Next Article in Journal
Long-Term Outcomes of Laparoscopic Greater Curvature Plication and Laparoscopic Sleeve Gastrectomy: Critical Appraisal of the Role of Gastric Plication in Bariatric Surgery
Next Article in Special Issue
Bioinformatic Analysis of the BCL-xL/BCL2L1 Interactome in Patients with Pancreatic Cancer
Previous Article in Journal
Evaluation of Pharmacy Inquiries in Physician Order Reviews for Medication Safety: A Cross-Sectional Study
Previous Article in Special Issue
Antidiabetics, Anthelmintics, Statins, and Beta-Blockers as Co-Adjuvant Drugs in Cancer Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Management of Advanced Pancreatic Cancer through Stromal Depletion and Immune Modulation

1
Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
2
Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Medicina 2022, 58(9), 1298; https://doi.org/10.3390/medicina58091298
Submission received: 9 August 2022 / Revised: 10 September 2022 / Accepted: 12 September 2022 / Published: 17 September 2022

Abstract

:
Pancreatic cancer is one of the leading causes of cancer-related deaths worldwide. Unfortunately, therapeutic gains in the treatment of other cancers have not successfully translated to pancreatic cancer treatments. Management of pancreatic cancer is difficult due to the lack of effective therapies and the rapid development of drug resistance. The cytotoxic agent gemcitabine has historically been the first-line treatment, but combinations of other immunomodulating and stroma-depleting drugs are currently undergoing clinical testing. Moreover, the treatment of pancreatic cancer is complicated by its heterogeneity: analysis of genomic alterations and expression patterns has led to the definition of multiple subtypes, but their usefulness in the clinical setting is limited by inter-tumoral and inter-personal variability. In addition, various cell types in the tumor microenvironment exert immunosuppressive effects that worsen prognosis. In this review, we discuss current perceptions of molecular features and the tumor microenvironment in pancreatic cancer, and we summarize emerging drug options that can complement traditional chemotherapies.

1. Introduction

Pancreatic cancer accounts for 6–8% of annual cancer-related mortalities [1]. The mortality and incidence rates are nearly the same due to the heterogeneous nature of pancreatic cancer, which equates to a poor prognosis in most patients [2]. Despite the robust growth in basic research on pancreatic cancer and improvements in surgical techniques, the 5-year survival rate for patients remains relatively low at approximately 8–9 per 100,000, which is lower than the survival rates for most other cancers [3].
The obstacles in managing pancreatic cancer are multidimensional. Pancreatic cancer is a heterogeneous array of pathological and molecular conditions rather than a singular disease, which leads different patients to respond differently to the same treatment [4,5]. The lack of accurate, cost-effective screening strategies makes pancreatic cancer more difficult to diagnose early than breast, colorectal, and lung cancers [6]. Most patients with early-stage, localized pancreatic cancer present no symptoms or only mild, non-specific symptoms such as fatigue, epigastric discomfort, and loss of appetite. Progressive abdominal pain, weight loss, and jaundice are usually the symptoms that finally raise the alarm for patients and healthcare providers, but by this point, the disease has typically reached an advanced stage that is difficult to treat. By the time of diagnosis, only 20–30% of patients are eligible for surgical resection, and up to 50% may already be at an unresectable metastatic stage [7]. Even for the small subset of patients who can undergo surgical resection, the 5-year survival rate remains lower than 20% [8]. Distal metastasis occurs early during pancreatic cancer progression [9], and metastatic pancreatic cancer is associated with a mortality rate of 95% and median overall survival shorter than one year [9].
The traditional first-line treatment for patients with unresectable pancreatic cancer is the classical chemotherapy gemcitabine. However, many patients develop resistance to this drug within weeks of treatment initiation [10]. Whereas multiple forms of chemotherapy or targeted therapies are available for other cancers, treatment options for pancreatic cancer are severely limited.
Successful cancer management is based on a comprehensive molecular and cytological understanding of tumor cells. Technological innovation in biomedical research has profoundly enriched our knowledge about pancreatic tumorigenesis and progression over the last decade [6,11]. However, a series of promising drug targets have been discovered and may fill gaps in the treatment of advanced pancreatic cancer and enable individualized cancer therapy.
This review briefly summarizes the current treatment recommendations for pancreatic cancer, describes critical advances in subtyping pancreatic cancer and understanding the tumor microenvironment, and presents clinical trials focusing on relevant drug targets.

2. History and Current Recommendations of Advanced Pancreatic Cancer Management

Cytotoxic therapy has been the mainstay of advanced cancer management because it extends patient survival time, albeit only for a limited period of time. Gemcitabine is a hydrophilic nucleoside analog that can be transported into cells to disrupt normal DNA synthesis and inhibit the proliferation of tumor cells. Since the early 1990s, gemcitabine has been the standard of care for advanced pancreatic cancer because it showed better outcomes than fluorouracil [12]. Efforts to improve the outcomes of gemcitabine by combining it with cytotoxic agents or targeted drugs have shown some success [13,14,15,16,17,18]. Erlotinib, an inhibitor of the epidermal growth factor receptor widely used to treat unresectable non-small cell lung cancer, can improve the overall survival of patients with advanced pancreatic cancer by 0.33 months, but at the expense of greater adverse effects [19]. Nab-paclitaxel increases gemcitabine concentrations in the blood and significantly improved response rate and median survival in a phase III trial [20,21]. The US Food and Drug Administration approved this combination therapy in 2013 for advanced pancreatic adenocarcinoma. In 2017, gemcitabine and capecitabine proved to be an effective combination of chemotherapy among pancreatic cancer patients who have undergone pancreatic tumor resection [22]. The combination of leucovorin, fluorouracil, irinotecan, and oxaliplatin, a regimen known as “FOLFIRINOX”, prolonged median overall survival from 6.8 months to 11.1 months [23]. As a result, several countries incorporated FOLFIRINOX into their guidelines for pancreatic cancer management, yet its severe toxicity makes it suitable only for certain patients.
Current pancreatic cancer treatment takes a multi-pronged approach [24]. Pancreatic cancer can be divided into four basic categories based on anatomical resectability: resectable, borderline resectable, unresectable (locally advanced), and metastatic. The classification largely depends on high-resolution computed tomography and surgeon experience [25]. Neoadjuvant chemotherapies can improve the resectability of borderline resectable pancreatic cancer [26]. For unresectable pancreatic cancer, the recommended first-line treatments are combinations of gemcitabine with FOLFIRINOX, nab-paclitaxel, or erlotinib. If gemcitabine-based treatment fails upon cancer metastasis, recommended second-line therapies include: the combination of fluorouracil, folinic acid, and oxaliplatin (OFF); nanoliposomal irinotecan, with or without fluorouracil and folinic acid; and the combination of folinic acid, oxaliplatin, and fluorouracil (FOLFOX) [27,28,29].

3. Tumorigenesis and Tumor Microenvironment in Pancreatic Cancer

3.1. Hallmarks of Pancreatic Cancer

Cancer cells share typical hallmarks that distinguish them from normal cells, and cancers of different origins demonstrate unique characteristics that are potentially targetable [30]. Pancreatic cancer cells carry specific genomic alterations critical to tumorigenesis and progression [31]. Classically, pancreatic cancer tumorigenesis has been considered the result of a sequential accumulation of molecular perturbations in resident pancreatic acinous cells that lead to intraepithelial neoplasia and, ultimately, cancer [32,33]. These disturbances include: mutations in KRAS, TP53, CDK4/6, and BRCA1/2; deletion of BRAF; and gene rearrangements of NTRK, ALK, and NRG1 [32,34]. However, clinical efforts to target various genomic alterations for treatment have so far proved to be disappointing [35].
Metabolic reprogramming is another remarkable feature of pancreatic cancer cells [30]. The hypoxic intra-tumoral environment leads tumor cells to shift their metabolism from oxidative phosphorylation toward oxygen-independent glycolysis, known as the “Warburg effect” [36,37], to help tumor cells survive the scarcity of nutrients and oxygen in their microenvironment. As part of this metabolic shift, redundant glycolytic intermediates enter various metabolic bypass pathways [38,39], and glutamine transportation is hyperactivated [40]. Autophagy and macropinocytosis are hyperactivated to scavenge nutrients [41,42,43,44]. Novel therapeutic opportunities exist in inhibiting cancer cell mitochondrial bioenergetics or scavenging pathways, as reviewed previously [45].

