Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 1.
Published in final edited form as: Nat Rev Clin Oncol. 2020 May 12;17(9):527–540. doi: 10.1038/s41571-020-0363-5

The tumour microenvironment in pancreatic cancer — clinical challenges and opportunities

Won Jin Ho, Elizabeth M Jaffee, Lei Zheng
PMCID: PMC7442729  NIHMSID: NIHMS1606762  PMID: 32398706

Abstract

Metastatic pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumours despite the use of multi-agent conventional chemotherapy regimens. Such poor outcomes have fuelled ongoing efforts to exploit the tumour microenvironment (TME) for therapy, but strategies aimed at deconstructing the surrounding desmoplastic stroma and targeting the immunosuppressive pathways have largely failed. In fact, evidence has now shown that the stroma is multi-faceted, which illustrates the complexity of exploring features of the TME as isolated targets. In this Review, we describe ways in which the PDAC microenvironment has been targeted and note the current understanding of the clinical outcomes that have unexpectedly contradicted preclinical observations. We also consider the more sophisticated therapeutic strategies under active investigation — multi-modal treatment approaches and exploitation of biologically integrated targets — which aim to remodel the TME against PDAC.


Pancreatic cancer, comprising mostly pancreatic ductal adenocarcinoma (PDAC), is an extremely lethal disease1, with 45,750 estimated deaths in the USA in 2019 (REF.2). Symptoms are often non-specific, which means that patients often present at advanced stages. Conventional cytotoxic chemotherapy constitutes the current standard of care for advanced or metastatic PDAC, providing only months of overall survival benefit3,4.

Carcinogenesis of PDAC involves progressive accumulation of driver mutations, including the oncogene KRAS5 and tumour suppressor gene TP53 (REF.6). These molecular perturbations are accompanied by histological changes that represent the different stages of PDAC development. Morphological evolution begins with the formation of precursor lesions, termed pancreatic intraepithelial neoplasia (PanIN)7, with increasing histological grades followed by progression to invasive adenocarcinoma. As the cancer develops, it leads to changes in the surrounding tissue stroma. A key function of any non-transformed tissue stroma is to provide homeostatic response to injury with its immune, vascular and connective tissue components. However, cancer hijacks such physiological responses to create a favourable tumour microenvironment (TME) for its successful growth8. In the words of Harold Dvorak, cancer behaves like “wounds...that never heal”, and stromal transformation is a result of “wound healing gone awry”9.

Given the clear importance of the TME in tumorigenesis, approaches to target specific features within the TME have garnered much attention. For example, in the past decade advances in immuno-oncology have led to ground-breaking therapeutic options for multiple cancer types. However, even immunotherapeutic strategies, such as immune-checkpoint inhibition, have yielded limited responses in PDACs10. Furthermore, therapeutic strategies aimed at ablating the stromal barriers that restrict drug delivery have also demonstrated disappointing and contradictory responses11,12.

In this Review, we provide an overview of the complexities and the multi-faceted nature of several therapeutic targets within the PDAC microenvironment. We also examine some of the multi-modal strategies that are currently under investigation and designed to overcome the challenges by reprogramming the stroma into an antitumour milieu.

Limitations of targeting desmoplasia

A histopathological hallmark of PDAC is a desmoplastic reaction to the tumour; this hallmark is present in both primary and metastatic tumours13. Myofibroblast-like cells in the pancreas (that is, pancreatic stellate cells) are activated by cancer cells to produce fibrosis surrounding the tumour14,15. The resultant desmoplasia is known to be responsible for creating a mechanical barrier around the tumour cells, preventing appropriate vascularization and thus limiting exposure to chemotherapy and leading to poor immune cell infiltration16. Early research largely stemmed from the idea that the surrounding desmoplasia is tumour promoting (FIG. 1; BOX 1); this view of its role is an imperfect one. The current understanding is that desmoplasia is in fact multi-faceted and that a more holistic approach to targeting the stroma is warranted.

Fig. 1 |. Targeting PDAC-associated stroma.

Fig. 1 |

The role of the stroma to either promote or resist tumour formation and progression is influenced by the surrounding signals. Both cell–cell and paracrine interactions between cancer-associated fibroblasts (CAFs) and cancer cells are involved in programming the stroma. CAFs, key constituents of the pancreatic ductal adenocarcinoma (PDAC) stroma, are heterogeneous, and include myofibroblastic, inflammatory and antigen-presenting subtypes. Fibroblasts in proximity to cancer cells are induced by transforming growth factor-β (TGFβ) from cancer cells into myofibroblastic CAFs, producing the mechanical barrier that can be both tumour promoting and antitumour. Inflammatory CAFs, located in the stroma away from the cancer cells, are reprogrammed by cancer-secreted IL-1 to produce cytokines and chemokines (for example, IL-6), which further promote cancer growth. The subsequently developed antigen-presenting CAFs, which express MHC class II molecules, modulate the immune cells in the stroma. Approaches to deconstruct the stroma have included the use of matrix metalloproteinase (MMP) inhibitors, hyaluronidase, Sonic hedgehog (SHH) inhibitors, fibroblast activation protein (FAP) targeting agents and CXCR4 inhibitors. Ab, antibody; CAR, chimeric antigen receptor; ECM, extracellular matrix.

Box 1 | Limitations in preclinical assessment of novel therapies.

Studying therapies that target features within the tumour microenvironment (TME) requires that 1) the cancer resides within an intact biological stroma (that is, in vivo tissue space) and 2) the cancer is recognized by the surrounding stroma and the immune system as self. Therefore, the most ideal models for preclinical testing of TME-targeted therapies consist of syngeneic transplantation of cancer cells or sporadic models of carcinogenesis rather than any in vitro culture systems or xenograft models. Early studies in the 1980s commonly used a mouse model of pancreatic cancer that was generated in C57BL/6 mice using a local implantation of the carcinogen 3-methylcholanthrene186. The cell line established from this model, Panc02, can be syngeneically transplanted to assess therapeutics. Given the method of carcinogenesis, the Panc02 cell line, unsurprisingly, harbours numerous mutations (586 missense, 19 stop gains and 32 indels)187. While Panc02 exhibits a stop-gain mutation in Smad4 it does not have mutations in Kras or Tp53 (REF.187). Based on these genetic differences between Panc02 and the majority of human pancreatic ductal adenocarcinomas that bear KRAS and TP53 mutations188, successful translation of the findings in syngeneic models based on Panc02 are limited. Nonetheless, one of the first proof-of-concept animal models demonstrating the benefit of STING agonist vaccination in cancer was the Panc02 model121.

To overcome the limitations of the Panc02 model, genetically engineered mouse models were developed in which KrasG12D and Tp53R172H mutations were inserted under Cre recombinase expression driven by the pancreas-specific promoter Pdx1 (the ‘KPC model’)189. In fact, most of the TME-oriented studies that have led to clinical trials in pancreatic cancer in the past decade have utilized the KPC model16,41,47,66,119,152,174. In all of these studies, however, the most common method of assessing therapeutic efficacy has been to begin therapy at the time when ultrasound shows that a minimum tumour size is reached at the primary pancreatic site (for example, 5–10 mm in diameter). The mice are then followed for survival and maximal reduction of tumour size via ultrasound measurements. Although this method is perhaps the most accurate way to recapitulate the real-life heterogeneity in disease progression and metastatic spread, the model fails to emulate how most of the therapies are tested in clinical trials in which patients are enrolled with metastatic disease and very commonly after prior lines of therapy. Many studies have demonstrated that the TME in the primary site is different from that of metastatic sites and that prior therapy reprogrammes the tumour13,51,53,54. Furthermore, reliable biological or molecular correlates are often not defined during the preclinical stage as it is challenging to do so. Without defining key correlates a priori, additional development of or gain of insight from the failed therapy becomes even more limited. To improve the chances of translational success of preclinical findings, it might be preferable, in some cases, to design the preclinical model to mimic the disease state in which the therapy is to be tested. One such example is a murine pancreatic tumour model in which metastatic liver disease is modelled by the intraportal injection of KPC cells via the splenic vasculature190. Alternatively, neoadjuvant trial paradigms in humans enable evaluation of an intact TME and enable a deeper understanding of the effects of therapy, allowing ‘reverse’ translations (that is, analysis of clinical correlates informing preclinical target or therapy development).

Matrix metalloproteinases

The surrounding extracellular matrix (ECM) has long been implicated in the regulation of cancer progression (for example, migration and invasion). Efforts in the late 1990s and early 2000s focused on the non-specific alteration of the ECM within the surrounding stroma by targeting the proteins that remodel the ECM. Studies showed that proteolytic matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs are differentially expressed between non-transformed pancreas and PDAC tissues, with higher expression of particular MMPs being associated with metastatic disease and/or poorer prognosis1719. Increased expression of MMP2, a type IV collagenase detected within the stromal components in pancreatic cancer specimens, was found to increase invasiveness in vitro and to correlate with the degree of desmoplasia2023. MMP7, a zinc-dependent endopeptidase predominantly expressed by glandular epithelial cells, is overexpressed in PanIN and PDAC24,25 and contributes to tumour growth and metastasis in a mutant Kras-driven mouse model of PDAC26.

