z.; Beiman, M.; Dassa, L.; Achdout, H.; et al. The Interaction of TIGIT with PVR and PVRL2 Inhibits Human NK Cell Cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 17858–17863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The Surface Protein TIGIT Suppresses T Cell Activation by Promoting the Generation of Mature Immunoregulatory Dendritic Cells. Nat. Immunol. 2009, 10, 48–57. [Google Scholar] [CrossRef] [PubMed]
  85. Seo, Y.D.; Jiang, X.; Sullivan, K.M.; Jalikis, F.G.; Smythe, K.S.; Abbasi, A.; Vignali, M.; Park, J.O.; Daniel, S.K.; Pollack, S.M.; et al. Mobilization of CD8 + T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 3934–3945. [Google Scholar] [CrossRef] [Green Version]
  86. Vonderheide, R.H. The Immune Revolution: A Case for Priming, Not Checkpoint. Cancer Cell 2018, 33, 563–569. [Google Scholar] [CrossRef] [Green Version]
  87. Byrne, K.T.; Vonderheide, R.H. CD40 Stimulation Obviates Innate Sensors and Drives T Cell Immunity in Cancer. Cell Rep. 2016, 15, 2719–2732. [Google Scholar] [CrossRef] [Green Version]
  88. Winograd, R.; Byrne, K.T.; Evans, R.A.; Odorizzi, P.M.; Meyer, A.R.L.; Bajor, D.L.; Clendenin, C.; Stanger, B.Z.; Furth, E.E.; Wherry, E.J.; et al. Induction of T-Cell Immunity Overcomes Complete Resistance to PD-1 and CTLA-4 Blockade and Improves Survival in Pancreatic Carcinoma. Cancer Immunol. Res. 2015, 3, 399–411. [Google Scholar] [CrossRef] [Green Version]
  89. O’Hara, M.H.; O’Reilly, E.M.; Varadhachary, G.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; Cabanski, C.R.; et al. CD40 Agonistic Monoclonal Antibody APX005M (Sotigalimab) and Chemotherapy, with or without Nivolumab, for the Treatment of Metastatic Pancreatic Adenocarcinoma: An Open-Label, Multicentre, Phase 1b Study. Lancet Oncol. 2021, 22, 118–131. [Google Scholar] [CrossRef]
  90. Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 Trial of Single Agent Ipilimumab (Anti-CTLA-4) for Locally Advanced or Metastatic Pancreatic Adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef]
  91. O’Reilly, E.M.; Oh, D.Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.; Hezel, A.; Chang, S.C.; Vlahovic, G.; et al. Durvalumab with or without Tremelimumab for Patients with Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019, 5, 1431–1438. [Google Scholar] [CrossRef]
  92. Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schütz, E.; Khemka, V. Phase Ib/II Study of Gemcitabine, Nab-Paclitaxel, and Pembrolizumab in Metastatic Pancreatic Adenocarcinoma. Investig. New Drugs 2018, 36, 96–102. [Google Scholar] [CrossRef] [PubMed]
  93. Renouf, D.J.; Knox, J.J.; Kavan, P.; Jonker, D.; Welch, S.; Couture, F.; Lemay, F.; Tehfe, M.; Harb, M.; Aucoin, N.; et al. LBA65 The Canadian Cancer Trials Group PA.7 Trial: Results of a Randomized Phase II Study of Gemcitabine (GEM) and Nab-Paclitaxel (Nab-P) vs GEM, Nab-P, Durvalumab (D) and Tremelimumab (T) as First Line Therapy in Metastatic Pancreatic Ductal Adenocarcinoma (MPDAC). Ann. Oncol. 2020, 31, S1195. [Google Scholar] [CrossRef]