3.2. The Inter-Tumoral Microenvironment of Pancreatic Cancer

In the pancreatic tumor microenvironment, the extracellular matrix (ECM), immune and stromal cells, and secretory signals interact to promote cancer cell invasion, dissemination, and distal colonization [42,46]. Cancer-associated fibroblasts (CAFs) in the tumor microenvironment create a desmoplastic extracellular matrix that helps drive pancreatic cancer cell progression and drug resistance by secreting various fibrotic substances, such as collagen and hyaluronic acid [47]. CAFs also secrete various amino acids and lipids that support cancer cell survival [48,49,50]. There are at least two types of CAFs within the tumor microenvironment. Myofibroblasts are characterized by α-smooth muscle actin (α-SMA), and inflammatory CAFs lose myofibroblastic features and secrete various inflammatory cytokines [51,52].
Immune cells in the PTME play a critical role during cancer progression. Compared to other types of cancer, most pancreatic cancers, if not all, are immunologically cold, which is represented by the limited infiltration of effector T cells and low expression of PD-L1, thereby eliciting low responses to immune checkpoint inhibitor therapy [53]. The restrained proliferation and activation of cytotoxic T cells are closely related to the composition of immune mi, mainly comprising macrophages, myeloid-derived suppressor cells (MDSC), fibroblasts, neutrophils, and T regulatory cells [54,55].
Tumor-associated macrophages (TAMs) are key components of cancer stroma inflammation, presenting a spectrum of functional states with two opposing extremes [42]. While polarized M1 macrophages are pro-inflammatory and anti-neoplastic, M2 macrophages have the opposed phenotype, which promotes tumor growth and predominates during cancer development [42]. Multiple lines of evidence have shown that macrophage depletion efficiently impairs angiogenesis and reduces metastasis formation [56]. Inhibition of TAMs also alleviates gemcitabine resistance by inactivating CDA, an enzyme critically involved in the inactivation of gemcitabine [57].
Myeloid-derived suppressor cells (MDSCs) are strongly increased in both the circulation and the TME of PDAC patients, and a positive correlation exists between the cell count and tumor staging [58]. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-1β secreted by tumor cells are critical to the development of MDSCs [59]. MDSCs increase the expression of tumor PD-L1 expression via the activation of EGFR/MAPK signaling pathway and result in CD8+ T cell exhaustion and T regulatory cell expansion [58,60], thus abrogation of tumor-derived GM-CSF secretion prohibits the MDSCs infiltration and blocks tumor development by increasing the number of cytotoxic T cells [61].
The role of neutrophils educated by cancer cells, or tumor-associated neutrophils (TAN), has received less attention in the PTME. Evidence showed that TANs contribute to pancreatic cancer cell spreading by producing multiple enzymes, namely matrix metalloproteinase (MMP) 8 and MMP9, Cathepsin-B, and proteinase-3, which accelerate the degradation of the ECM, break down the barrier, and promote cancer invasion and metastasis [62]. Neutrophil extracellular traps (NETs) are responsible for the exclusion of cytotoxic CD8+ T cells from tumors and may have a role in driving pancreatic cancer metastasis [63,64]. In preclinical models, lorlatinib, a TANs suppressor, attenuates pancreatic cancer growth and improves the efficacy of immune checkpoint inhibitors [65]. Similarly, the inhibition of neutrophil activator IL17 increased immune checkpoint blockade (PD-1, CTLA4) sensitivity [63].
The ECM in the pancreatic tumor microenvironment also appears to contribute to immunosuppression [66]: the thick, fibrotic ECM inhibits tumor vascularization and immune cell infiltration, as well as the delivery of drugs into tumors [67]. Thus, a suggested strategy is to bring back the normalization in the tumor immune ecosystem and make the immune backbone of the tumor hot or immunogenic so that it would be more responsive to therapy [53,68].
Although some studies have shown that an immunologic subtype of pancreatic cancer does exist and generally has a better prognosis than the unselected patient population [69,70], the treatment of immunosuppressive PDAC remains a major obstacle. The targeting of various contributors of the immune microenvironment in combination with immune checkpoint inhibitors is promising for synergistic antitumoral effects in preclinical models of pancreatic cancer and some ongoing clinical trials.

4. Molecular and Cellular Subtyping of Pancreatic Cancer

Morphologically and pathologically indistinguishable pancreatic tumors can have quite different genomic features and respond differently to antitumor therapy. Thus, a molecular classification system is required to guide clinical management adequately [71].
Molecular subtyping is a necessary stepping stone toward individualized anticancer therapy. Early attempts to classify pancreatic cancers relied on single genetic markers, including point mutations, structural variations, and protein markers [34,72,73,74,75,76,77]. Though most classification schemes have failed to enter the clinic, some markers, such as KRAS, TP53, and ERBB2, have been validated as prognostic markers and surgical indications [78]. In 2011, microarray data were used to define three subtypes of pancreatic ductal adenocarcinoma (PDAC): “classical”, “quasimesenchymal”, and “exocrine-like”. Each subtype shows distinguishable histopathological features that may indicate prognosis and responsiveness to gemcitabine and erlotinib [79]. More recently, an “immunogenic” subtype has been described, and is characterized by strong immune infiltration of the tumor microenvironment [33,79].
Pancreatic cancer has been subtyped as “classical” or “basal-like” based on its cellular composition [80,81]. The classical subtype arises from an endodermal-like stromal lineage and shows activation of the transcription factor GATA6 and KRAS expression. In contrast, basal-like cells are associated with altered chromatin modification and a worse prognosis. Classical PDAC appears to respond better to first-line pancreatic cancer chemotherapy than basal-like PDAC [82]. More recent work has identified additional cell-based subtypes, including “desmoplastic”, “immune classical”, and “stroma-activated” [71].
Multi-omics approaches may be key to elucidating on the complex nature of pancreatic cancer. Indeed, integrating genomic and transcriptomic information revealed that the classical and basal-like subtypes still contain substantial heterogeneity, which should be studied further [83]. For example, the basal-like subtype may contain a “squamous” subpopulation associated with altered chromatin modification and faster disease progression [84,85]. Comprehensive genomic and transcriptomic profiling may help pave the way for personalized treatments against pancreatic cancer.

5. TME Normalization for the Management of Advanced Pancreatic Cancer

The highly desmoplastic tumor stroma and presence of desmoplasia-inducing stromal cells are major contributors to the lack of efficacy of, and resistance to, various anticancer drugs [86]. Hedgehog (Hh) signaling is abnormally activated in pancreatic intraepithelial neoplasia and PDAC, and it is critical for pancreatic cancer stroma formation and stabilization [87,88]. Blocking Hh in mouse models increased the tumor vascularization and intra-tumoral concentration of gemcitabine [89]. However, adding a Hh inhibitor to either gemcitabine alone or FOLFIRINOX failed to improve outcomes for advanced pancreatic cancer patients [90,91,92]. This result was understandable because the Hh pathway involves many other biological and pathological processes. An ongoing clinical trial investigates the safety and efficacy of inhibiting focal adhesion kinase (Table 1), a crucial regulator of the fibrotic and immunosuppressive pancreatic tumor microenvironment [93]. However, inhibiting a specific signal transduction pathway on its own may prove ineffective: for example, inhibiting focal adhesion kinase appears to promote drug resistance [94]. Drug targets with more specific antitumor effects are warranted.
The cell–cell communication between the tumor cell and the non-tumor stromal constituents promotes the TME to shift towards a ‘hungry’ state that largely promotes tumor growth, invasion, and metastasis. Various stromal cells and immune cells present with cancer-associated phenotypes [42], and induced neovascularization is a shared hallmark of the TME of almost all solid tumors in response to the increased need for oxygen and nutrients [95,96]. These alterations form the fertilized “soil” that facilitates tumor growth. Rather than directly developing novel therapies that present a higher potency to kill tumor cells, “stroma normalization strategies” aim at reversing the TME state to a relatively normal one from different angles by increasing the function of vessels or modulating the phenotype of various populations of stromal cells in order to enhance the efficacy of well-established chemo-, radio-, and immunotherapies. CAF reprogramming, vascular reconstruction, and immune modulation are three promising ways that TME normalization strategies can manage solid tumors, which will be elaborated on in the following sections.

5.1. CAF Reprogramming and ECM Depletion

Targeting CAFs may be another way to control tumor growth [50], such as by depleting them, reprogramming them to adopt an anti-tumorigenic or quiescent state, or inhibiting their communication with the tumor stroma [97,98,99,100]. Inhibition of molecular markers of CAFs, such as α-SMA and fibroblast activation protein (FAP), can disrupt desmoplasia in the pancreatic tumor microenvironment and block tumor growth [101]. However, targeting CAFs or the fibrotic tumor microenvironment is a difficult issue. For unknown reasons, disruption of the tumor microenvironment’s desmoplastic nature can lead to anticancer treatment failure [94]. Deleting α-SMA-expressing myofibroblasts in pre-cancerous or PDAC tumors in mice exacerbated hypoxia and suppressed immune cell infiltration, which reduced survival [102]. The multiple roles of CAFs make it difficult to predict the effects of disrupting them [4]; an alternative approach may be to target specific signals secreted by CAFs. For example, pharmacologic depletion of CXCR4 and immunotherapy inhibited cancer progression in a mouse model of PDAC [103]. A phase II trial suggested that CXCR4 inhibition combined with pembrolizumab can reduce populations of immunosuppressive cells in the tumor microenvironment of patients with chemotherapy-resistant advanced pancreatic cancer [104].
The therapeutic rationale for disrupting constituents of the ECM is to break down the mechanical barrier forged by dense desmoplasia. During cell differentiation, fibroblasts secrete major components of the ECM Hyaluronan, collagen, and fibronectin as physical barriers to impede the transport and diffusion of antitumor drugs [67]. Therefore, targeting these fibrosis-related components should increase such drugs’ local concentration and efficacy. Hyaluronidase degrades hyaluronan in the ECM and decreases interstitial fluid pressure [105]. Recombinant hyaluronidase has been tested as a complementary therapy to enhance combination chemotherapy [106,107], but preclinical and clinical studies have given conflicting results. For example, the hyaluronidase PEGPH20 improved the benefits of chemotherapy in a phase II trial, yet it increased rates of drug-related adverse effects in a subsequent phase III trial [108,109,110,111]. Therefore, targeting components in the ECM may be insufficient to overcome drug resistance. Efforts are underway to explore the simultaneous pharmaceutical inhibition of multiple distinct pathways or biological processes, particularly immunomodulatory pathways (Figure 1).

5.2. Anti-Angiogenic Therapy and Vascular Normalization

Although angiogenesis is extensively induced in solid tumors, the mismatch between oxygen and nutrient supply and cancer cell division largely creates a hypoxic intra-tumoral environment [95]. Hyperpermeability and branch tortuosity of the newly formed vessels, as well as the increased physical compression by uncontrolled expansion of tumor cell mass, further compromise the blood perfusion of the tumor, which limits the access of various immune cells [53]. Therefore, strategies that alter the intra-tumor vasculature have long been considered for their antitumor potential. The earliest therapy targeting tumor vasculature was the anti-vascular endothelial growth factor (anti-VEGF) agents, such as bevacizumab, which has been shown to improve the efficacy of traditional chemotherapy or immunotherapy for some solid tumors [112,113]. Nevertheless, this success cannot be replicated in pancreatic cancer because multiple phase III trials with anti-VEGF agents have failed to increase the overall survival of advanced pancreatic cancer patients compared to chemotherapy alone [114]. In several phase III trials, other regimens of the same category, such as sorafenib, axitinib, and ZIV-aflibercept in combination with gemcitabine, also failed to improve the outcome [115]. Though the mechanism remains incompletely understood, the simple hypovascularized nature of pancreatic ductal adenocarcinoma is not enough to account for the failure [69]. Using a PDAC mouse model, Aguilera et al. found that chronic treatment with bevacizumab-induced intra-tumoral hypoxia accelerated collagen deposition and increased the overall tumor burden [116].
Instead of a simple blockade of vasculature genesis, tumor vasculature normalization is the concept of eliminating excess endothelial cells, therefore pruning immature and unproductive vasculature to provide easier access for the anticancer drug delivery to cancer cells. Furthermore, the normalization can increase pericyte coverage, fortify the immature vessels, and improve tumor perfusion. In preclinical animal models, Semaphorin 3A (SEMA3A) was secreted by endothelial cells and acted as a vasculature-normalizing regulator by negatively impacting integrin expression [117]. Expression of SEMA3A gradually decreases during the transition of premalignant lesions to the tumor, consistent with the development of a dysfunctional abnormal tumor vasculature system during this course [117]. Restored expression of SEMA3A resulted in the normalization of the tumor vasculature, impairment of metastatic disease progression, and sensitization of immunotherapy [118,119]. Moving forward, anti-angiogenic therapy in PDAC should consider proper experimental models that recapitulate the angiogenic feature of the real tumor and possibly classify the pancreatic cancers in terms of vasculature extent to improve the benefit of therapy.