The interplay between stromal cells and cancer cells via MMPs is further exemplified by the role of the tumour cell-associated MMP inducer (EMMPRIN) in stimulating MMP2, MMP9 and EMMPRIN production following co-culture of EMMPRIN-expressing tumour cells and fibroblasts27. These observations supported the rationale behind the development and application of multi-MMP inhibitors to suppress cancer progression in the B16 melanoma, colon xenograft and gastric xenograft mouse models2830. Despite the preclinical successes in other cancers and overall tolerability in patients, MMP inhibitors such as marimastat and tanomastat failed to show any significant clinical activity in patients with advanced-stage pancreatic cancer3133, suggesting that non-specific targeting of the ECM alone is not effective in pancreatic cancer.

Hyaluronan

A more specific approach to disrupting the ECM within the desmoplastic barrier is targeting hyaluronan, a non-sulfated glycosaminoglycan. Hyaluronan is a major constituent of the stromal ECM, and high deposition of hyaluronan in PDAC is associated with poor prognosis13,34. On the basis of this association, researchers investigated an enzymatic approach to targeting the desmoplastic barrier using human recombinant PH20 hyaluronidase (PEGPH20). A 2013 study demonstrated that targeted depletion of hyaluronan led to improved vascular permeability and increased drug delivery in a mouse model of PDAC, leading to improved chemotherapeutic efficacy when used in combination with cytotoxic chemotherapy with gemcitabine34.

Subsequently, clinical trials investigated the effects of PEGPH20 with two standard-of-care combination chemotherapeutic regimens, gemcitabine plus nab-paclitaxel12 and FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan and oxaliplatin)11. A randomized phase II trial showed that the addition of PEGPH20 enhanced the effects of gemcitabine plus nab-paclitaxel as measured by improved progression-free survival (PFS)12, but another phase Ib/II trial showed that adding PEGPH20 reduced overall survival in patients receiving FOLFIRINOX11. Furthermore, a follow-up phase III trial revealed that combining PEGPH20 with gemcitabine plus nab-paclitaxel did not improve overall survival when compared with gemcitabine plus nab-paclitaxel alone (HR 1.00; P = 0.97)35. The failure of PEGPH20 to enhance the efficacy of chemotherapy does not necessarily exclude ECM-targeting agents from future anticancer therapeutic developments but suggests that this component of the desmoplastic barrier does not sufficiently account for the ineffectiveness of chemotherapeutics in PDAC.

Sonic hedgehog signaling

Distinct from directly targeting a specific component of the ECM, another approach targeting desmoplasia is to focus on a specific signalling pathway responsible for the development of tumour stroma. The hedgehog signalling pathway is key in pancreas development. During embryogenesis, repression of endodermal Sonic hedgehog (SHH) by inhibin-βB and FGF2 permits expression of Pdx1 and insulin (Ins), which then initiates pancreatic differentiation36. The fact that SHH inhibition initiates pancreatic differentiation was corroborated by the observation that an inhibitor of SHH, cyclopamine, promotes heterotopic expansion of pancreatic tissues into adjacent endodermic areas37.

Unsurprisingly, studies have shown that dysregulated hedgehog signalling leads to pancreatic carcinogenesis. SHH expression under Pdx1 control results in the development of tubular structures mimicking PanIN in mice genetically engineered to express mutated Kras38. Also, injury responses in the pancreas induce hedgehog signalling, which then leads to metaplasia with features of PanIN38. A 2008 study also showed that hedgehog signalling promotes desmoplasia, and that antibody-mediated inhibition and overexpression of SHH were associated with reduced and increased presence of desmoplasia, respectively39. Moreover, SHH overexpression provided paracrine stimulation of stellate cell differentiation and myofibroblast invasion39.

Given the evidence indicating that SHH contributes to both cell-intrinsic carcinogenesis and desmoplastic processes, inhibition of SHH was investigated as a therapeutic strategy in PDAC. In a mouse model of PDAC driven by Kras and Cdkn2a, tumours were found to overexpress SHH, and administration of cyclopamine suppressed tumour growth and prolonged survival in cancer-bearing mice40. Furthermore, similar to findings with hyaluronan, SHH inhibition in the KPC mouse model (where pancreatic cancer is driven by Kras and Tp53 mutations) led to improved vascular density within the tumour and increased antitumour efficacy of gemcitabine, resulting in improved survival41.

The results of clinical trials of SHH inhibition, however, have been largely disappointing. A randomized phase II study was stopped early because preliminary results showed that the combination of the SHH inhibitor saridegib and gemcitabine led to a higher rate of progressive disease than did placebo and gemcitabine42. A randomized phase Ib/II study showed that the addition of the SHH inhibitor vismodegib to gemcitabine did not improve overall survival or PFS43. Another study showed that adding vismodegib to gemcitabine plus nab-paclitaxel did not improve PFS over historical rates observed with chemotherapy alone44.

Reconciling contradictions

A number of lessons can be learned from the observed contradictions between preclinical and clinical responses and across several clinical trials focused on stromal desmoplasia. First, the potential for toxic effects occurring in humans that were not observed in mice makes incorporating novel therapies into any existing treatment paradigm challenging. In humans, the addition of PEGPH20 to nab-paclitaxel plus gemcitabine was associated with increased rates of thrombotic events and ultimately the need for the use of prophylactic anticoagulation12. MMP inhibitors and PEGPH20 were associated with the development of musculoskeletal symptoms, not unexpectedly as both MMP inhibitors and PEGPH20 could in theory alter physiological connective tissue remodelling12,31,32,45. Also, higher rates and severity of nausea, vomiting and diarrhoea were seen with PEGPH20 and FOLFIRINOX compared with FOLFIRINOX alone, which led to reduced treatment duration and dose reductions with the combination11. With these toxic effects occurring in the setting of novel combinations, the efficacy of standard regimens might be adversely affected by such reduced treatment durations and dose reductions. These examples illustrate that even judicious combinations based on rigorous preclinical studies of novel therapies with known mechanisms of action and safety data can lead to unexpected outcomes in clinical trials.

Second, although the tumour-promoting role of desmoplasia is well established, accumulating evidence demonstrates that desmoplasia is not solely tumour promoting but rather a neutral reactive process to carcinogenesis that also has antitumour functions. Specifically, when studying the role of SHH in KPC mice, Shh deletion specifically within pancreatic cancer cells leads to decreased myofibroblastic and desmoplastic content in the stroma in association with reduced survival of the KPC mice, a poorly differentiated histology and increased metastatic ability46. The tumour-suppressive effects of SHH signalling were also observed in another study using Kras-driven and KPC models with genetic deficiency of Shh as well as in mice treated with the SHH inhibitor vismodegib from 5 weeks of age47. Shh-null KPC tumours were also shown to exhibit significantly increased vascular density and sensitivity to anti-VEGFR therapy46.

The contradictory observations between the two outcomes of SHH inhibition in mouse experiments can be partly reconciled by the differences in the duration of SHH inhibition. In other words, acute SHH inhibition seems to help in breaking down the barrier to facilitate enhanced drug delivery, but chronic or early SHH inhibition eventually benefits the tumour. The optimal duration of stroma-targeted therapy is unclear at present. Together, these studies suggest that the natural function of the stroma is to restrain tumour growth, tumour angiogenesis and metastatic spread; however, during cancer development, the stromal cells (that is, surrounding fibroblasts) are reprogrammed by tumour cells to support tumour growth48. Indeed, the stroma is composed of a variety of different stromal cells that have both antitumour and tumour-promoting functions49,50.

Of note, the studies did not take into account the heterogeneity of stromal composition in PDAC. Although the stroma-targeted therapies in the preclinical models are tested against a relatively more homogeneous stromal content, previous observations established that patients with PDAC do in fact show a wide range of diversity within the desmoplasia13. Variability occurs not only between patients but also within patients, manifested by site-to-site variability. One could speculate that stroma-targeted agents would be beneficial in sites with high stromal density and that they could be harmful in sites with low stromal density. Thus, a valuable approach might be to further develop non-invasive methods such as imaging to characterize or quantify the degree and type of stromal content within the tumour as a key component of trial design51. Also, according to consensus clustering of expression levels of key genes, the stroma could be classified as being ‘normal’ or ‘activated’, each portending a different prognosis52. Moreover, stromal heterogeneity is not an independent entity; rather, stromal heterogeneity is inherently linked with tumour heterogeneity as it is able to programme tumour behaviour (via quasi-mesenchymal or epithelial phenotype switching)53. The tumours of patients with metastatic PDAC on presentation were more likely to have a quasi-mesenchymal signature than an epithelial signature54. Interestingly, the quasi-mesenchymal and epithelial subtypes showed different responses to chemotherapy regimens54, with epithelial phenotype tumours being associated with a longer metastasis-free survival than the quasi-mesenchymal phenotype. Therefore, a possibility exists that therapies aimed at the stroma might yield divergent effects owing to molecular heterogeneity.