  94. Rahma, O.E.; Katz, M.H.G.; Wolpin, B.M.; Dias-Costa, A.; Nowak, J.; Rodig, S.J.; Dougan, S.; Bekaii-Saab, T.S.; Stucky, C.-C.H.; Elias, R.; et al. Randomized Multicenter Phase Ib/II Study of Neoadjuvant Chemoradiation Therapy (CRT) Alone or in Combination with Pembrolizumab in Patients with Resectable or Borderline Resectable Pancreatic Cancer. J. Clin. Oncol. 2021, 39, 4128. [Google Scholar] [CrossRef]
  95. Borazanci, E.H.; Jameson, G.S.; Borad, M.J.; Ramanathan, R.K.; Korn, R.L.; Caldwell, L.; Ansaldo, K.; Hendrickson, K.; Marceau, K.; Von Hoff, D.D. A Phase II Pilot Trial of Nivolumab (N) + Albumin Bound Paclitaxel (AP) + Paricalcitol (P) + Cisplatin (C) + Gemcitabine (G) (NAPPCG) in Patients with Previously Untreated Metastatic Pancreatic Ductal Adenocarcinoma (PDAC). J. Clin. Oncol. 2018, 36, 358. [Google Scholar] [CrossRef]
  96. Le, D.T.; Lutz, E.; Uram, J.N.; Sugar, E.A.; Onners, B.; Solt, S.; Zheng, L.; Diaz, L.A.; Donehower, R.C.; Jaffee, E.M.; et al. Evaluation of Ipilimumab in Combination with Allogeneic Pancreatic Tumor Cells Transfected with a GM-CSF Gene in Previously Treated Pancreatic Cancer. J. Immunother. 2013, 36, 382–389. [Google Scholar] [CrossRef] [Green Version]
  97. Heumann, T.R.; Judkins, C.; Lim, S.J.; Wang, H.; Parkinson, R.; Gai, J.; Celiker, B.; Durham, J.N.; Laheru, D.A.; De Jesus-Acosta, A.; et al. Neoadjuvant and Adjuvant Antitumor Vaccination Alone or Combination with PD1 Blockade and CD137 Agonism in Patients with Resectable Pancreatic Adenocarcinoma. J. Clin. Oncol. 2022, 40, 558. [Google Scholar] [CrossRef]
  98. Hardacre, J.M.; Mulcahy, M.; Small, W.; Talamonti, M.; Obel, J.; Krishnamurthi, S.; Rocha-Lima, C.S.; Safran, H.; Lenz, H.J.; Chiorean, E.G. Addition of Algenpantucel-L Immunotherapy to Standard Adjuvant Therapy for Pancreatic Cancer: A Phase 2 Study. J. Gastrointest. Surg. 2013, 17, 94–101. [Google Scholar] [CrossRef]
  99. Hewitt, D.B.; Nissen, N.; Hatoum, H.; Musher, B.; Seng, J.; Coveler, A.L.; Al-Rajabi, R.; Yeo, C.J.; Leiby, B.; Banks, J.; et al. A Phase 3 Randomized Clinical Trial of Chemotherapy with or without Algenpantucel-L (HyperAcute-Pancreas) Immunotherapy in Subjects with Borderline Resectable or Locally Advanced Unresectable Pancreatic Cancer. Ann. Surg. 2022, 275, 45–53. [Google Scholar] [CrossRef]
  100. Russell, S.J.; Peng, K.-W.; Bell, J.C. Oncolytic virotherapy. Nat. Biotechnol. 2012, 30, 658–670. [Google Scholar] [CrossRef] [Green Version]
  101. Vähä-Koskela, M.J.; Heikkilä, J.E.; Hinkkanen, A.E. Oncolytic viruses in cancer therapy. Cancer Lett. 2007, 254, 178–216. [Google Scholar] [CrossRef]
  102. Rahal, A.; Musher, B.L. Oncolytic viral therapy for pancreatic cancer. J. Surg. Oncol. 2017, 116, 94–103. [Google Scholar] [CrossRef] [PubMed]
  103. Haller, S.D.; Monaco, M.L.; Essani, K. The Present Status of Immuno-Oncolytic Viruses in the Treatment of Pancreatic Cancer. Viruses 2020, 12, 1318. [Google Scholar] [CrossRef]