5.3. Immune Checkpoint Blockade (ICB)

Adoptive cell transfer and immune checkpoint blockade (ICB) are two powerful treatment paradigms that have achieved groundbreaking success in multiple cancers, such as melanoma and leukemia [42]. However, both have been largely unsuccessful against pancreatic cancer [42]. A single classical ICB has an average response rate of only 5% in treating advanced pancreatic cancer [120]. Nevertheless, preclinical and clinical trials continue in PDAC animal models and patients to target tumor antigens or components of the tumor microenvironment [121,122,123]. Routine evaluation of tumor mutational burden has been recommended to identify patients that may potentially benefit from ICB [124]. For example, CD8 and T cell receptor clonality expression have been correlated with responses to ICB targeting PD-1/PD-L1 [125]. Innate lymphoid cells are involved in activating tissue-specific tumor immunity in PDAC [126]. “Switchable” adoptive cell transfer, which exploits a recombinant peptide that bridges tumor antigen and the Fab domain of the T-cell receptor (TCR) to control CAR-T cell response, can potentially achieve tunable antitumor and avoid off-tumor effects in preclinical PDAC models [127]. This strategy potentially improves the safety of CAR-T therapy in solid tumors while preserving antitumor responses to the largest extent.

5.4. Pancreatic Tumor Vaccine

“Cancer vaccine” strategies aim to increase the efficacy of existing ICBs by using immunostimulatory agents to sensitize the host immune system against tumors. One potential vaccine, GVAX pancreas, has been formulated with allogeneic pancreatic tumor cells that secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) to induce T cell infiltration of the pancreatic tumor. In contrast, the vaccine CRS-207 contains the live attenuated pathogen Listeria monocytogenes, which stimulate an immune response against mesothelin, a tumor-associated antigen overproduced by the pancreatic tumor [128,129]. One trial failed to demonstrate the clinical benefit of the combination of GVAX pancreas, cyclophosphamide, and CRS-207 against advanced pancreatic cancer [130,131]. The whole-cell immunomodulator HyperAcute-Pancreas algenpantucel-L (HAPa) comprises allogenic pancreatic cancer cells that elicit antibody-dependent cell-mediated cytotoxicity [132]. However, it failed to improve the survival of patients with borderline resectable or locally advanced, unresectable PDAC who received chemotherapy alone or with radiation in a phase III trial [132]. Conversely, adding a tumor vaccine targeting VEGFR1/2 or personalized neoantigen peptides improved the efficacy of gemcitabine [133,134]. Although vaccines or immune-enhancing approaches on their own may be ineffective, their combinations should be explored further.

5.5. Cytokine-Based Therapy

Another immune-modulating strategy is the regulation of immunosuppressive cells in the pancreatic tumor microenvironment, such as regulatory T cells, cancer-associated macrophages, and myeloid-derived suppressor cells [58,135,136,137,138]. Activation of classical tumor necrosis factor receptor family member CD40 enhances antigen presentation by dendritic cells and activates cytotoxic T cells in animal models [139]. CD40 agonists can also polarize macrophages in the tumor microenvironment to adopt a pro-inflammatory state that can kill tumors in preclinical studies [140,141]. Combining CD40 priming with other immune modulation may enhance antitumor immune responses [142] and potentiate ICB and chemotherapy [143,144]. However, adding the monoclonal antibody sotigalimab or nivolumab, which acts as a CD40 agonist, to gemcitabine and nab-paclitaxel failed to provide satisfactory efficacy in a phase II trial [145,146]. This may reflect that gemcitabine antagonizes the ability of CD40- and ICB-based therapy to restrict tumor progression in animal models [147].
Cytokines and chemokines regulate the differentiation and maturation of various immune cells. In mouse models, inhibiting CCR2 and CSF-1R can antagonize the ability of tumor-associated macrophages to suppress T cell responses [148,149]. However, whether combining CCR2 inhibitors with nab-paclitaxel plus gemcitabine is effective in patients remains unclear [150]. In fact, the combination of the CSF-1R inhibitor cabiralizumab, the PD-1 inhibitor nivolumab, and gemcitabine failed to improve the progression-free survival of advanced pancreatic cancer patients compared to gemcitabine alone. This may reflect that CAFs induce tumor infiltration by CXCR2-expressing MDSCs [151], which argues for multi-pronged therapy. IL-10 activates cytotoxic T cells and stimulates their proliferation [105], yet recombinant human IL-10 has not been proven effective against advanced chemotherapy-resistant PDAC [152].

5.6. STING DNA-Sensing Pathway Agonists

Stimulator of Interferon Genes (STING) is a transmembrane endoplasmic reticulum protein normally activated by cytosolic DNA, which binds to cGAMP and activates transcriptional gene cascades and ultimately results in type I interferon (IFN) production. It is a key regulator for generating cytotoxic T cells [153]. Activation of the STING pathway in antigen-presenting cells (APCs) is essential for checkpoint blockade and anti-PD1 therapies [154]. In preclinical models, systemic or intra-tumoral administration of STING agonists is powerful in reversing immune suppression of the tumor and improving cytotoxic T-cell infiltration [154,155,156]. STING-dependent vaccines can inhibit tumor growth and improve long-term antitumor memory [157]. As one of the earliest investigated STING agonists, 5,6-dimethylxanthenone-4-acetic acid (DMXAA) can modulate the immune system and result in anticancer responses in mice [154]. Similarly, cytosolic cyclic dinucleotides (CDNs) enhance type I interferons’ production by activating TBK1/IRF3 and NF-κB pathways [158]. More evidence is emerging that STING activation is an effective antitumor strategy that can be applied in various forms.

6. Challenges and Future Perspectives

Pancreatic cancer is one of the deadliest malignancies worldwide due to limited therapeutic options. Treating and managing it effectively will require a multidisciplinary effort, including improvements in basic research, disease screening and diagnosis, drug development, surgical techniques, and, most importantly, evidence-based clinical decision-making. Progress in elucidating intra- and inter-tumoral heterogeneity has identified several promising therapeutic strategies, such as stromal depletion, immune modulation, and pathway targeting.
Effective therapies will most likely depend on combining two or more of these therapies at once. For example, treatments targeting only stromal depletion or immune modulation have generally proven disappointing in clinical trials. In part, this reflects the dynamic crosstalk between the stroma and immune cells in the pancreatic microenvironment, such that alterations in one can lead to dramatic changes in the other. Combining stroma-targeting and immune-targeting treatments may be more effective: for example, cancer vaccine-induced T cell infiltration could be enhanced by the hyaluronidase PEGPH20 in murine models [159].
Several pancreatic cancer treatments that showed promise in preclinical research proved ineffective or harmful in patients. This reflects the difficulty in creating preclinical in vitro and in vivo models that emulate the clinical characteristics of pancreatic cancer. For example, the earliest pancreatic cancer studies used 3-methylcholanthrene as a carcinogen to induce pancreatic tumor formation in wild-type C57BL/6 mice, and the widely used Panc02 cell line was derived from these animals [160]. This in vitro pancreatic cancer cell model was flawed due to the absence of hallmark gene mutations that we now know are important in pancreatic tumorigenesis [31]. This limitation was overcome by genetically engineering the KPC mouse model to include the mutations and more closely mimic human cancer progression [161]. Preclinical studies of stroma-depleting therapies have also failed to model the tumor stroma accurately. The stroma exerts complex influences on pancreatic tumors, which in different contexts can suppress tumor growth or promote metastasis [162,163]. Desmoplasia in the tumor microenvironment can vary substantially between patients, between tumors in the same patient, and even within the same tumor [4,80,164]. The same stromal depletion approach may lead to opposite outcomes in different contexts, highlighting the need for individualized assessment of patient suitability for stroma-depleting therapies.

7. Conclusions

In conclusion, creating therapeutic possibilities for patients with advanced pancreatic cancer will require deepening our biological and molecular understanding of the disease while improving the preclinical development and clinical translation of promising treatments.

Author Contributions

Conceptualization, S.C. and Z.W.; methodology, T.L.; software, T.L.; resources, Q.X.; writing—original draft preparation, T.L.; writing—review and editing, S.C.; supervision, Z.W.; project administration, Q.X.; funding acquisition, Z.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Beijing Municipal Key Clinical Specialty Excellence Program, provided by Beijing Municipal Health Commission and Beijing Municipal Bureau of Finance.