Stromal cells

Another approach towards reprogramming the ECM has been to focus on the cells that deposit the components of the ECM. Cancer-associated stromal cells or cancer-associated fibroblasts (CAFs) are a heterogeneous group of cells known to be major producers of ECM proteins. These generally spindle-shaped cells are positive for one or more activated fibroblast markers (for example, fibroblast activation protein (FAP) and a-smooth muscle actin) and classically have been linked with various tumour-promoting functions including tumorigenesis, angiogenesis, immunosuppression and metastasis, as reviewed elsewhere55.

Targeting of fibroblasts to treat patients with cancer was first assessed using inhibitors targeting FAP The first clinical trial of FAP inhibition was a phase II trial that used a humanized monoclonal antibody, sibrotuzumab, to inhibit CAFs in patients with colorectal cancer. This trial failed to meet its end point, and sibrotuzumab was not investigated further56.

Small molecule inhibitors of FAP have also been explored in pancreatic cancers. In subcutaneous mouse models of PDAC, the small molecule inhibitor UAMC-1110 did not demonstrate any meaningful activity as a single agent57. Similarly, a phase II trial showed that the combination of talabostat and gemcitabine, while relatively well tolerated, had very limited efficacy against metastatic PDAC58.

Given the lack of success with targeted FAP inhibition, researchers investigated the cellular depletion of activated fibroblasts. On the one hand, genetic deletion of a-smooth muscle actin-expressing fibroblasts in mouse models of PanIN or PDAC led to a disease with a more aggressive phenotype59, suggesting that fibroblasts naturally have a cancer-restraining function. Also consistent with the tumour-constraining functions of the stroma, pancreatic cancer cells co-cultured with irradiated fibroblasts showed increased invasiveness over pancreatic cells co-cultured with non-irradiated fibroblasts60. On the other hand, the adoptive transfer of T cells engineered with chimeric antigen receptors (CARs) specific to FAP (to deplete FAP-expressing CAFs) has been shown to disrupt tumour-promoting desmoplasia and to have antitumour efficacy in a mouse model of lung cancer and in KPC-based pancreatic cancer61,62, motivating the clinical translation of CAR-T cell therapy against FAP (NCT03932565).

The direct targeting of CAFs, however, is complex and can result in unexpected biological outcomes. Studies on fibroblasts in the pancreatic stroma have revealed the heterogeneity of CAFs by highlighting their phenotypic and functional diversity49,50,53,63 (FIG. 1). Fibroblasts are cells that typically facilitate homeostatic wound repair, but cancer has the ability to co-opt their function. Specifically, researchers previously discovered that cancer-led signalling via IL-1 or transforming growth factor-β (TGFβ) can differentiate surrounding fibroblasts into inflammatory CAF and myofibroblastic CAF phenotypes, respectively64. IL-6 secreted by inflammatory CAFs then provides pro-proliferative effects on the tumour whereas myofibroblastic CAFs are stimulated by TGFβ to produce the surrounding stroma. Subsequently, a third subtype of CAFs was characterized that express MHC class II molecules and have the ability to present antigens to CD4+ T cells, suggesting that some CAFs are important for shaping the antitumour immune responses49.

Again, instead of simply eliminating the stromal fibroblasts from the TME, a more sophisticated approach might be to exploit the altered microenvironment, particularly the immune TME, that occurs as a result of stromal reprogramming by tumour cells. As proof of concept, a 2019 murine study demonstrated that the addition of PEGPH20 significantly enhanced the effects of cancer-specific vaccines in promoting T cell infiltration into the TME65. Another study in KPC mice showed that depletion of FAP+ CAFs resulted in the immune control of tumour growth and an effective response to immune-checkpoint inhibitors (ICIs)66. Both of these studies elucidated the importance of CXCL12–CXCR4 signalling as a means of stromal–immune crosstalk, presenting yet another target for therapy.

Related to these preclinical observations, although not specific to pancreatic cancer, a clinical trial examining the efficacy of combining the antibody targeting PD-1 (pembrolizumab) and the FAP inhibitor talabostat is ongoing (NCT03910660). One small molecule inhibitor of CXCR4, AMD3100, which demonstrated efficacy in the KPC mouse model in combination with anti-PD-1/PD-L1 signalling66, is now being studied in patients with metastatic pancreatic cancer in combination with an anti-PD-1 inhibitor (NCT04177810). Another CXCR4 antagonist, the peptide-based motixafortide (BL-8040), is also under active investigation to treat PDAC in combination with standard chemotherapy and pembrolizumab, with encouraging results (NCT02826486)67. These findings are generally consistent with the perspective that a multi-modal alteration of the TME combining stromal and immune modulation is probably a more appropriate therapeutic approach instead of targeted depletion; however, caution is needed given that the combination of nivolumab, an antibody against PD-1, and the anti-CXCR4 monoclonal antibody ulocuplumab failed to demonstrate efficacy against PDAC (NCT02472977).

Immune compartment targeting in PDAC

Immune cells in the TME have a key role in the development and progression of pancreatic cancer. Inflammation has long been linked with PDAC according to epidemiological studies, as reviewed in depth elsewhere68. In a KrasG12V-driven model of PDAC, pancreatic inflammation from exposure to caerulein was essential for carcinogenesis69. Furthermore, using this model, inflammation was shown to inhibit an oncogene-induced senescence programme that physiologically prevents adult acinar cells or precursor lesions from persistent progression towards invasive carcinoma70. In addition, studies have established the Kras-specific immune recognition of mutant Kras-driven cancers using a murine lung tumour model71, T cells from patients with colon cancer72,73 and T cells from patients with pancreatic cancer73,74. These studies have demonstrated a clear link between immunological processes and PDAC carcinogenesis. Despite these findings, PDACs are typically known as immunologically ‘cold’ tumours. Analyses of large PDAC genomic datasets showed that only a subset of pancreatic cancers are immunologically active75,76. Studies have identified high tumour mutation burdens exhibiting neoantigenicity to be a key characteristic of inflamed tumours, especially melanomas and lung cancers. However, PDACs have relatively low tumour mutation burdens7779, which is consistent with the limited responses observed when PDACs are treated with ICIs80,81.

Presenting PDAC antigens to the immune system

To overcome the issue of low immune recognition of PDACs, researchers have explored several vaccine therapy approaches to enhance antigen presentation and drive expansion of tumour-specific T cell clones82 as a way to elicit novel or boost pre-existent immune responses. Strategies targeting PDAC-associated antigens (including telomerase83, KRAS84,85, gastrin86, CEA87, MUC1 (REF88) and mesothelin89,90) have included peptide-based vaccines8385, virus-based vaccines87,91, Listeria-based vaccines90, DNA-based vaccines (neoantigens)92,93 and cell-based vaccines88,89,94. On the basis of the results of a multitude of studies, vaccination strategies have now been well established to yield antigen-specific immunological responses in patients with PDAC83,85,90,95.

Studies have also demonstrated that the use of lethally irradiated allogeneic cell-based vaccines engineered to express granulocyte–macrophage colony-stimulating factor (GM-CSF), such as GM-CSF secreting allogeneic pancreatic tumour cell vaccine (GVAX), successfully recruited immune cell aggregates into the TME with activated signatures and enhanced T cell repertoires96,97. Despite positive immunological responses and encouraging findings in early phase trials, many vaccines including TeloVac (telomerase), Primo Vax (telomerase), PANVAC-V (CEA and MUC1) and algenpantucel-L (two allogeneic PDAC cell lines) failed to show significant clinical benefit in phase III trials82.

The major theme from this series of findings is that vaccination strategies alone might not be sufficient for generating clinically meaningful antitumour effects. Thus, studies are ongoing to explore the effects of combining vaccination strategies with other therapeutic modalities. Importantly, the fact that the overall clinical effect of vaccination strategies is limited despite positive immune recognition of tumours suggests that other immunosuppressive pathways that restrict successful antitumour immune responses are present and could be targeted and reversed.

Targeting immunosuppressive cells to modulate the immune TME

PDAC development is intertwined with multiple types of immunosuppressive cells, including regulatory T (Treg) cells, myeloid-derived suppressor cells (MDSCs) and tumour-associated macrophages (TAMs), and leads to an inherently immunosuppressed TME.