  104. Ahn, D.H.; Bekaii-Saab, T.S. The Continued Promise and Many Disappointments of Oncolytic Virotherapy in Gastrointestinal Malignancies. Biomedicines 2017, 5, 10. [Google Scholar] [CrossRef] [PubMed]
  105. Mahalingam, D.; Wilkinson, G.A.; Eng, K.H.; Fields, P.; Raber, P.; Moseley, J.L.; Cheetham, K.; Coffey, M.; Nuovo, G.; Kalinski, P.; et al. Pembrolizumab in Combination with the Oncolytic Virus Pelareorep and Chemotherapy in Patients with Advanced Pancreatic Adenocarcinoma: A Phase Ib Study. Clin. Cancer Res. 2019, 26, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Soucek, L.; Buggy, J.J.; Kortlever, R.; Adimoolam, S.; Monclús, H.A.; Allende, M.T.S.; Swigart, L.B.; Evan, G.I. Modeling Pharmacological Inhibition of Mast Cell Degranulation as a Therapy for Insulinoma. Neoplasia 2011, 13, 1093–1100. [Google Scholar] [CrossRef] [Green Version]
  107. Overman, M.; Javle, M.; Davis, R.E.; Vats, P.; Kumar-Sinha, C.; Xiao, L.; Mettu, N.B.; Parra, E.R.; Benson, A.B.; Lopez, C.D.; et al. Randomized Phase II Study of the Bruton Tyrosine Kinase Inhibitor Acalabrutinib, Alone or with Pembrolizumab in Patients with Advanced Pancreatic Cancer. J. Immunother. Cancer 2020, 8, e000587. [Google Scholar] [CrossRef] [Green Version]
  108. Di Federico, A.; Tateo, V.; Parisi, C.; Formica, F.; Carloni, R.; Frega, G.; Rizzo, A.; Ricci, D.; Di Marco, M.; Palloni, A.; et al. Hacking Pancreatic Cancer: Present and Future of Personalized Medicine. Pharmaceuticals 2021, 14, 677. [Google Scholar] [CrossRef]
  109. Singhi, A.D.; George, B.; Greenbowe, J.R.; Chung, J.; Suh, J.; Maitra, A.; Klempner, S.J.; Hendifar, A.; Milind, J.M.; Golan, T.; 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.e4. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Representation of immune cross talk in the complex pancreatic tumor microenvironment. CAFs: cancer-associated fibroblasts; TANs: tumor-associated neutrophils; TAMs: tumor-associated macrophages; NK: natural killer; MDSC: myeloid-derived suppressor cells; PD-1: programmed death-1; PD-L1: programmed death-ligand 1; GM-CSF: granulocyte-macrophage colony-stimulating factor. Created with BioRender (www.biorender.com (accessed on 3 May 2022)).
Figure 1. Representation of immune cross talk in the complex pancreatic tumor microenvironment. CAFs: cancer-associated fibroblasts; TANs: tumor-associated neutrophils; TAMs: tumor-associated macrophages; NK: natural killer; MDSC: myeloid-derived suppressor cells; PD-1: programmed death-1; PD-L1: programmed death-ligand 1; GM-CSF: granulocyte-macrophage colony-stimulating factor. Created with BioRender (www.biorender.com (accessed on 3 May 2022)).
Cancers 14 02429 g001
Table 1. Ongoing phase II–III clinical trials evaluating the use of immune-checkpoint inhibitors selectively in patients with pancreatic cancer.
Table 1. Ongoing phase II–III clinical trials evaluating the use of immune-checkpoint inhibitors selectively in patients with pancreatic cancer.