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Acknowledgments

The authors gratefully acknowledge A. Chapin Rodriguez for his help in language editing of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Najafi, M.; Majidpoor, J.; Toolee, H.; Mortezaee, K. The current knowledge concerning solid cancer and therapy. J. Biochem. Mol. Toxicol. 2021, 35, e22900. [Google Scholar] [CrossRef] [PubMed]
  4. Neuzillet, C.; Tijeras-Raballand, A.; Ragulan, C.; Cros, J.; Patil, Y.; Martinet, M.; Erkan, M.; Kleeff, J.; Wilson, J.; Apte, M.; et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J. Pathol. 2019, 248, 51–65. [Google Scholar] [CrossRef]
  5. Bernard, V.; Semaan, A.; Huang, J.; San Lucas, F.A.; Mulu, F.C.; Stephens, B.M.; Guerrero, P.A.; Huang, Y.; Zhao, J.; Kamyabi, N.; et al. Single-Cell Transcriptomics of Pancreatic Cancer Precursors Demonstrates Epithelial and Microenvironmental Heterogeneity as an Early Event in Neoplastic Progression. Clin. Cancer Res. 2019, 25, 2194–2205. [Google Scholar] [CrossRef]
  6. Singhi, A.D.; Wood, L.D. Early detection of pancreatic cancer using DNA-based molecular approaches. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 457–468. [Google Scholar] [CrossRef]
  7. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef]
  8. van Dam, J.L.; Janssen, Q.P.; Besselink, M.G.; Homs, M.Y.V.; van Santvoort, H.C.; van Tienhoven, G.; de Wilde, R.F.; Wilmink, J.W.; van Eijck, C.H.J.; Groot Koerkamp, B. Dutch Pancreatic Cancer Group Neoadjuvant therapy or upfront surgery for resectable and borderline resectable pancreatic cancer: A meta-analysis of randomised controlled trials. Eur. J. Cancer 2022, 160, 140–149. [Google Scholar] [CrossRef]
  9. Strobel, O.; Neoptolemos, J.; Jäger, D.; Büchler, M.W. Optimizing the outcomes of pancreatic cancer surgery. Nat. Rev. Clin. Oncol. 2019, 16, 11–26. [Google Scholar] [CrossRef]
  10. Yu, S.; Zhang, C.; Xie, K.-P. Therapeutic resistance of pancreatic cancer: Roadmap to its reversal. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188461. [Google Scholar] [CrossRef]
  11. Thomasian, N.M.; Kamel, I.R.; Bai, H.X. Machine intelligence in non-invasive endocrine cancer diagnostics. Nat. Rev. Endocrinol. 2022, 18, 81–95. [Google Scholar] [CrossRef] [PubMed]
  12. Burris, H.A.; Moore, M.J.; Andersen, J.; Green, M.R.; Rothenberg, M.L.; Modiano, M.R.; Cripps, M.C.; Portenoy, R.K.; Storniolo, A.M.; Tarassoff, P.; et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J. Clin. Oncol. 1997, 15, 2403–2413. [Google Scholar] [CrossRef] [PubMed]
  13. Stathopoulos, G.P.; Syrigos, K.; Aravantinos, G.; Polyzos, A.; Papakotoulas, P.; Fountzilas, G.; Potamianou, A.; Ziras, N.; Boukovinas, J.; Varthalitis, J.; et al. A multicenter phase III trial comparing irinotecan-gemcitabine (IG) with gemcitabine (G) monotherapy as first-line treatment in patients with locally advanced or metastatic pancreatic cancer. Br. J. Cancer 2006, 95, 587–592. [Google Scholar] [CrossRef] [PubMed]
  14. Chauffert, B.; Mornex, F.; Bonnetain, F.; Rougier, P.; Mariette, C.; Bouché, O.; Bosset, J.F.; Aparicio, T.; Mineur, L.; Azzedine, A.; et al. Phase III trial comparing intensive induction chemoradiotherapy (60 Gy, infusional 5-FU and intermittent cisplatin) followed by maintenance gemcitabine with gemcitabine alone for locally advanced unresectable pancreatic cancer. Definitive results of the 2000-01 FFCD/SFRO study. Ann. Oncol. 2008, 19, 1592–1599. [Google Scholar] [CrossRef]
  15. Colucci, G.; Labianca, R.; Di Costanzo, F.; Gebbia, V.; Cartenì, G.; Massidda, B.; Dapretto, E.; Manzione, L.; Piazza, E.; Sannicolò, M.; et al. Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC) Randomized phase III trial of gemcitabine plus cisplatin compared with single-agent gemcitabine as first-line treatment of patients with advanced pancreatic cancer: The GIP-1 study. J. Clin. Oncol. 2010, 28, 1645–1651. [Google Scholar] [CrossRef]
  16. Rocha Lima, C.M.; Green, M.R.; Rotche, R.; Miller, W.H.; Jeffrey, G.M.; Cisar, L.A.; Morganti, A.; Orlando, N.; Gruia, G.; Miller, L.L. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J. Clin. Oncol. 2004, 22, 3776–3783. [Google Scholar] [CrossRef]
  17. Oettle, H.; Richards, D.; Ramanathan, R.K.; van Laethem, J.L.; Peeters, M.; Fuchs, M.; Zimmermann, A.; John, W.; Von Hoff, D.; Arning, M.; et al. A phase III trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer. Ann. Oncol. 2005, 16, 1639–1645. [Google Scholar] [CrossRef]
  18. Poplin, E.; Feng, Y.; Berlin, J.; Rothenberg, M.L.; Hochster, H.; Mitchell, E.; Alberts, S.; O’Dwyer, P.; Haller, D.; Catalano, P.; et al. Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: A trial of the Eastern Cooperative Oncology Group. J. Clin. Oncol. 2009, 27, 3778–3785. [Google Scholar] [CrossRef]
  19. Moore, M.J.; Goldstein, D.; Hamm, J.; Figer, A.; Hecht, J.R.; Gallinger, S.; Au, H.J.; Murawa, P.; Walde, D.; Wolff, R.A.; et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 2007, 25, 1960–1966. [Google Scholar] [CrossRef]
  20. Von Hoff, D.D.; Ramanathan, R.K.; Borad, M.J.; Laheru, D.A.; Smith, L.S.; Wood, T.E.; Korn, R.L.; Desai, N.; Trieu, V.; Iglesias, J.L.; et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: A phase I/II trial. J. Clin. Oncol. 2011, 29, 4548–4554. [Google Scholar] [CrossRef] [Green Version]
  21. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed]
  22. 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]
  23. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.-L.; Gourgou-Bourgade, S.; de la Fouchardière, C.; et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
  24. Dumont, R.; Puleo, F.; Collignon, J.; Meurisse, N.; Chavez, M.; Seidel, L.; Gast, P.; Polus, M.; Loly, C.; Delvenne, P.; et al. A single center experience in resectable pancreatic ductal adenocarcinoma: The limitations of the surgery-first approach. Critical review of the literature and proposals for practice update. Acta. Gastroenterol. Belg. 2017, 80, 451–461. [Google Scholar] [CrossRef]
  25. Hong, S.B.; Lee, S.S.; Kim, J.H.; Kim, H.J.; Byun, J.H.; Hong, S.M.; Song, K.-B.; Kim, S.C. Pancreatic Cancer CT: Prediction of Resectability according to NCCN Criteria. Radiology 2018, 289, 710–718. [Google Scholar] [CrossRef]
  26. Del Chiaro, M.; Søreide, K. Trials and tribulations of neoadjuvant therapy in pancreatic cancer. Br. J. Surg. 2018, 105, 1387–1389. [Google Scholar] [CrossRef]
  27. Wang-Gillam, A.; Li, C.-P.; Bodoky, G.; Dean, A.; Shan, Y.-S.; Jameson, G.; Macarulla, T.; Lee, K.-H.; Cunningham, D.; Blanc, J.F.; et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): A global, randomised, open-label, phase 3 trial. Lancet 2016, 387, 545–557. [Google Scholar] [CrossRef]
  28. Pelzer, U.; Schwaner, I.; Stieler, J.; Adler, M.; Seraphin, J.; Dörken, B.; Riess, H.; Oettle, H. Best supportive care (BSC) versus oxaliplatin, folinic acid and 5-fluorouracil (OFF) plus BSC in patients for second-line advanced pancreatic cancer: A phase III-study from the German CONKO-study group. Eur. J. Cancer 2011, 47, 1676–1681. [Google Scholar] [CrossRef]
  29. Gill, S.; Ko, Y.-J.; Cripps, C.; Beaudoin, A.; Dhesy-Thind, S.; Zulfiqar, M.; Zalewski, P.; Do, T.; Cano, P.; Lam, W.Y.H.; et al. PANCREOX: A randomized phase III study of fluorouracil/leucovorin with or without oxaliplatin for second-line advanced pancreatic cancer in patients who have received gemcitabine-based chemotherapy. J. Clin. Oncol. 2016, 34, 3914–3920. [Google Scholar] [CrossRef]
  30. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  31. Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 2017, 32, 185–203.e13. [Google Scholar] [CrossRef] [PubMed]
  32. Makohon-Moore, A.; Iacobuzio-Donahue, C.A. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat. Rev. Cancer 2016, 16, 553–565. [Google Scholar] [CrossRef] [PubMed]
  33. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.-M.; Gingras, M.-C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.C.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef] [PubMed]
  34. Humphris, J.L.; Patch, A.-M.; Nones, K.; Bailey, P.J.; Johns, A.L.; McKay, S.; Chang, D.K.; Miller, D.K.; Pajic, M.; Kassahn, K.S.; et al. Hypermutation in pancreatic cancer. Gastroenterology 2017, 152, 68–74.e2. [Google Scholar] [CrossRef]
  35. Nevala-Plagemann, C.; Hidalgo, M.; Garrido-Laguna, I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat. Rev. Clin. Oncol. 2020, 17, 108–123. [Google Scholar] [CrossRef]
  36. Warburg, O.; Wind, F.; Negelein, E. The metabolism of tumors in the body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef]
  37. Denko, N.C. Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat. Rev. Cancer 2008, 8, 705–713. [Google Scholar] [CrossRef]
  38. Encarnación-Rosado, J.; Kimmelman, A.C. Harnessing metabolic dependencies in pancreatic cancers. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 482–492. [Google Scholar] [CrossRef]