A mechanistic link between KRAS mutations and immunosuppressed TME of PDAC has previously been characterized, in which KrasG12D-dependent upregulation of GM-CSF can lead to recruitment of Gr1+CD11b+ MDSCs and limit antitumour T cell activity98. In fact, infiltration of immunosuppressive cells is detected very early in PDAC carcinogenesis. In the Kras-driven mouse model of PDAC, Treg cells and MDSCs dominated the immune infiltration in early PanIN; effector T cells were scarce and generally lacking activation99. Similarly, in a TGFα-overexpressed Tp53-mutated mouse model of pancreatic cancer, MDSCs were detected in premalignant lesions within the pancreas100.

Analogous to the findings in mouse models, Treg cells are observed in human PanIN and increase with progression to PDAC, and increased prevalence of Treg cells confers poor prognosis for patients with PDAC101. In delineating the function of Treg cells and MDSCs, several depletion experiments established Treg cells to be suppressors of antitumour immune responses, as reviewed elsewhere102. However, a precise understanding of MDSCs has been difficult to achieve given their heterogeneity in both mouse and human contexts103. Nevertheless, MDSCs — which can be further subtyped into being monocytic or granulocytic — are known to also exert immunosuppressive effects on T cells via arginase, nitric oxide synthase, TGFβ, IL-10 and COX2 (REF.103).

In addition to Treg cells and MDSCs, TAMs are known to be involved in PDAC carcinogenesis as their infiltration accompanies KRAS G12D-mediated inflammation104. Macrophages have been shown to drive pancreatic acinar-to-ductal metaplasia via secretion of TNF, RANTES and induction of MMP9 (REF.105). They also secrete IL-6 to drive progression of early lesions via the JAK-STAT3 signalling pathway106. TAMs are not just able to promote cancer growth, they also foster cancer invasiveness by stimulating angiogenesis and inhibit natural killer and T cell function by expressing non-classical MHC class I molecules (for example, HLA-G) and ligands of co-inhibitory receptors PD-1 (PD-L1 and PD-L2) and cytotoxic T lymphocyte antigen 4 (CTLA-4)107. In the early stages of carcinogenesis, PanIN interacts with macrophages in the TME via IL-13 to polarize them towards a more immunosuppressive phenotype (that is, an M2 subtype107). Moreover, persistence of colony-stimulating factor-1 (CSF-1) in the TME polarizes macrophages towards the M2 subtype (whereas GM-CSF shifts macrophages towards CD80+ MHC class IIhigh proinflammatory macrophages)108.

Compared with TAMs, tumour-associated neutrophils (TANs) are less mechanistically established with pancreatic carcinogenesis. However, TANs are detected even in PanIN109, and their presence in the TME is associated with poor prognosis in cancers in general110. Importantly, inhibition of TAN infiltration into KPC tumours by knocking out CXCR2, the key chemotactic receptor for neutrophils, resulted in T cell-dependent suppression of tumour growth111. Therefore, Treg cells, MDSCs, TAMs and TANs provide targets for immune modulation of the PDAC microenvironment.

Strategies to directly target immunosuppressive cells in the TME have been explored (FIG. 2; TABLE 1). One well-studied example is the incorporation of cyclophosphamide in treatment regimens to target Treg cells. Evidence supports the idea that low-dose cyclophosphamide selectively eliminates Treg cells112. Therapeutic strategies have successfully utilized cyclophosphamide in combination with GVAX to augment immune responses to PDAC89,97,113.

Fig. 2 |. Myeloid and Treg targeting strategies to treat PDAC.

Fig. 2 |

Antigen-presenting machinery relying on dendritic cells or inflammatory macrophages (TAM M1) and supported by helper T cells (CD4+ T cells) steers the antitumour immune response to eliminate pancreatic ductal adenocarcinoma (PDAC) — for example, via cytotoxic T cells (CD8+ T cells). However, myeloid-derived suppressor cells (MDSCs), anti-inflammatory tumour-associated macrophages (TAM M2), and regulatory T (Treg) cells regulate these processes via several inhibitory pathways, establishing an immunosuppressive tumour microenvironment. Many strategies to abrogate or overcome these immunological targets have been proposed. Clinically tested approaches are listed in the corresponding boxes with the specific types and names of the agents in parentheses. APC, antigen-presenting cell; CSF-1R, colony-stimulating factor-1 receptor; CTLA-4, cytotoxic T lymphocyte antigen 4; GVAX, granulocyte–macrophage colony-stimulating factor secreting allogeneic pancreatic tumour cell vaccine; TCR, T cell receptor.

Table 1 |.

Clinically investigated novel immune-modulating agents for PDAC

Category Mechanism Agent Clinical trial Phase Notes
Chemokine inhibition CCR2 and CCR5 inhibitor (small molecule) BMS-813160 NCT03184870 I/II With chemotherapy or anti-PD-1 (nivolumab)
CCR2 inhibitor (small molecule) CCX872 NCT02345408 Ib Monotherapy
PF-04136309 NCT02732938 Ib/II With chemotherapy
CXCR2 inhibitor (small molecule) AZD5069 NCT02583477 I/II With anti-PD-L1 (durvalumab)
CXCR4 inhibitor (small molecule) Motixafortide (BL-8040) NCT02826486 II With chemotherapy and anti-PD-1 (pembrolizumab)
Epigenetic modifier Hypomethylating agent Azacitidine NCT01845805 II Adjuvant setting
DNMT inhibitor Guadecitabine NCT03257761 I With anti-PD-L1 (durvalumab)
HDAC inhibitor Entinostat NCT03760614 I With chemotherapy
Cytokine Pegylated IL-10 Pegilodecakin NCT02923921 III With chemotherapy
Checkpoint inhibition Bispecific LAG3–CTLA-4 Ab XmAb22841 NCT03849469 I With anti-PD-1 (pembrolizumab)
Bispecific PD-1–ICOS Ab XmAb23104 NCT03752398 I Monotherapy
Co-stimulator agonism CD40 agonist (mAb) APX005M NCT03214250 I/II With chemotherapy and anti-PD-1 (nivolumab)
STING agonist (mAb) MK-1454 NCT03010176 I With anti-PD-1 (pembrolizumab)
ICOS agonist KY1044 NCT03829501 I/II With anti-PD-L1 (atezolizumab)
Myeloid-specific agents CSF-1R inhibitor (small molecule) BLZ945 NCT02829723 I/II With anti-PD-1 (spartalizumab)
CSF-1R inhibitor (mAb) Cabiralizumab NCT03336216 II With chemotherapy and anti-PD-1 (nivolumab)
IMC-CS4 NCT03153410 I With GVAX, low-dose Cy and anti-PD-1 (pembrolizumab)
CD11b agonist (mAb) GB1275 NCT04060342 I/II With chemotherapy or anti-PD-1 (pembrolizumab)
Vaccines Allogeneic GM-CSF-secreting cells GVAX NCT03190265 II With GVAX, low-dose Cy and anti-PD-1 (nivolumab) and anti-CTLA4 (ipilimumab)
Listeria-based vaccine CRS-207
Dendritic cell-based vaccine mDC3/8-KRAS NCT03592888 I Adjuvant setting
Personalized DNA (neoantigen) NCT03122106 I Adjuvant setting
Personalized mRNA RO7198457 NCT03289962 I With anti-PD-L1 (atezolizumab)
Based on two recombinant poxviruses CV301 NCT03376659 I/II With anti-PD-L1 (durvalumab)
Metabolism targeting Adenosine-generating enzyme CD39 (mAb) TTX-030 NCT03884556 I With chemotherapy or anti-PD-1 (pembrolizumab)
Adenosine-generating enzyme CD73 (mAb) CPI-006 NCT03454451 I With anti-PD-1 (pembrolizumab)
SLC6A8 (creatine transporter) inhibitor RGX-202 NCT03597581 I With chemotherapy
Tyrosine hydroxylase inhibitor Racemetyrosine (SM-88) NCT03512756 II/III With chemotherapy
IDO1 inhibitor Epacadostat NCT03006302 II With GVAX, low-dose Cy and anti-PD-1 (pembrolizumab)
Oncolytic virus Adenovirus (intratumoural injection) LOAd703 NCT02705196 I/II With chemotherapy and anti-PD-L1 (atezolizumab)
Reovirus (intravenous injection) Pelareorep NCT03723915 II With anti-PD-1 (pembrolizumab)
Type I herpes simplex virus (intratumoural injection) Talimogene laherparepvec NCT03086642 I Monotherapy

Ab, antibody; CSF-1R, colony-stimulating factor-1 receptor; CTLA-4, cytotoxic T lymphocyte antigen 4; Cy, cyclophosphamide; DNMT, DNA methyltransferase; GM-CSF, granulocyte–macrophage colony-stimulating factor; GVAX, GM-CSF secreting allogeneic pancreatic tumour cell vaccine; HDAC, histone deacetylase; mAb, monoclonal antibody; PDAC, pancreatic ductal adenocarcinoma.