NCT
(Acronym)
PhaseNumber of PatientsSettingTreatment ArmsPrimary EndpointStatus
NCT03989310I/II50Locally advanced/Metastatic(1) Manganese chloride + Nab-paclitaxel + Gemcitabine + Anti-PD-1 antibody
(2) Nab-paclitaxel + Gemcitabine + Anti-PD-1 antibody
Safety
DCR
Recruiting
NCT04548752II88Maintenance, BRCA mutated(1) Olaparib + Pembrolizumab
29 Olaparib
PFSRecruiting
NCT04156087II20Locally AdvancedMIS-MWA + Durvalumab + TremelimumabPFSRecruiting
NCT05116917II30MetastaticNivolumab + Ipilimumab + Influenza vaccine + SBRTORRRecruiting
NCT03161379II30Borderline resectable, neoadjuvantCyclophosphamide + Nivolumab + GVAX + SBRTCD8 count (cells/mm3) in the tumor microenvironmentActive, not recruiting
NCT04324307I/II60Locally Advanced/
Metastatic
(1) 2nd line PD-L1/CTLA4 inhibitor
(2) 1st line PD-L1/CTLA4 inhibitor + gemcitabine/nab-paclitaxel
(3) 1st line PD-L1/CTLA4 inhibitor + FOLFIRINOX
ORRRecruiting
NCT03193190Ib/II290MetastaticSeverals, combinations of Nab-Paclitaxel, Gemcitabine, Oxaliplatin, Fluorouracil, Atezolizumab, Cobimetinib, PEGPH20, BL-8040, Selicrelumab, Bevacizumab, RO6874281, AB928, Tiragolumab and Tocilizumab.ORR
Safety
Active, not recruiting
NCT04361162II30MetastaticNivolumab + Ipilimumab + RadiationORRRecruiting
NCT04543071II10MetastaticMotixafortide, Cemiplimab, Gemcitabine, Nab-PaclitaxelORRRecruiting
NCT03336216II179Advanced, pretreated(1) Gemcitabine/Nab-Paclitaxel or 5-FU/Leucovorin/Irinotecan Liposome
(2) Cabiralizumab + Nivolumab
(3) Gemcitabine + Nab-Paclitaxel + Cabiralizumab + Nivolumab
(4) Cabiralizumab + Nivolumab + FOLFOX
PFSActive, not recruiting
NCT03977272III110Metastatic(1) modified-FOLFIRINOX/FOLFIRINOX
(2) modified-FOLFIRINOX/FOLFIRINOX + anti PD-1 antibody 200 mg
OSRecruiting
NCT03983057III830Locally advanced/borderline resectable(1) modified-FOLFIRINOX
(2) modified-FOLFIRINOX + anti PD-1 antibody 3 mg/kg
PFSRecruiting
NCT04377048II38MetastaticTegafur-Gimeracil-Oteracil + Nivolumab + GemcitabineORRNot yet recruiting
NCT04493060II20Metastatic, germline or somatic BRCA1/2 and PALB2 related cancerDostarlimab + NiraparibDCRRecruiting
NCT02648282II58Locally advancedCyclophosphamide + Pembrolizumab + GVAX + SBRTDMFSActive, not recruiting
NCT04887805II28Maintenance after 1st or 2nd line chemotherapyLenvatinib + PembrolizumabPFSRecruiting
NCT04247165I/II20Locally advancedNivolumab + Ipilimumab + Gemcitabine + Nab-paclitaxel + SBRTSafetyRecruiting
NCT05093231II20MetastaticPembrolizumab + OlaparibORRRecruiting
NCT04940286II30Resectable/borderline resectable, neoadjuvantDurvalumab + Oleclumab + Nab-paclitaxel + GemcitabineMajor pathological response rate (≤5% viable tumor cells)
Safety
Recruiting
NCT02305186I/II68Resectable/borderline resectable, neoadjuvant(1) chemoradioterapy (with Capecitabine)
(2) chemoradioterapy (with Capecitabine) + Pembrolizumab
TILs per HPF
Safety
Recruiting
NCT04827953I/II24AdvancedGemcitabine + nab-paclitaxel + NLM-001 + ZalifrelimabORRRecruiting
NCT03563248II160Resectable/borderline resectable/locally advanced, neoadjuvant(1) FOLFIRINOX → SBRT → Surgery
(2) FOLFIRINOX + Losartan → SBRT + Losartan → Surgery
(3) FOLFIRINOX + Losartan → SBRT + Nivolumab + Losartan → Surgery