  39. Dey, P.; Li, J.; Zhang, J.; Chaurasiya, S.; Strom, A.; Wang, H.; Liao, W.-T.; Cavallaro, F.; Denz, P.; Bernard, V.; et al. Oncogenic KRAS-Driven Metabolic Reprogramming in Pancreatic Cancer Cells Utilizes Cytokines from the Tumor Microenvironment. Cancer Discov. 2020, 10, 608–625. [Google Scholar] [CrossRef]
  40. Zhao, J.-S.; Shi, S.; Qu, H.-Y.; Keckesova, Z.; Cao, Z.-J.; Yang, L.-X.; Yu, X.; Feng, L.; Shi, Z.; Krakowiak, J.; et al. Glutamine synthetase licenses APC/C-mediated mitotic progression to drive cell growth. Nat. Metab. 2022, 4, 239–253. [Google Scholar] [CrossRef]
  41. Perera, R.M.; Stoykova, S.; Nicolay, B.N.; Ross, K.N.; Fitamant, J.; Boukhali, M.; Lengrand, J.; Deshpande, V.; Selig, M.K.; Ferrone, C.R.; et al. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature 2015, 524, 361–365. [Google Scholar] [CrossRef]
  42. Ho, W.J.; Jaffee, E.M.; Zheng, L. The tumour microenvironment in pancreatic cancer—Clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 2020, 17, 527–540. [Google Scholar] [CrossRef] [PubMed]
  43. Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020, 581, 100–105. [Google Scholar] [CrossRef] [PubMed]
  44. Gupta, S.; Yano, J.; Mercier, V.; Htwe, H.H.; Shin, H.R.; Rademaker, G.; Cakir, Z.; Ituarte, T.; Wen, K.W.; Kim, G.E.; et al. Lysosomal retargeting of Myoferlin mitigates membrane stress to enable pancreatic cancer growth. Nat. Cell Biol. 2021, 23, 232–242. [Google Scholar] [CrossRef] [PubMed]
  45. Halbrook, C.J.; Lyssiotis, C.A. Employing metabolism to improve the diagnosis and treatment of pancreatic cancer. Cancer Cell 2017, 31, 5–19. [Google Scholar] [CrossRef]
  46. 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]
  47. Menezes, S.; Okail, M.H.; Jalil, S.M.A.; Kocher, H.M.; Cameron, A.J.M. Cancer-associated fibroblasts in pancreatic cancer: New subtypes, new markers, new targets. J. Pathol. 2022, 257, 526–544. [Google Scholar] [CrossRef]
  48. Sousa, C.M.; Biancur, D.E.; Wang, X.; Halbrook, C.J.; Sherman, M.H.; Zhang, L.; Kremer, D.; Hwang, R.F.; Witkiewicz, A.K.; Ying, H.; et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016, 536, 479–483. [Google Scholar] [CrossRef]
  49. Gabitova-Cornell, L.; Surumbayeva, A.; Peri, S.; Franco-Barraza, J.; Restifo, D.; Weitz, N.; Ogier, C.; Goldman, A.R.; Hartman, T.R.; Francescone, R.; et al. Cholesterol Pathway Inhibition Induces TGF-β Signaling to Promote Basal Differentiation in Pancreatic Cancer. Cancer Cell 2020, 38, 567–583.e11. [Google Scholar] [CrossRef]
  50. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF subpopulations: A new reservoir of stromal targets in pancreatic cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef] [Green Version]
  51. 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]
  52. Biffi, G.; Oni, T.E.; Spielman, B.; Hao, Y.; Elyada, E.; Park, Y.; Preall, J.; Tuveson, D.A. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFβ to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov. 2019, 9, 282–301. [Google Scholar] [CrossRef] [PubMed]
  53. Mortezaee, K. Enriched cancer stem cells, dense stroma, and cold immunity: Interrelated events in pancreatic cancer. J. Biochem. Mol. Toxicol. 2021, 35, e22708. [Google Scholar] [CrossRef] [PubMed]
  54. Evans, R.A.; Diamond, M.S.; Rech, A.J.; Chao, T.; Richardson, M.W.; Lin, J.H.; Bajor, D.L.; Byrne, K.T.; Stanger, B.Z.; Riley, J.L.; et al. Lack of immunoediting in murine pancreatic cancer reversed with neoantigen. JCI Insight 2016, 1, e88328. [Google Scholar] [CrossRef] [PubMed]
  55. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  56. Mazzieri, R.; Pucci, F.; Moi, D.; Zonari, E.; Ranghetti, A.; Berti, A.; Politi, L.S.; Gentner, B.; Brown, J.L.; Naldini, L.; et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell 2011, 19, 512–526. [Google Scholar] [CrossRef]
  57. Halbrook, C.J.; Pontious, C.; Kovalenko, I.; Lapienyte, L.; Dreyer, S.; Lee, H.-J.; Thurston, G.; Zhang, Y.; Lazarus, J.; Sajjakulnukit, P.; et al. Macrophage-Released Pyrimidines Inhibit Gemcitabine Therapy in Pancreatic Cancer. Cell Metab. 2019, 29, 1390–1399.e6. [Google Scholar] [CrossRef]
  58. Greten, T.F. Myeloid-derived suppressor cells in pancreatic cancer: More than a hidden barrier for antitumour immunity? Gut 2014, 63, 1690–1691. [Google Scholar] [CrossRef]
  59. 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]
  60. Zhang, Y.; Velez-Delgado, A.; Mathew, E.; Li, D.; Mendez, F.M.; Flannagan, K.; Rhim, A.D.; Simeone, D.M.; Beatty, G.L.; Pasca di Magliano, M. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017, 66, 124–136. [Google Scholar] [CrossRef] [Green Version]
  61. 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]
  62. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, Y.; Chandra, V.; Riquelme Sanchez, E.; 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, e20190354. [Google Scholar] [CrossRef] [PubMed]
  64. Deng, J.; Kang, Y.; Cheng, C.-C.; Li, X.; Dai, B.; Katz, M.H.; Men, T.; Kim, M.P.; Koay, E.A.; Huang, H.; et al. DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight 2021, 6, e146133. [Google Scholar] [CrossRef]
  65. Nielsen, S.R.; Strøbech, J.E.; Horton, E.R.; Jackstadt, R.; Laitala, A.; Bravo, M.C.; Maltese, G.; Jensen, A.R.D.; Reuten, R.; Rafaeva, M.; et al. Suppression of tumor-associated neutrophils by lorlatinib attenuates pancreatic cancer growth and improves treatment with immune checkpoint blockade. Nat. Commun. 2021, 12, 3414. [Google Scholar] [CrossRef]
  66. Whatcott, C.J.; Diep, C.H.; Jiang, P.; Watanabe, A.; LoBello, J.; Sima, C.; Hostetter, G.; Shepard, H.M.; Von Hoff, D.D.; Han, H. Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin. Cancer Res. 2015, 21, 3561–3568. [Google Scholar] [CrossRef] [PubMed]
  67. Shah, V.M.; Sheppard, B.C.; Sears, R.C.; Alani, A.W. Hypoxia: Friend or Foe for drug delivery in Pancreatic Cancer. Cancer Lett. 2020, 492, 63–70. [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, JCO2102616. [Google Scholar] [CrossRef]
  69. Zhang, Z.; Ji, S.; Zhang, B.; Liu, J.; Qin, Y.; Xu, J.; Yu, X. Role of angiogenesis in pancreatic cancer biology and therapy. Biomed. Pharmacother. 2018, 108, 1135–1140. [Google Scholar] [CrossRef]
  70. Danilova, L.; Ho, W.J.; Zhu, Q.; Vithayathil, T.; De Jesus-Acosta, A.; Azad, N.S.; Laheru, D.A.; Fertig, E.J.; Anders, R.; Jaffee, E.M.; et al. Programmed Cell Death Ligand-1 (PD-L1) and CD8 Expression Profiling Identify an Immunologic Subtype of Pancreatic Ductal Adenocarcinomas with Favorable Survival. Cancer Immunol. Res. 2019, 7, 886–895. [Google Scholar] [CrossRef]
  71. Collisson, E.A.; Bailey, P.; Chang, D.K.; Biankin, A.V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 207–220. [Google Scholar] [CrossRef] [PubMed]
  72. Sivakumar, S.; de Santiago, I.; Chlon, L.; Markowetz, F. Master regulators of oncogenic KRAS response in pancreatic cancer: An integrative network biology analysis. PLoS Med. 2017, 14, e1002223. [Google Scholar] [CrossRef] [PubMed]
  73. Jones, S.; Hruban, R.H.; Kamiyama, M.; Borges, M.; Zhang, X.; Parsons, D.W.; Lin, J.C.-H.; Palmisano, E.; Brune, K.; Jaffee, E.M.; et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 2009, 324, 217. [Google Scholar] [CrossRef]
  74. DiGiuseppe, J.A.; Redston, M.S.; Yeo, C.J.; Kern, S.E.; Hruban, R.H. p53-independent expression of the cyclin-dependent kinase inhibitor p21 in pancreatic carcinoma. Am. J. Pathol. 1995, 147, 884–888. [Google Scholar]
  75. Moskaluk, C.A.; Hruban, R.H.; Kern, S.E. p16 and K-ras gene mutations in the intraductal precursors of human pancreatic adenocarcinoma. Cancer Res. 1997, 57, 2140–2143. [Google Scholar]
  76. Iacobuzio-Donahue, C.A.; Fu, B.; Yachida, S.; Luo, M.; Abe, H.; Henderson, C.M.; Vilardell, F.; Wang, Z.; Keller, J.W.; Banerjee, P.; et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J. Clin. Oncol. 2009, 27, 1806–1813. [Google Scholar] [CrossRef] [PubMed]
  77. Biankin, A.V.; Kench, J.G.; Colvin, E.K.; Segara, D.; Scarlett, C.J.; Nguyen, N.Q.; Chang, D.K.; Morey, A.L.; Lee, C.-S.; Pinese, M.; et al. Expression of S100A2 calcium-binding protein predicts response to pancreatectomy for pancreatic cancer. Gastroenterology 2009, 137, 558–568.e11. [Google Scholar] [CrossRef]
  78. Dreyer, S.B.; Pinese, M.; Jamieson, N.B.; Scarlett, C.J.; Colvin, E.K.; Pajic, M.; Johns, A.L.; Humphris, J.L.; Wu, J.; Cowley, M.J.; et al. Precision oncology in surgery: Patient selection for operable pancreatic cancer. Ann. Surg. 2020, 272, 366–376. [Google Scholar] [CrossRef]
  79. Collisson, E.A.; Sadanandam, A.; Olson, P.; Gibb, W.J.; Truitt, M.; Gu, S.; Cooc, J.; Weinkle, J.; Kim, G.E.; Jakkula, L.; et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 2011, 17, 500–503. [Google Scholar] [CrossRef]
  80. Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.H.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178. [Google Scholar] [CrossRef]