In addition to low-dose cyclophosphamide, CTLA-4 (REF.114) and neuropilin-1 (REF.115) have been investigated as targets for intratumoural Treg cells. Another example of targeting immunosuppressive cells in the TME, TAMs in particular, is antagonizing the CSF-1 receptor (CSF-1R). The CSF-1R is a member of the receptor protein tyrosine kinase family of growth factor receptors that is expressed by TAMs and MDSCs116. Inhibition of CSF-1R has been shown to substantially deplete TAMs and increase the CD8+:CD4+ T cell ratio in mouse models and has demonstrated efficacy in patients with diffuse-type giant cell tumours108. In pancreatic cancer models, CSF-1R inhibition resulted in increased expression of immune checkpoints, PD-L1 and CTLA-4, and targeting of PD-1 and CTLA-4 demonstrated superior antitumour efficacy to CSF-1R inhibition alone when used in combination with CSF-1R inhibition117. The utility of CSF-1R in augmenting the antitumour immune response was recapitulated in a study showing its efficacy in combination with GVAX and anti-PD-1 therapy in a liver metastatic mouse model of PDAC118. Antibody-based and small molecule inhibition of CSF-1R strategies are actively being investigated clinically in multiple cancer types including PDAC116.

Another approach to reprogramming immunosuppressive cells in the TME is to target CD40, a costimulatory molecule present on antigen-presenting cells including macrophages. Targeting CD40 with an antibody has been shown to induce TAMs to express higher levels of CD86 and MHC class II molecules and to be tumoricidal against PDAC in KPC mice119. Anti-CD40 therapy was also associated with substantial stromal degradation119, establishing CD40 as another integrated therapeutic target that can modify the TME. Early-phase clinical trials for PDAC of an anti-CD40 antibody with gemcitabine and with or without nivolumab (an anti-PD-1 monoclonal antibody) are ongoing and showing promising results120. Other modes of enhancing co-stimulation include targeting STING121,122 and ICOS123 signalling. An agonist cellular vaccine of the STING pathway demonstrated anticancer efficacy in multiple murine models of cancer including a metastatic pancreatic cancer model (Panc02)121. A STING vaccine, MK-1454, is currently being tested in a phase I clinical trial in lymphoma and solid tumours including PDAC with an overall acceptable safety profile and encouraging efficacy (NCT03010176)122. Targeting the ICOS pathway has shown generally promising results in other cancer types with regards to tolerability and efficacy and has been reviewed elsewhere124. The use of KY1044, a fully human antibody that depletes ICOShigh Treg cells while stimulating ICOSlow effector T cells, in solid tumours including PDAC is actively being investigated (NCT03829501)123.

Modulation of chemokine signalling is another approach to altering the immune TME. CCR2, expressed on myeloid cells, interacts with the multi-functional ligand CCL2 (as well as CCL3 and CCL5) to recruit monocytes into the TME125. In mouse models of PDAC, small molecule inhibitors of CCR2 led to blockade of TAM infiltration and improved resistance against tumour progression125. This change in the TME also potentiated anti-PD-1 therapy126. Thus far, early phase trials using the CCR2 inhibitors PF-04136309 and CCX872 in combination with conventional chemotherapy regimens in patients with PDAC have had varying results (NCT02732938, NCT01413022 and NCT02345408)127129. A phase Ib study of PF-04136309 in combination with gemcitabine and nab-paclitaxel raised concerns about pulmonary toxicity and did not show superior efficacy compared with gemcitabine and nab-paclitaxel alone (NCT02732938)129, but more encouraging results were reported when both PF-04136409 and CCX872 were combined with FOLFIRINOX (NCT01413022 and NCT02345408)127,128. Myeloid-targeted effects of CCX872 were also noted as peripheral monocyte counts at baseline inversely correlated with overall survival (NCT02345408)127.

The strategy of targeting CXCR2 was explored in order to inhibit TAN infiltration and demonstrated that small molecule inhibition of CXCR2 can abrogate PDAC metastasis, augment T cell infiltration and synergize with anti-PD-1 therapy to extend survival130. An orthotopic PDAC model also showed that the chemotherapy response could be enhanced by CXCR2 inhibition131. In light of these findings, a phase Ib/II clinical trial investigating AZD5069, an oral small molecule inhibitor of CXCR2, in combination with the PD-L1 inhibitor durvalumab in patients with PDAC has just completed, demonstrating limited efficacy (with median PFS and overall survival durations of 1.6 months and 2.8 months, respectively) (NCT02583477). Additional studies incorporating CCR2 and CXCR2 inhibition into the treatment paradigm are warranted.

Other clinically investigated immune-oriented methods of targeting the PDAC microenvironment include the use of oncolytic viruses (adenovirus132, reovirus133 and herpes simplex virus 1 (REF.134)), epigenetic modifiers135 and bispecific antibodies against immune checkpoints136 (FIG. 2; TABLE 1). A 2018 paper revealed that yet another immune-modulatory agent, pegylated IL-10 (pegilodecakin)137, has failed to meet the primary end point of overall survival in a phase III trial for treating PDAC in combination with chemotherapy, but the search for the next novel effective therapy for PDAC is far from over; many of these approaches are just beginning to be tested in patients with PDAC. Additionally, even unsuccessful immunotherapeutic agents might benefit from more in-depth preclinical investigation, especially when being tested in novel combinations with other modalities.

Exploiting integrated targets of TME

Despite the aforementioned failures of TME-targeted therapies, exploiting the unique features within the PDAC microenvironment as therapeutic targets warrants further investigation. Prior experiences have made it clear that an entirely stroma-based or immune-oriented approach to treating PDAC is of limited benefit (FIG. 3). Instead, more effective remodelling of the TME might be achieved by building on these prior efforts and exploiting particular points of biological convergence.

Fig. 3 |. Remodelling the PDAC microenvironment.

Fig. 3 |

Pancreatic ductal adenocarcinoma (PDAC) is classically surrounded by desmoplastic stroma composed of cancer-associated fibroblasts and extracellular matrix (ECM). The stroma provides a dense mechanical barrier (both antitumour and tumour promoting) against vascularization, immune cell trafficking and cancer invasiveness. The tumour microenvironment is also characterized by the presence of multiple immunosuppressive pathways. Exploiting biologically integrated targets of the stroma (such as glutamine metabolism, transforming growth factor-β (TGFβ) and focal adhesion kinase (FAK) signalling) and the immunosuppressive pathways is the most likely approach to remodel the tumour microenvironment into an effective antitumour environment. MDSC, myeloid-derived suppressor cell; Treg cell, regulatory T cell.

Targeting a metabolic convergence to enable TME remodeling

Cancer cell metabolism can be described by the Warburg effect, in which cancer cells maintain high glycolytic activity in order to grow138. Cancer cells require glutamine to fuel the tricarboxylic acid cycle for continued anabolic metabolism139. Importantly, T cells depend on similar metabolic pathways for successful activation and proliferation140. Thus, cancer cells are able to divert the stroma into a tumour-promoting metabolic environment that hinders T cells from providing proper antitumour immune responses.

Using multiple mouse models, one study demonstrated that broad pharmacological blockade of glutamine metabolism enhanced antitumour immune response by augmenting the nutrient availability with which CD8+ T cells can thrive and maintain an activated phenotype141. The use of anti-PD-1 ICIs in conjunction with a glutamine antagonist was superior to either therapy alone. Of note, glutamine blockade also led to a significant decrease in the activity of the hexosamine biosynthesis pathway141. This pathway is a source of uridine diphosphate N-acetylglucosamine142, an important substrate for hyaluronan synthesis, and thus has an important role in nutrient sensing in the context of stroma generation143,144.

Another study fully recapitulated this concept by showing that glutamine antagonism led to a reduction of hyaluronan in the TME, an increase in CD8+ T cell infiltration and improved sensitivity to anti-PD-1 therapy145. Other researchers demonstrated that IFNγ released from CD8+ T cells downregulates the expression of SLC3A2 and SLC7A11, two subunits of the glutamate–cystine antiporter system xc, and impairs the uptake of cystine by tumour cells146. Cystine is known to counteract lipid peroxidation, and this study demonstrated that lack of cystine induces tumour cell peroxidation and ferroptosis, another independent mechanism of tumour cell death.

These clear examples show how a point of biological convergence, as related to desmoplasia, cancer metabolism and the immune microenvironment, might be a target in order to optimize therapeutic strategies. A well-studied glutamine antagonist, 6-diazo-5-oxo-L-norleucine (DON), was limited by toxic effects in previous trials, but a novel prodrug form of DON has been developed and is under investigation147. Other metabolic targets including the adenosine-generating enzymes CD39 and CD73 (REF.148), creatine transporter SLC6A8 (REFS149,150) and tryptophan catabolic enzyme IDO1 (REF.151) are also under investigation (FIG. 2; TABLE 1).

TME remodelling by targeting the focal adhesion kinase pathway

Studies have established the importance of the focal adhesion kinase (FAK) signalling pathway in shaping the PDAC stroma152,153. FAK signalling has long been implicated in processes of wound healing and pathologic fibrosis across various organs154158. FAK has a mechanosensing role in propagating activating signals toward tissue fibrosis within fibrogenic cells such as cardiac myocytes and fibroblasts following stretching or loading157,159.