(4) FOLFIRINOX × 8 → SBRT + Nivolumab → Surgery
Proportion of patients with R0 resectionRecruiting
NCT04177810II21MetastaticPlerixafor + CemiplimabORRRecruiting
NCT05014776II20Metastatic, pretreatedTadalafil + Pembrolizumab + Ipilimumab + CRS-207irORRRecruiting
NCT03190265II63Metastatic, pretreated(1) Cyclophosphamide + Nivolumab + Ipilimumab + GVAX + CRS-207
(2) Nivolumab + Ipilimumab + CRS-207
ORRActive, not recruiting
NCT02907099IIb18MetastaticCXCR4 antagonist BL-8040 + PembrolizumabORRActive, not recruiting
NCT04116073II25Unresectable/Metastatic, pretreatedINCMGA00012 (PD-1 antibody)DCR4Recruiting
NCT04753879II38MetastaticNab-paclitaxel + Gemcitabine + Cisplatin + Irinotecan + Capecitabine →
Maintenance with Pembrolizumab + Olaparib
PFSRecruiting
NCT03767582I/II30Locally advanced(1) SBRT + Nivolumab + CCR2/CCR5 dual antagonist
(2) SBRT + Nivolumab + GVAX + CCR2/CCR5 dual antagonist
Safety
Immune response
Recruiting
NCT03727880II36Neoadjuvant/Adjuvant, resectable at diagnosis(1) Pembrolizumab + Defactinib
(2) Pembrolizumab
pCRRecruiting
NCT04624217Ib/II54Advanced, pretreatedGemcitabine + Nab-paclitaxel + SHR-1701ORR
RP2D
Active, not recruiting
PD-1: programmed death-1; DCR: disease control rate; PD-L1: programmed death-ligand 1; PFS: progression-free survival; MIS-MWA: Minimally Invasive Surgical Microwave Ablation; SBRT: Stereotactic Body Radiation Therapy; GVAX: granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine; CTLA-4: Cytotoxic T-Lymphocyte Antigen 4; OS: overall survival; DCR: disease control rate; DMFS: distant metastasis-free survival; TILs: tumor-infiltrating Lymphocytes; HPF: high-powered field; irORR: Objective response rate using immune Response Evaluation Criteria for Solid Tumors (iRECIST); CXCR4: C-X-C Motif Chemokine Receptor 4; DCR4: disease control rate at 4 months; CCR2: Chemokine (C-C motif) receptors 2; CCR5: Chemokine (C-C motif) receptors 5; pCR: pathologic complete response; RP2D: recommended phase 2 dose.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Di Federico, A.; Mosca, M.; Pagani, R.; Carloni, R.; Frega, G.; De Giglio, A.; Rizzo, A.; Ricci, D.; Tavolari, S.; Di Marco, M.; et al. Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? A Focus on Tumor Immune Microenvironment, Predictive Biomarkers and Treatment Outcomes. Cancers 2022, 14, 2429. https://doi.org/10.3390/cancers14102429

AMA Style

Di Federico A, Mosca M, Pagani R, Carloni R, Frega G, De Giglio A, Rizzo A, Ricci D, Tavolari S, Di Marco M, et al. Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? A Focus on Tumor Immune Microenvironment, Predictive Biomarkers and Treatment Outcomes. Cancers. 2022; 14(10):2429. https://doi.org/10.3390/cancers14102429

Chicago/Turabian Style

Di Federico, Alessandro, Mirta Mosca, Rachele Pagani, Riccardo Carloni, Giorgio Frega, Andrea De Giglio, Alessandro Rizzo, Dalia Ricci, Simona Tavolari, Mariacristina Di Marco, and et al. 2022. "Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? A Focus on Tumor Immune Microenvironment, Predictive Biomarkers and Treatment Outcomes" Cancers 14, no. 10: 2429. https://doi.org/10.3390/cancers14102429

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