  81. Puleo, F.; Nicolle, R.; Blum, Y.; Cros, J.; Marisa, L.; Demetter, P.; Quertinmont, E.; Svrcek, M.; Elarouci, N.; Iovanna, J.; et al. Stratification of pancreatic ductal adenocarcinomas based on tumor and microenvironment features. Gastroenterology 2018, 155, 1999–2013.e3. [Google Scholar] [CrossRef] [PubMed]
  82. Aung, K.L.; Fischer, S.E.; Denroche, R.E.; Jang, G.-H.; Dodd, A.; Creighton, S.; Southwood, B.; Liang, S.-B.; Chadwick, D.; Zhang, A.; et al. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin. Cancer Res. 2018, 24, 1344–1354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Wang, S.; Zheng, Y.; Yang, F.; Zhu, L.; Zhu, X.-Q.; Wang, Z.-F.; Wu, X.-L.; Zhou, C.-H.; Yan, J.-Y.; Hu, B.-Y.; et al. The molecular biology of pancreatic adenocarcinoma: Translational challenges and clinical perspectives. Signal Transduct. Target. Ther. 2021, 6, 249. [Google Scholar] [CrossRef]
  84. Hayashi, A.; Fan, J.; Chen, R.; Ho, Y.-J.; Makohon-Moore, A.P.; Lecomte, N.; Zhong, Y.; Hong, J.; Huang, J.; Sakamoto, H.; et al. A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma. Nat. Cancer 2020, 1, 59–74. [Google Scholar] [CrossRef] [PubMed]
  85. Chan-Seng-Yue, M.; Kim, J.C.; Wilson, G.W.; Ng, K.; Figueroa, E.F.; O’Kane, G.M.; Connor, A.A.; Denroche, R.E.; Grant, R.C.; McLeod, J.; et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat. Genet. 2020, 52, 231–240. [Google Scholar] [CrossRef] [PubMed]
  86. Ren, B.; Cui, M.; Yang, G.; Wang, H.; Feng, M.; You, L.; Zhao, Y. Tumor microenvironment participates in metastasis of pancreatic cancer. Mol. Cancer 2018, 17, 108. [Google Scholar] [CrossRef] [PubMed]
  87. Thayer, S.P.; di Magliano, M.P.; Heiser, P.W.; Nielsen, C.M.; Roberts, D.J.; Lauwers, G.Y.; Qi, Y.P.; Gysin, S.; Fernández-del Castillo, C.; Yajnik, V.; et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003, 425, 851–856. [Google Scholar] [CrossRef]
  88. Bailey, J.M.; Swanson, B.J.; Hamada, T.; Eggers, J.P.; Singh, P.K.; Caffery, T.; Ouellette, M.M.; Hollingsworth, M.A. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin. Cancer Res. 2008, 14, 5995–6004. [Google Scholar] [CrossRef]
  89. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef]
  90. Catenacci, D.V.T.; Junttila, M.R.; Karrison, T.; Bahary, N.; Horiba, M.N.; Nattam, S.R.; Marsh, R.; Wallace, J.; Kozloff, M.; Rajdev, L.; et al. Randomized phase ib/ii study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2015, 33, 4284–4292. [Google Scholar] [CrossRef]
  91. Ko, A.H.; LoConte, N.; Tempero, M.A.; Walker, E.J.; Kate Kelley, R.; Lewis, S.; Chang, W.-C.; Kantoff, E.; Vannier, M.W.; Catenacci, D.V.; et al. A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma. Pancreas 2016, 45, 370–375. [Google Scholar] [CrossRef] [PubMed]
  92. De Jesus-Acosta, A.; Sugar, E.A.; O’Dwyer, P.J.; Ramanathan, R.K.; Von Hoff, D.D.; Rasheed, Z.; Zheng, L.; Begum, A.; Anders, R.; Maitra, A.; et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br. J. Cancer 2020, 122, 498–505. [Google Scholar] [CrossRef] [PubMed]
  93. Jiang, H.; Hegde, S.; Knolhoff, B.L.; Zhu, Y.; Herndon, J.M.; Meyer, M.A.; Nywening, T.M.; Hawkins, W.G.; Shapiro, I.M.; Weaver, D.T.; et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016, 22, 851–860. [Google Scholar] [CrossRef] [PubMed]
  94. Jiang, H.; Liu, X.; Knolhoff, B.L.; Hegde, S.; Lee, K.B.; Jiang, H.; Fields, R.C.; Pachter, J.A.; Lim, K.-H.; DeNardo, D.G. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut 2020, 69, 122–132. [Google Scholar] [CrossRef] [PubMed]
  95. Abdollahi, A.; Schwager, C.; Kleeff, J.; Esposito, I.; Domhan, S.; Peschke, P.; Hauser, K.; Hahnfeldt, P.; Hlatky, L.; Debus, J.; et al. Transcriptional network governing the angiogenic switch in human pancreatic cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 12890–12895. [Google Scholar] [CrossRef]
  96. Hill, A.; Sadda, J.; LaBarge, M.A.; Hurria, A. How cancer therapeutics cause accelerated aging: Insights from the hallmarks of aging. J. Geriatr. Oncol. 2020, 11, 191–193. [Google Scholar] [CrossRef]
  97. Duperret, E.K.; Trautz, A.; Ammons, D.; Perales-Puchalt, A.; Wise, M.C.; Yan, J.; Reed, C.; Weiner, D.B. Alteration of the Tumor Stroma Using a Consensus DNA Vaccine Targeting Fibroblast Activation Protein (FAP) Synergizes with Antitumor Vaccine Therapy in Mice. Clin. Cancer Res. 2018, 24, 1190–1201. [Google Scholar] [CrossRef]
  98. Shi, Y.; Gao, W.; Lytle, N.K.; Huang, P.; Yuan, X.; Dann, A.M.; Ridinger-Saison, M.; DelGiorno, K.E.; Antal, C.E.; Liang, G.; et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 2019, 569, 131–135. [Google Scholar] [CrossRef]
  99. Dauer, P.; Zhao, X.; Gupta, V.K.; Sharma, N.; Kesh, K.; Gnamlin, P.; Dudeja, V.; Vickers, S.M.; Banerjee, S.; Saluja, A. Inactivation of Cancer-Associated-Fibroblasts Disrupts Oncogenic Signaling in Pancreatic Cancer Cells and Promotes Its Regression. Cancer Res. 2018, 78, 1321–1333. [Google Scholar] [CrossRef]
  100. Chen, X.; Jia, F.; Li, Y.; Deng, Y.; Huang, Y.; Liu, W.; Jin, Q.; Ji, J. Nitric oxide-induced stromal depletion for improved nanoparticle penetration in pancreatic cancer treatment. Biomaterials 2020, 246, 119999. [Google Scholar] [CrossRef]
  101. Lo, A.; Wang, L.-C.S.; Scholler, J.; Monslow, J.; Avery, D.; Newick, K.; O’Brien, S.; Evans, R.A.; Bajor, D.J.; Clendenin, C.; et al. Tumor-Promoting Desmoplasia Is Disrupted by Depleting FAP-Expressing Stromal Cells. Cancer Res. 2015, 75, 2800–2810. [Google Scholar] [CrossRef]
  102. Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.-C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.B.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [PubMed]
  104. Bockorny, B.; Macarulla, T.; Semenisty, V.; Borazanci, E.; Feliu, J.; Ponz-Sarvise, M.; Abad, D.G.; Oberstein, P.; Alistar, A.; Muñoz, A.; et al. Motixafortide and Pembrolizumab Combined to Nanoliposomal Irinotecan, Fluorouracil, and Folinic Acid in Metastatic Pancreatic Cancer: The COMBAT/KEYNOTE-202 Trial. Clin. Cancer Res. 2021, 27, 5020–5027. [Google Scholar] [CrossRef] [PubMed]
  105. Autio, K.; Oft, M. Pegylated Interleukin-10: Clinical Development of an Immunoregulatory Cytokine for Use in Cancer Therapeutics. Curr. Oncol. Rep. 2019, 21, 19. [Google Scholar] [CrossRef]
  106. Jacobetz, M.A.; Chan, D.S.; Neesse, A.; Bapiro, T.E.; Cook, N.; Frese, K.K.; Feig, C.; Nakagawa, T.; Caldwell, M.E.; Zecchini, H.I.; et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013, 62, 112–120. [Google Scholar] [CrossRef]
  107. Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef]
  108. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.-Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; et al. Randomized Phase III Trial of Pegvorhyaluronidase Alfa with Nab-Paclitaxel Plus Gemcitabine for Patients with Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J. Clin. Oncol. 2020, 38, 3185–3194. [Google Scholar] [CrossRef]
  109. 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]
  110. 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]
  111. A Study of PEGylated Recombinant Human Hyaluronidase in Combination with Nab-Paclitaxel Plus Gemcitabine Compared with Placebo Plus Nab-Paclitaxel and Gemcitabine in Participants with Hyaluronan-High Stage IV Previously Untreated Pancreatic Ductal Adenocarcinoma—Study Results—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/results/NCT02715804?term=ICOS&cond=pancreatic+cancer&phase=123&draw=3&rank=15 (accessed on 17 June 2022).
  112. Mpekris, F.; Panagi, M.; Voutouri, C.; Martin, J.D.; Samuel, R.; Takahashi, S.; Gotohda, N.; Suzuki, T.; Papageorgis, P.; Demetriou, P.; et al. Normalizing the Microenvironment Overcomes Vessel Compression and Resistance to Nano-immunotherapy in Breast Cancer Lung Metastasis. Adv. Sci 2021, 8, 2001917. [Google Scholar] [CrossRef] [PubMed]
  113. Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kindler, H.L.; Niedzwiecki, D.; Hollis, D.; Sutherland, S.; Schrag, D.; Hurwitz, H.; Innocenti, F.; Mulcahy, M.F.; O’Reilly, E.; Wozniak, T.F.; et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: Phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J. Clin. Oncol. 2010, 28, 3617–3622. [Google Scholar] [CrossRef] [PubMed]
  115. Hosein, A.N.; Brekken, R.A.; Maitra, A. Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 487–505. [Google Scholar] [CrossRef]
  116. Aguilera, K.Y.; Rivera, L.B.; Hur, H.; Carbon, J.G.; Toombs, J.E.; Goldstein, C.D.; Dellinger, M.T.; Castrillon, D.H.; Brekken, R.A. Collagen signaling enhances tumor progression after anti-VEGF therapy in a murine model of pancreatic ductal adenocarcinoma. Cancer Res. 2014, 74, 1032–1044. [Google Scholar] [CrossRef]
  117. Maione, F.; Molla, F.; Meda, C.; Latini, R.; Zentilin, L.; Giacca, M.; Seano, G.; Serini, G.; Bussolino, F.; Giraudo, E. Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models. J. Clin. Invest. 2009, 119, 3356–3372. [Google Scholar] [CrossRef]