In direct relevance to cancer cells, FAK overexpression has been well established as a feature of PDAC, and inhibition of FAK has been shown to suppress pancreatic cancer cell growth, survival and spread160165. Accumulating evidence suggests the importance of FAK in integrating cell–cell or cell–matrix interaction signals with immunomodulation. Specifically, FAK functions downstream of αvβ3 integrin to positively regulate interferon signalling towards expression of PD-L1 upon binding of αvβ3 integrin to ECM166. Also within cancer cells, FAK signalling primes a more immunosuppressive TME via recruitment of Treg cells via transcriptional activation of Ccl5 expression167. This pathway is bolstered by FAK-induced expression and nuclear translocation of IL-33 (REF.168).

Leveraging the biology that converges on FAK signalling, a 2016 study demonstrated that small molecule inhibition of FAK resulted in significantly suppressed tumour growth and increased survival of KPC mice in association with decreased stromal fibrosis and reduced presence of immunosuppressive cells within the TME152. Notably, this change in the TME was maximized by combining FAK inhibition with anti-PD-1 therapy, signifying a successful reprogramming of the TME towards immune-responsiveness. Nevertheless, a follow-up study showed that as FAK inhibition led to progressive fibroblast depletion, eventual loss of TGFβ production by the stroma conferred resistance against FAK inhibition through decreased suppression of the STAT3 signalling pathway153. As such, the importance of understanding the downstream effects of any therapeutic strategy and how these effects affect the TME cannot be overstated. At least three FAK inhibitors have been clinically tested in PDAC, one of which — defactinib — is being actively studied in PDAC in combination with the PD-1 inhibitor pembrolizumab (NCT03727880 and NCT02546531)169171.

Disrupting TGFβ signalling in the TME

TGFβ is a pleiotropic molecule that generates both tumour-promoting and antitumour effects. Although TGFβ initially suppresses epithelial cell proliferation, it promotes stromal support of cancer and immunosuppression172. With regards to immunosuppression, TGFβ induces Treg cells and directly represses several effector T cell functions172.

In patients with metastatic urothelial cancer, high levels of TGFβ predicted poor response to anti-PD-Ll therapy173. Accordingly, inhibition of TGFβ enhanced the actions of ICIs in several mouse models including the KPC model of PDAC173175. In a mouse model of liver metastatic PDAC, combining anti-TGFβ therapy with GVAX was also able to reshape the immune TME with greater infiltration of CD8+ T cells and reduction of Treg cells in association with better survival176.

Galunisertib, a small molecule inhibitor of TGFβ, has been tested in patients with unresectable PDAC. A randomized phase II trial demonstrated that galunisertib in combination with gemcitabine led to improved overall survival versus gemcitabine alone177. The combination of galunisertib and durvalumab was also investigated in patients with metastatic PDAC178. Further investigation of galunisertib has since been terminated by the sponsor179. Instead, newer generation TGFβ pathway inhibitors, such as TGFβR180 inhibitors and TGFβ-checkpoint traps181, are being developed. In addition, as blockade of the angiotensin II type I receptor leads to reduced TGFβ levels in fibroblasts182,183, the angiotensin receptor blocker losartan was tested both in preclinical models of pancreatic cancer184 and subsequently in a phase II trial in the neoadjuvant setting in combination with FOLFIRINOX and enabled 69% (30 of 49) of patients with locally advanced disease to have an R0 resection185. A randomized phase II trial assessing the effect of losartan in combination with FOLFIRINOX and stereotactic body radiation therapy again in the neoadjuvant setting is ongoing (NCT03563248). Given the multi-faceted nature of TGFβ, the clinical outcome of targeting TGFβ in PDAC is difficult to predict and might depend on how TGFβ inhibition is combined with other modalities. More studies are needed to clarify the utility of TGFβ in treating PDAC.

Conclusions

Accumulating evidence illustrates the importance of understanding the multi-faceted roles of the complex TME components in tumour suppression and progression. Future approaches should therefore prioritize integrated or convergent targets that would reprogramme the TME rather than deplete particular targets. Combination and/or multi-modal strategies that target multiple features of the TME simultaneously might also be successful (FIG. 3). Nevertheless, combination approaches must take into account the complementarity of the targeted pathways. When developing novel therapeutic strategies we should investigate whether there are TME features that are organ specific and should be considered when treating metastatic cancers (for example, differences of primary versus hepatic versus lung). In addition, we should be aware of the feedback responses that occur with any treatment and consider how they should be leveraged in a combinatorial fashion, taking into account how the preceding conventional chemotherapy and/or radiation will affect the efficacy of a therapeutic strategy of interest. Another important point to consider is how we can inform therapeutic decisions on the basis of individualized characterization of the TME in an individual patient. Given the relatively low incidence of PDACs, future trials should involve deep profiling of the TME and personalization of therapeutics when possible to accelerate progress towards more effective treatment strategies. This goal is ever closer to becoming a reality as multiplexed imaging, immunophenotyping and mutational analysis tools are increasingly high-throughput. Many trials have failed, but given the progress in our understanding of the PDAC microenvironment and the emerging strategies we have reasons to be hopeful for the future successful treatment of pancreatic cancers.

Key points.

  • Therapeutic approaches to target stromal desmoplasia, a histopathological hallmark of pancreatic ductal adenocarcinoma, have classically focused on depleting the stromal constituents; results have been generally disappointing, owing to the multi-faceted nature of tumour stroma.

  • Isolated strategies to overcome specific immune targets have also met with limited success, likely owing to the presence of multiple immunoregulatory pathways within the pancreatic ductal adenocarcinoma microenvironment.

  • In recognition of the functional complexity of the tumour microenvironment (TME), combining complementary stromal-targeted and immune-targeted treatment modalities to leverage the changes in the TME offers a more rational treatment approach.

  • Points of biological convergence, such as stromal–immune crosstalk, including glutamine metabolism, focal adhesion kinase and transforming growth factor-β signalling, are promising targets for remodelling the TME into an antitumour milieu.

Acknowledgements

W.J.H. is the recipient of an ASCO Young Investigator Award and an AACR Incyte Immuno-Oncology Research Fellowship, and is supported by NIH T32CA00971-38.

Competing interests

W.J.H. could potentially receive patent-related royalties from Rodeo Therapeutics. E.M.J. receives commercial research grants from Aduro Biotech, Amgen, Bristol–Myers Squibb, Corvus and Hertix, has ownership interest in Aduro Biotech, and is a consultant and/or advisory board member for Achilles Therapeutics, Adaptive Biotechnologies, CStone Pharmaceuticals, Dragonfly Therapeutics, Genocea and the Parker Institute for Cancer Immunotherapy. She is a member of the National Cancer Advisory Board and the Chief Medical Advisor for the Lustgarten Foundation. L.Z. receives grant support from Amgen, Bristol–Myers Squibb, Halozyme, Inxmed, iTeos, Merck and NovaRock, and received royalties for licensing GVAX to Aduro Biotech. He is a paid consultant and/or advisory board member for Akrevia, Alphamab, Biosion, Da tare vive, Found atio n Med icin e, F usun Biopharmaceutical, Mingruzhiyao, NovaRock and Sound Biologics, and holds shares in Alphamab and Mingruzhiyao.

Footnotes

Peer review information

Nature Reviews Clinical Oncology thanks I. Garrido-Laguna, A. Maitra, M. Moore and D. Ting for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