  118. Casazza, A.; Laoui, D.; Wenes, M.; Rizzolio, S.; Bassani, N.; Mambretti, M.; Deschoemaeker, S.; Van Ginderachter, J.A.; Tamagnone, L.; Mazzone, M. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 2013, 24, 695–709. [Google Scholar] [CrossRef]
  119. Gioelli, N.; Maione, F.; Camillo, C.; Ghitti, M.; Valdembri, D.; Morello, N.; Darche, M.; Zentilin, L.; Cagnoni, G.; Qiu, Y.; et al. A rationally designed NRP1-independent superagonist SEMA3A mutant is an effective anticancer agent. Sci. Transl. Med. 2018, 10, e14903. [Google Scholar] [CrossRef]
  120. Schizas, D.; Charalampakis, N.; Kole, C.; Economopoulou, P.; Koustas, E.; Gkotsis, E.; Ziogas, D.; Psyrri, A.; Karamouzis, M.V. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat. Rev. 2020, 86, 102016. [Google Scholar] [CrossRef]
  121. Watanabe, K.; Luo, Y.; Da, T.; Guedan, S.; Ruella, M.; Scholler, J.; Keith, B.; Young, R.M.; Engels, B.; Sorsa, S.; et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight 2018, 3, e99573. [Google Scholar] [CrossRef]
  122. Lesch, S.; Blumenberg, V.; Stoiber, S.; Gottschlich, A.; Ogonek, J.; Cadilha, B.L.; Dantes, Z.; Rataj, F.; Dorman, K.; Lutz, J.; et al. T cells armed with C-X-C chemokine receptor type 6 enhance adoptive cell therapy for pancreatic tumours. Nat. Biomed. Eng. 2021, 5, 1246–1260. [Google Scholar] [CrossRef] [PubMed]
  123. Leidner, R.; Sanjuan Silva, N.; Huang, H.; Sprott, D.; Zheng, C.; Shih, Y.-P.; Leung, A.; Payne, R.; Sutcliffe, K.; Cramer, J.; et al. Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. N. Engl. J. Med. 2022, 386, 2112–2119. [Google Scholar] [CrossRef] [PubMed]
  124. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
  125. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.M.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef] [PubMed]
  126. Moral, J.A.; Leung, J.; Rojas, L.A.; Ruan, J.; Zhao, J.; Sethna, Z.; Ramnarain, A.; Gasmi, B.; Gururajan, M.; Redmond, D.; et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 2020, 579, 130–135. [Google Scholar] [CrossRef] [PubMed]
  127. Raj, D.; Yang, M.-H.; Rodgers, D.; Hampton, E.N.; Begum, J.; Mustafa, A.; Lorizio, D.; Garces, I.; Propper, D.; Kench, J.G.; et al. Switchable CAR-T cells mediate remission in metastatic pancreatic ductal adenocarcinoma. Gut 2019, 68, 1052–1064. [Google Scholar] [CrossRef]
  128. Nair, N.; Chen, S.-Y.; Lemmens, E.; Chang, S.; Le, D.T.; Jaffee, E.M.; Murphy, A.; Whiting, C.; Müller, T.; Brockstedt, D.G. Single-Cell Immune Competency Signatures Associate with Survival in Phase II GVAX and CRS-207 Randomized Studies in Patients with Metastatic Pancreatic Cancer. Cancer Immunol. Res. 2020, 8, 609–617. [Google Scholar] [CrossRef]
  129. Kitahata, Y.; Kawai, M.; Hirono, S.; Okada, K.-I.; Miyazawa, M.; Motobayashi, H.; Ueno, M.; Hayami, S.; Miyamoto, A.; Yamaue, H. Circulating Tumor DNA as a Potential Prognostic Marker in Patients with Borderline-Resectable Pancreatic Cancer Undergoing Neoadjuvant Chemotherapy Followed by Pancreatectomy. Ann. Surg. Oncol. 2022, 29, 1596–1605. [Google Scholar] [CrossRef]
  130. 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]
  131. 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.; 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]
  132. 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] [PubMed]
  133. Suzuki, N.; Hazama, S.; Iguchi, H.; Uesugi, K.; Tanaka, H.; Hirakawa, K.; Aruga, A.; Hatori, T.; Ishizaki, H.; Umeda, Y.; et al. Phase II clinical trial of peptide cocktail therapy for patients with advanced pancreatic cancer: VENUS-PC study. Cancer Sci. 2017, 108, 73–80. [Google Scholar] [CrossRef] [PubMed]
  134. Bassani-Sternberg, M.; Digklia, A.; Huber, F.; Wagner, D.; Sempoux, C.; Stevenson, B.J.; Thierry, A.-C.; Michaux, J.; Pak, H.; Racle, J.; et al. A Phase Ib Study of the Combination of Personalized Autologous Dendritic Cell Vaccine, Aspirin, and Standard of Care Adjuvant Chemotherapy Followed by Nivolumab for Resected Pancreatic Adenocarcinoma—A Proof of Antigen Discovery Feasibility in Three Patients. Front. Immunol. 2019, 10, 1832. [Google Scholar] [CrossRef]
  135. Siret, C.; Collignon, A.; Silvy, F.; Robert, S.; Cheyrol, T.; André, P.; Rigot, V.; Iovanna, J.; van de Pavert, S.; Lombardo, D.; et al. Deciphering the Crosstalk Between Myeloid-Derived Suppressor Cells and Regulatory T Cells in Pancreatic Ductal Adenocarcinoma. Front. Immunol. 2019, 10, 3070. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, Y.; Lazarus, J.; Steele, N.G.; Yan, W.; Lee, H.-J.; Nwosu, Z.C.; Halbrook, C.J.; Menjivar, R.E.; Kemp, S.B.; Sirihorachai, V.R.; et al. Regulatory T-cell Depletion Alters the Tumor Microenvironment and Accelerates Pancreatic Carcinogenesis. Cancer Discov. 2020, 10, 422–439. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, Y.; Herndon, J.M.; Sojka, D.K.; Kim, K.-W.; Knolhoff, B.L.; Zuo, C.; Cullinan, D.R.; Luo, J.; Bearden, A.R.; Lavine, K.J.; et al. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017, 47, 323–338.e6. [Google Scholar] [CrossRef]
  138. Aykut, B.; Chen, R.; Miller, G. Regulatory T cells keep pancreatic cancer at bay. Cancer Discov. 2020, 10, 345–347. [Google Scholar] [CrossRef]
  139. Vonderheide, R.H. CD40 agonist antibodies in cancer immunotherapy. Annu. Rev. Med. 2020, 71, 47–58. [Google Scholar] [CrossRef]
  140. Beatty, G.L.; Chiorean, E.G.; Fishman, M.P.; Saboury, B.; Teitelbaum, U.R.; Sun, W.; Huhn, R.D.; Song, W.; Li, D.; Sharp, L.L.; et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011, 331, 1612–1616. [Google Scholar] [CrossRef]
  141. Byrne, K.T.; Betts, C.B.; Mick, R.; Sivagnanam, S.; Bajor, D.L.; Laheru, D.A.; Chiorean, E.G.; O’Hara, M.H.; Liudahl, S.M.; Newcomb, C.; et al. Neoadjuvant Selicrelumab, an Agonist CD40 Antibody, Induces Changes in the Tumor Microenvironment in Patients with Resectable Pancreatic Cancer. Clin. Cancer Res. 2021, 27, 4574–4586. [Google Scholar] [CrossRef]
  142. Hegde, S.; Krisnawan, V.E.; Herzog, B.H.; Zuo, C.; Breden, M.A.; Knolhoff, B.L.; Hogg, G.D.; Tang, J.P.; Baer, J.M.; Mpoy, C.; et al. Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell 2020, 37, 289–307.e9. [Google Scholar] [CrossRef] [PubMed]
  143. Zippelius, A.; Schreiner, J.; Herzig, P.; Müller, P. Induced PD-L1 expression mediates acquired resistance to agonistic anti-CD40 treatment. Cancer Immunol. Res. 2015, 3, 236–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. 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] [PubMed]
  145. Padrón, L.J.; Maurer, D.M.; O’Hara, M.H.; O’Reilly, E.M.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; et al. Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: Clinical and immunologic analyses from the randomized phase 2 PRINCE trial. Nat. Med. 2022, 28, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
  146. 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]
  147. Morrison, A.H.; Diamond, M.S.; Hay, C.A.; Byrne, K.T.; Vonderheide, R.H. Sufficiency of CD40 activation and immune checkpoint blockade for T cell priming and tumor immunity. Proc. Natl. Acad. Sci. USA 2020, 117, 8022–8031. [Google Scholar] [CrossRef]
  148. Mitchem, J.B.; Brennan, D.J.; Knolhoff, B.L.; Belt, B.A.; Zhu, Y.; Sanford, D.E.; Belaygorod, L.; Carpenter, D.; Collins, L.; Piwnica-Worms, D.; et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013, 73, 1128–1141. [Google Scholar] [CrossRef]
  149. Candido, J.B.; Morton, J.P.; Bailey, P.; Campbell, A.D.; Karim, S.A.; Jamieson, T.; Lapienyte, L.; Gopinathan, A.; Clark, W.; McGhee, E.J.; et al. CSF1R+ Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype. Cell Rep. 2018, 23, 1448–1460. [Google Scholar] [CrossRef]
  150. 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. Invest. New Drugs 2020, 38, 800–811. [Google Scholar] [CrossRef]
  151. Kumar, V.; Donthireddy, L.; Marvel, D.; Condamine, T.; Wang, F.; Lavilla-Alonso, S.; Hashimoto, A.; Vonteddu, P.; Behera, R.; Goins, M.A.; et al. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017, 32, 654–668.e5. [Google Scholar] [CrossRef]
  152. Hecht, J.R.; Lonardi, S.; Bendell, J.; Sim, H.-W.; Macarulla, T.; Lopez, C.D.; Van Cutsem, E.; Muñoz Martin, A.J.; Park, J.O.; Greil, R.; et al. Randomized Phase III Study of FOLFOX Alone or with Pegilodecakin as Second-Line Therapy in Patients with Metastatic Pancreatic Cancer That Progressed After Gemcitabine (SEQUOIA). J. Clin. Oncol. 2021, 39, 1108–1118. [Google Scholar] [CrossRef] [PubMed]
  153. Conlon, J.; Burdette, D.L.; Sharma, S.; Bhat, N.; Thompson, M.; Jiang, Z.; Rathinam, V.A.K.; Monks, B.; Jin, T.; Xiao, T.S.; et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J. Immunol. 2013, 190, 5216–5225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Jing, W.; McAllister, D.; Vonderhaar, E.P.; Palen, K.; Riese, M.J.; Gershan, J.; Johnson, B.D.; Dwinell, M.B. STING agonist inflames the pancreatic cancer immune microenvironment and reduces tumor burden in mouse models. J. Immunother. Cancer 2019, 7, 115. [Google Scholar] [CrossRef] [PubMed]
  155. Demaria, O.; De Gassart, A.; Coso, S.; Gestermann, N.; Di Domizio, J.; Flatz, L.; Gaide, O.; Michielin, O.; Hwu, P.; Petrova, T.V.; et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc. Natl. Acad. Sci. USA 2015, 112, 15408–15413. [Google Scholar] [CrossRef]
  156. Ager, C.R.; Reilley, M.J.; Nicholas, C.; Bartkowiak, T.; Jaiswal, A.R.; Curran, M.A. Intratumoral STING Activation with T-cell Checkpoint Modulation Generates Systemic Antitumor Immunity. Cancer Immunol. Res. 2017, 5, 676–684. [Google Scholar] [CrossRef]
  157. Luo, M.; Wang, H.; Wang, Z.; Cai, H.; Lu, Z.; Li, Y.; Du, M.; Huang, G.; Wang, C.; Chen, X.; et al. A STING-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 2017, 12, 648–654. [Google Scholar] [CrossRef]
  158. Ager, C.R.; Boda, A.; Rajapakshe, K.; Lea, S.T.; Di Francesco, M.E.; Jayaprakash, P.; Slay, R.B.; Morrow, B.; Prasad, R.; Dean, M.A.; et al. High potency STING agonists engage unique myeloid pathways to reverse pancreatic cancer immune privilege. J. Immunother. Cancer 2021, 9, e003246. [Google Scholar] [CrossRef]
  159. Blair, A.B.; Kim, V.M.; Muth, S.T.; Saung, M.T.; Lokker, N.; Blouw, B.; Armstrong, T.D.; Jaffee, E.M.; Tsujikawa, T.; Coussens, L.M.; et al. Dissecting the stromal signaling and regulation of myeloid cells and memory effector T cells in pancreatic cancer. Clin. Cancer Res. 2019, 25, 5351–5363. [Google Scholar] [CrossRef]
  160. Corbett, T.H.; Roberts, B.J.; Leopold, W.R.; Peckham, J.C.; Wilkoff, L.J.; Griswold, D.P.; Schabel, F.M. Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Res. 1984, 44, 717–726. [Google Scholar]
  161. Hingorani, S.R.; Wang, L.; Multani, A.S.; Combs, C.; Deramaudt, T.B.; Hruban, R.H.; Rustgi, A.K.; Chang, S.; Tuveson, D.A. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005, 7, 469–483. [Google Scholar] [CrossRef]
  162. Steele, N.G.; Biffi, G.; Kemp, S.B.; Zhang, Y.; Drouillard, D.; Syu, L.; Hao, Y.; Oni, T.E.; Brosnan, E.; Elyada, E.; et al. Inhibition of hedgehog signaling alters fibroblast composition in pancreatic cancer. Clin. Cancer Res. 2021, 27, 2023–2037. [Google Scholar] [CrossRef] [PubMed]
  163. Rhim, A.D.; Oberstein, P.E.; Thomas, D.H.; Mirek, E.T.; Palermo, C.F.; Sastra, S.A.; Dekleva, E.N.; Saunders, T.; Becerra, C.P.; Tattersall, I.W.; et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014, 25, 735–747. [Google Scholar] [CrossRef] [PubMed]
  164. Grünwald, B.T.; Devisme, A.; Andrieux, G.; Vyas, F.; Aliar, K.; McCloskey, C.W.; Macklin, A.; Jang, G.H.; Denroche, R.; Romero, J.M.; et al. Spatially confined sub-tumor microenvironments in pancreatic cancer. Cell 2021, 184, 5577–5592.e18. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Landscape of immunomodulatory and stroma-depleting strategies to treat pancreatic cancer. Various immune cells and stromal cells constitute the pancreatic tumor microenvironment and can provide druggable targets. Promising multi-modal therapy usually involves two mechanistically independent immunomodulatory strategies or one immunomodulatory therapy combined with stroma-depleting therapy. Enhancing antigen presentation, depleting or inhibiting immunosuppressive components, or activating cytotoxic T cells are immunomodulatory strategies that can reinforce the antitumor efficacy of other therapies. APC, antigen-presenting cell; α-SMA, α-smooth muscle actin; CAF, cancer-associated fibroblast; CCR, chemokine receptor; CSF, colony-stimulating factor; CSF-1R, colony-stimulating factor-1 receptor; CTLA-4, Cytotoxic T-lymphocyte antigen 4; CXCR, C-X-C chemokine receptor; DC, dendritic cell; FAK, focal adhesion kinase; FAP, fibroblast activation protein; HA, hyaluronic acid; MDSC, myeloid-derived suppressor cell; MHC, major histocompability complex; TAM, tumor-associated macrophage.
Figure 1. Landscape of immunomodulatory and stroma-depleting strategies to treat pancreatic cancer. Various immune cells and stromal cells constitute the pancreatic tumor microenvironment and can provide druggable targets. Promising multi-modal therapy usually involves two mechanistically independent immunomodulatory strategies or one immunomodulatory therapy combined with stroma-depleting therapy. Enhancing antigen presentation, depleting or inhibiting immunosuppressive components, or activating cytotoxic T cells are immunomodulatory strategies that can reinforce the antitumor efficacy of other therapies. APC, antigen-presenting cell; α-SMA, α-smooth muscle actin; CAF, cancer-associated fibroblast; CCR, chemokine receptor; CSF, colony-stimulating factor; CSF-1R, colony-stimulating factor-1 receptor; CTLA-4, Cytotoxic T-lymphocyte antigen 4; CXCR, C-X-C chemokine receptor; DC, dendritic cell; FAK, focal adhesion kinase; FAP, fibroblast activation protein; HA, hyaluronic acid; MDSC, myeloid-derived suppressor cell; MHC, major histocompability complex; TAM, tumor-associated macrophage.
Medicina 58 01298 g001
Table 1. Ongoing clinical trials involving potential therapies against pancreatic cancer that target the tumor microenvironment.
Table 1. Ongoing clinical trials involving potential therapies against pancreatic cancer that target the tumor microenvironment.
Trial IdentifierDrugCancer StageMechanismPhaseStarting YearStatus *Notes
Therapeutic strategy: Stromal depletion
NCT03941093PamrevlumabLocally advanced PDACAntifibroticIII2019ActiveWith or without chemo
NCT03634332PEGPH20+PembrolizumabAdvanced PDAChyaluronidase+PD-1 inhibitorII2018RecruitingNo control group
Therapeutic strategy: Immune modulation
NCT02345408CCX872Unresectable PDACCCR2 inhibitorIb2015ActiveWith or without chemo
NCT02767557TocilizumabAdvanced PDACIL-6 inhibitorII2016ActiveWith chemo (Gem+Nab-P) compared to chemo alone
NCT03184870BMS-813160Advanced PCa and CRCCCR2/5 inhibitorI/II2017ActiveWith or without chemo or nivolumab
NCT03941093ABBV-927Advanced PDACCD40 inhibitorII2019ActiveWith or without chemo
NCT03336216Cabiralizumab+NivolumabAdvanced PDACCSF-1R inhibitor+PD-1 inhibitorII2017ActiveWith or without chemo
Therapeutic strategy: Tumor vaccine
NCT03190265CY/GVAX+CyclophosphamidePreviously treated advanced PDACAllogeneic cells secreting GM-CSF, as well as alkylating agentsII2017ActiveAddition to Nivolumab+Ipilimumab+CRS-207
NCT03006302CY/GVAXAdvanced PDACAllogeneic cells secreting GM-CSFII2016ActiveAddition to Epacadostat+Pembrolizumab+CRS-207 (IDO1 inhibitor+PD-1 inhibitor+Listeria monocytogenes-expressing mesothelin (tumor vaccine)
NCT02705196LOAd703+chemoEarly-stage resectable PDACRecombinant adenovirus (intratumoral injection)I/II2016ActiveWith or without atezolizumab (PD-L1 inhibitor)
Therapeutic strategy: Multiple immunomodulatory approaches
NCT02583477MEDI4736+AZD5069Advanced PDACCXCR2 inhibitor+PD1 inhibitorI/II2015ActiveCompared to MEDI4736+ nab-paclitaxel+ gemcitabine
NCT02826486BL-8040+pembrolizumabAdvanced PDACCXCR4 inhibitor+PD1 inhibitorII2016ActiveCompared to BL-8040+5-FU
NCT02826486BL-8040+ PembrolizumabAdvanced PDACCXCR4 inhibitor+PD-1 inhibitorII2016ActiveWith or without chemo (Onivyde)
NCT03376659CV301+DurvalumabAdvanced PDAC and CRCPD-L1 inhibitor+recombinant polypeptidesI/II2017ActiveWith or without chemo
NCT04191421Siltuximab+ SpartalizumabAdvanced PDACIL-6 inhibitor+PD-1 inhibitorI/II2019RecruitingNone
NCT05419479APX005M+Domvanalimab+ImberelimabAdvanced PCaCD40 agonist+TIGIT inhibitor+PD1 inhibitorI/II2022RecruitingWith or without chemo
Therapeutic strategy: The combination of stromal depletion and immune modulation
NCT04171219Talabostat/PembrolizumabAdvanced PDACFAP inhibitor combining PD1 inhibitorII2019RecruitingNone
NCT04177810Plerixafor/CemiplimabAdvanced PDACCXCR4 inhibitor/PD1 inhibitorII2019RecruitingNone
* As of 30 June 2022. Abbreviations: CRC, colorectal cancer; CCR, chemokine receptors; CXCR4, C-X-C chemokine receptor type 4; chemo, chemotherapy; DNMT, DNA methyltransferase; Gem, gemcitabine; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL-6, Interleukin 6; MPS, methoxsalen, phenytoin, and sirolimus; Nab-P, nab-paclitaxel; PD-1, Programmed cell death protein 1; PCa, pancreatic cancer; PDAC, pancreatic ductal adenocarcinoma; SCLC, small cell lung cancer; TIGIT, T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, T.; Cheng, S.; Xu, Q.; Wang, Z. Management of Advanced Pancreatic Cancer through Stromal Depletion and Immune Modulation. Medicina 2022, 58, 1298. https://doi.org/10.3390/medicina58091298

AMA Style

Liu T, Cheng S, Xu Q, Wang Z. Management of Advanced Pancreatic Cancer through Stromal Depletion and Immune Modulation. Medicina. 2022; 58(9):1298. https://doi.org/10.3390/medicina58091298

Chicago/Turabian Style

Liu, Tiantong, Sihang Cheng, Qiang Xu, and Zhiwei Wang. 2022. "Management of Advanced Pancreatic Cancer through Stromal Depletion and Immune Modulation" Medicina 58, no. 9: 1298. https://doi.org/10.3390/medicina58091298

Article Metrics

Back to TopTop