RELATED LINKS

US NIH ClinicalTrials.gov database: https://www.clinicaltrials.gov

References

  • 1.Hidalgo M Pancreatic cancer. N. Engl. J. Med 362, 1605–1617 (2010). [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD & Jemal A Cancer statistics, 2019. CA Cancer J. Clin 69, 7–34 (2019). [DOI] [PubMed] [Google Scholar]
  • 3.Von Hoff DD et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med 369, 1691–1703 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Conroy T et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med 364, 1817–1825 (2011). [DOI] [PubMed] [Google Scholar]
  • 5.Moskaluk CA, Hruban RH & Kern SE p16 and K-ras gene mutations in the intraductal precursors of human pancreatic adenocarcinoma. Cancer Res. 57, 2140–2143 (1997). [PubMed] [Google Scholar]
  • 6.DiGiuseppe JA, Redston MS, Yeo CJ, Kern SE & Hruban RH p53-independent expression of the cyclin-dependent kinase inhibitor p21 in pancreatic carcinoma. Am. J. Pathol 147, 884–888 (1995). [PMC free article] [PubMed] [Google Scholar]
  • 7.Hruban RH et al. Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am. J. Surg. Pathol 25, 579–586 (2001). [DOI] [PubMed] [Google Scholar]
  • 8.Foster DS, Jones RE, Ransom RC, Longaker MT & Norton JA The evolving relationship of wound healing and tumor stroma. JCI Insight 3, 99911 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Flier JS, Underhill LH & Dvorak HF Tumors: wounds that do not heal. N. Engl. J. Med 315, 1650–1659 (1986). [DOI] [PubMed] [Google Scholar]
  • 10.Brahmer JR et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med 366, 2455–2465 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ramanathan RK 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 37, 1062–1069 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hingorani SR 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 36, 359–366 (2018). [DOI] [PubMed] [Google Scholar]
  • 13.Whatcott CJ et al. Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin. Cancer Res 21, 3561–3568 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Vonlaufen A et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 68, 2085–2093 (2008). [DOI] [PubMed] [Google Scholar]
  • 15.Apte MV et al. Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis. Gut 44, 534–541 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Provenzano PP et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21, 418–429 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bramhall SR, Neoptolemos JP, Stamp GWH & Lemoine NR Imbalance of expression of matrix metalloproteinases (MMPs) and tissue inhibitors of the matrix metalloproteinases (TIMPs) in human pancreatic carcinoma. J. Pathol 182, 347–355 (1997). [DOI] [PubMed] [Google Scholar]
  • 18.Jones LE, Humphreys MJ, Campbell F, Neoptolemos JP & Boyd MT Comprehensive analysis of matrix metalloproteinase and tissue inhibitor expression in pancreatic cancer: increased expression of matrix metalloproteinase-7 predicts poor survival. Clin. Cancer Res 10, 2832–2845 (2004). [DOI] [PubMed] [Google Scholar]
  • 19.Matsuyama Y, Takao S & Aikou T Comparison of matrix metalloproteinase expression between primary tumors with or without liver metastasis in pancreatic and colorectal carcinomas. J. Surg. Oncol 80, 105–110 (2002). [DOI] [PubMed] [Google Scholar]
  • 20.Okada Y et al. Nerve growth factor stimulates MMP-2 expression and activity and increases invasion by human pancreatic cancer cells. Clin. Exp. Metastasis 21, 285–292 (2004). [DOI] [PubMed] [Google Scholar]
  • 21.Schnelderhan W et al. Pancreatic stellate cells are an important source of MMP-2 in human pancreatic cancer and accelerate tumor progression in a murine xenograft model and CAM assay. J. Cell Sci 120, 512–519 (2007). [DOI] [PubMed] [Google Scholar]
  • 22.Gress TM et al. Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. Int. J. Cancer 62, 407–413 (1995). [DOI] [PubMed] [Google Scholar]
  • 23.Ellenrieder V et al. Role of MT-MMPs and MMP-2 in pancreatic cancer progression. Int. J. Cancer 85, 14–20 (2000). [DOI] [PubMed] [Google Scholar]
  • 24.Yamamoto H et al. Expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases in human pancreatic adenocarcinomas: clinicopathologic and prognostic significance of matrilysin expression. J. Clin. Oncol 19, 1118–1127 (2001). [DOI] [PubMed] [Google Scholar]
  • 25.Crawford HC, Scoggins CR, Washington MK, Matrisian LM & Leach SD Matrix metalloproteinase-7 is expressed by pancreatic cancer precursors and regulates acinar-to-ductal metaplasia in exocrine pancreas. J. Clin. Invest 109, 1437–1444 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fukuda A et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell 19, 441–455 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tang Y, Kesavan P, Nakada MT & Yan L Tumor-stroma interaction: positive feedback regulation of extracellular matrix metalloproteinase inducer (EMMPRIN) expression and matrix metalloproteinase-dependent generation of soluble EMMPRIN. Mol. Cancer Res 2, 73–80 (2004). [PubMed] [Google Scholar]
  • 28.Chirvi RGS et al. Inhibition of the metastatic spread and growth of B16-BL6 murine melanoma by a synthetic matrix metalloproteinase inhibitor. Int. J. Cancer 58, 460–464 (1994). [DOI] [PubMed] [Google Scholar]
  • 29.Watson SA et al. Inhibition of organ invasion by the matrix metalloproteinase inhibitor batimastat (BB-94) in two human colon carcinoma metastasis models. Cancer Res. 55, 3629–3633 (1995). [PubMed] [Google Scholar]
  • 30.Watson SA et al. Inhibition of tumour growth by marimastat in a human xenograft model of gastric cancer: relationship with levels of circulating CEA. Br. J. Cancer 81, 19–23 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bramhall SR et al. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br. J. Cancer 87, 161–167 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bramhall SR et al. Marimastat as first-line therapy for patients with unresectable pancreatic cancer: a randomized trial. J. Clin. Oncol 19, 3447–3455 (2001). [DOI] [PubMed] [Google Scholar]
  • 33.Moore MJ et al. Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12–9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol 21, 3296–3302 (2003). [DOI] [PubMed] [Google Scholar]
  • 34.Jacobetz MA et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 62, 112–120 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.HemOnc today. Phase 3 trial of pancreatic cancer therapy misses primary endpoint https://www.healio.com/hematology-oncology/gastrointestinal-cancer/news/online/%7Be57ffed4-505b-40e5-ac9a-21f050ee850e%7D/phase-3-trial-of-pancreaticcancer-therapy-misses-primary-endpoint (2019).
  • 36.Hebrok M, Kim SK & Melton DA Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev. 12, 1705–1713 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kim SK & Melton DA Pancreas development is promoted by cyclopamine, a hedgehog signaling inhibitor. Proc. Natl Acad. Sci. USA 95, 13036–13041 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Strobel O et al. Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia. Gastroenterology 138, 1166–1177 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bailey JM et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin. Cancer Res 14, 5995–6004 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Feldmann G et al. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut 57, 1420–1430 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Olive KP et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Madden J Infinity Reports Update from Phase 2 Study of Saridegib Plus Gemcitabine in Patients with Metastatic Pancreatic Cancer. https://www.businesswire.com/news/home/20120127005146/en/Infinity-Reports-Update-Phase-2-Study-Saridegib (2012).
  • 43.Catenacci DV 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 33, 4284–4292 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.De Jesus-Acosta A et al. A phase II study of vismodegib, a hedgehog (Hh) pathway inhibitor, combined with gemcitabine and nab-paclitaxel (nab-P) in patients (pts) with untreated metastatic pancreatic ductal adenocarcinoma (PDA). J. Clin. Oncol 32, 257 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hingorani SR et al. Phase Ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin. Cancer Res. 22, 2848–2854 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rhim AD et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lee JJ et al. Stromal response to hedgehog signaling restrains pancreatic cancer progression. Proc. Natl Acad. Sci. USA 111, E3091–E3100 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Xiao Q et al. Cancer-associated fibroblasts in pancreatic cancer are reprogrammed by tumor-induced alterations in genomic DNA methylation. Cancer Res. 76, 5395–5404 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Elyada E et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Öhlund D et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med 214, 579–596 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Torphy RJ et al. Stromal content is correlated with tissue site, contrast retention, and survival in pancreatic adenocarcinoma. JCO Precis. Oncol 2018, 1–12 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Moffitt RA et al. Virtual microdissection identifies distinct tumor-and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet 47, 1168–1178 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ligorio M et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic. Cancer Cell 178, 160–175 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mahadevan KK et al. Quasimesenchymal phenotype predicts systemic metastasis in pancreatic ductal adenocarcinoma. Mod. Pathol 32, 844–854 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chen X & Song E Turning foes to friends: targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov 18, 99–115 (2019). [DOI] [PubMed] [Google Scholar]
  • 56.Hofheinz RD et al. Stromal antigen targeting by a humanised monoclonal antibody: an early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Onkologie 26, 44–48 (2003). [DOI] [PubMed] [Google Scholar]
  • 57.Gunderson AJ et al. Blockade of fibroblast activation protein in combination with radiation treatment in murine models of pancreatic adenocarcinoma. PLoS One 14, e0211117 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nugent FW et al. Phase 2 study of talabostat/gemcitabine in stage IV pancreatic cancer. J. Clin. Oncol 25, 4616 (2007).17925557 [Google Scholar]
  • 59.Özdemir BC et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ohuchida K et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 64, 3215–3222 (2004). [DOI] [PubMed] [Google Scholar]
  • 61.Lo A et al. Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Res. 75, 2800–2810 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kakarla S et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol. Ther 21, 1611–1620 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hosein AN et al. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight 4, e129212 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Biffi G et al. Il1-induced Jak/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 9, 282–301 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Blair AB et al. Dissecting the stromal signaling and regulation of myeloid cells and memory effector T cells in pancreatic cancer. Clin. Cancer Res 25, 5351–5363 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Feig C et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110, 20212–20217 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hidalgo M et al. A multi-center phase IIA trial to assess the safety and efficacy of BL-8040 (a CXCR4 inhibitor) in combination with pembrolizumab and chemotherapy in patients with metastatic pancreatic adenocarcinoma (PDAC). Ann. Oncol 30, xi33 (2019). [Google Scholar]
  • 68.Yadav D & Lowenfels AB The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 144, 1252–1261 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Guerra C et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11, 291–302 (2007). [DOI] [PubMed] [Google Scholar]
  • 70.Guerra C et al. Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence. Cancer Cell 19, 728–739 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.DuPage M et al. Endogenous T cell responses to antigens expressed in lung adenocarcinomas delay malignant tumor progression. Cancer Cell 19, 72–85 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fossum B, Olsen AC, Thorsby E & Gaudernack G CD8+ T cells from a patient with colon carcinoma, specific for a mutant p21-Ras-derived peptide (GLY13→ASP), are cytotoxic towards a carcinoma cell line harbouring the same mutation. Cancer Immunol. Immunother. 40, 165–172 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Qin H et al. CD4+ T-cell immunity to mutated ras protein in pancreatic and colon cancer patients. Cancer Res. 55, 2984–2987 (1995). [PubMed] [Google Scholar]
  • 74.Kubuschok B et al. Naturally occurring T-cell response against mutated p21 Ras oncoprotein in pancreatic cancer. Clin. Cancer Res 12, 1365–1372 (2006). [DOI] [PubMed] [Google Scholar]
  • 75.Bailey P et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016). [DOI] [PubMed] [Google Scholar]
  • 76.Danilova L 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 7, 886–895 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zehir A et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med 23, 703–713 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Alexandrov LB et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chalmers ZR et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 9, 34 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Osipov A et al. Tumor mutational burden (TMB) and response rates to immune checkpoint inhibitors (ICIs) targeting PD-1, CTLA-4, and combination. J. Clin. Oncol 37, 2578–2578 (2019). [Google Scholar]
  • 81.Yarchoan M, Hopkins A & Jaffee EM Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med 377, 2500–2501 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Salman B, Zhou D, Jaffee EM, Edil BH & Zheng L Vaccine therapy for pancreatic cancer. Oncoimmunology 2, 1–8 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bernhardt SL et al. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br. J. Cancer 95, 1474–1482 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gjertsen MK et al. Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet 346, 1399–1400 (1995). [DOI] [PubMed] [Google Scholar]
  • 85.Gjertsen MK et al. Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: clinical and immunological responses in patients with pancreatic adenocarcinoma. Int. J. Cancer 92, 441–450 (2001). [DOI] [PubMed] [Google Scholar]
  • 86.Gilliam AD et al. An international multicenter randomized controlled trial of G17DT in patients with pancreatic cancer. Pancreas 41, 374–379 (2012). [DOI] [PubMed] [Google Scholar]
  • 87.Kaufman HL et al. Poxvirus-based vaccine therapy for patients with advanced pancreatic cancer. J. Transl Med 5, (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lepisto AJ et al. A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther. 6, 955–964 (2008). [PMC free article] [PubMed] [Google Scholar]
  • 89.Laheru D et al. Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin. Cancer Res. 14, 1455–1463 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Le DT et al. A live-attenuated listeria vaccine (ANZ-100) and a live-attenuated listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction. Clin. Cancer Res. 18, 858–868 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Gatti-Mays ME et al. A phase I dose-escalation trial of BN-CV301, a recombinant poxviral vaccine targeting MUC1 and CEA with costimulatory molecules. Clin. Cancer Res 25, 4933–4944 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hu Z, Ott PA & Wu CJ Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol 18, 168–182 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kinkead HL et al. Combining STING-based neoantigen-targeted vaccine with checkpoint modulators enhances antitumor immunity in murine pancreatic cancer. JCI Insight 3, 122857 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jaffee EM et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J. Clin. Oncol 19, 145–156 (2001). [DOI] [PubMed] [Google Scholar]
  • 95.Eric L et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma: a phase II trial of safety, efficacy, and immune activation. Ann. Surg 253, 328–335 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hopkins AC et al. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight 3, 122092 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lutz ER et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol. Res 2, 616–631 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G & Bar-Sagi D Oncogenic kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 21, 836–847 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Clark CE et al. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 67, 9518–9527 (2007). [DOI] [PubMed] [Google Scholar]
  • 100.Zhao F et al. Increase in frequency of myeloid-derived suppressor cells in mice with spontaneous pancreatic carcinoma. Immunology 128, 141–149 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hiraoka N, Onozato K, Kosuge T & Hirohashi S Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin. Cancer Res. 12, 5423–5434 (2006). [DOI] [PubMed] [Google Scholar]
  • 102.Joshi NS et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity 43, 579–590 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gabrilovich DI Myeloid-derived suppressor cells. Cancer Immunol. Res 5, 3–8 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Liou GY et al. Mutant KRAS-induced expression of ICAM-1 in pancreatic acinar cells causes attraction of macrophages to expedite the formation of precancerous lesions. Cancer Discov. 5, 52–63 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Liou GY et al. Macrophage-secreted cytokines drive pancreatic acinar-to-ductal metaplasia through NF-kB and MMPs. J. Cell Biol 202, 563–577 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lesina M et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell 19, 456–469 (2011). [DOI] [PubMed] [Google Scholar]
  • 107.Liou GY et al. The presence of interleukin-13 at pancreatic ADM/PanIN lesions alters macrophage populations and mediates pancreatic tumorigenesis. Cell Rep. 19, 1322–1333 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ries CH et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 25, 846–859 (2014). [DOI] [PubMed] [Google Scholar]
  • 109.Reid MD et al. Tumor-infiltrating neutrophils in pancreatic neoplasia. Mod. Pathol 24, 1612–1619 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Shen M et al. Tumor-associated neutrophils as a new prognostic factor in cancer: a systematic review and meta-analysis. PLoS One 9, e98259 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chao T, Furth EE & Vonderheide RH CXCR2-dependent accumulation of tumor-associated neutrophils regulates T-cell immunity in pancreatic ductal adenocarcinoma. Cancer Immunol. Res 4, 968–982 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Le DT & Jaffee EM Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res. 72, 3439–3444 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kim VM et al. Anti-pancreatic tumor efficacy of a listeria-based, annexin A2-targeting immunotherapy in combination with anti-PD-1 antibodies. J. Immunother. Cancer 7, 132 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Selby MJ et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol. Res 1, 32–42 (2013). [DOI] [PubMed] [Google Scholar]
  • 115.Jang JE et al. Crosstalk between regulatory T cells and tumor-associated dendritic cells negates anti-tumor immunity in pancreatic cancer. Cell Rep. 20, 558–571 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Cannarile MA et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 5, 53 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhu Y et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 74, 5057–5069 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Saung MT et al. Targeting myeloid-inflamed tumor with anti-CSF-1R antibody expands CD137+ effector T-cells in the murine model of pancreatic cancer. J. Immunother. Cancer 6, 118 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Beatty GL et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331, 1612–1616 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Vonderheide RH CD40 agonist antibodies in cancer immunotherapy. Annu. Rev. Med 71, 47–58 (2020). [DOI] [PubMed] [Google Scholar]
  • 121.Fu J et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci. Transl Med 7, 283ra52 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Harrington KJ et al. Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol 29, viii712 (2018). [Google Scholar]
  • 123.Quaratino S et al. A first-in-human study of KY1044, a fully human anti-ICOS IgG1 antibody as monotherapy and in combination with atezolizumab in patients with selected advanced malignancies. J. Clin. Oncol 37, TPS2644–TPS2644 (2019). [Google Scholar]
  • 124.Solinas C, Gu-Trantien C & Willard-Gallo K The rationale behind targeting the ICOS-ICOS ligand costimulatory pathway in cancer immunotherapy. ESMO Open 5, e000544 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Mitchem JB et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 73, 1128–1141 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Janson C et al. Inhibition of CCR2 potentiates checkpoint inhibitor immunotherapy in murine model of pancreatic cancer [abstract 5655]. Cancer Res. 10.1158/1538-7445.am2017-5655 (2017). [DOI] [Google Scholar]
  • 127.Linehan D et al. Overall survival in a trial of orally administered CCR2 inhibitor CCX872 in locally advanced/metastatic pancreatic cancer: correlation with blood monocyte counts. J. Clin. Oncol 36, 92–92 (2018). [Google Scholar]
  • 128.Wang-Gillam A et al. Phase IB study of FOLFIRINOX plus PF-04136309 in patients with borderline resectable and locally advanced pancreatic adenocarcinoma (PC). J. Clin. Oncol 33, 338–338 (2015). [Google Scholar]
  • 129.Noel M 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 10.1007/s10637-019-00830-3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Steele CW et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell 29, 832–845 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Nywening TM et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 67, 1112–1123 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Eriksson E et al. Shaping the tumor stroma and sparking immune activation by CD40 and 4–1BB signaling induced by an armed oncolytic virus. Clin. Cancer Res. 23, 5846–5857 (2017). [DOI] [PubMed] [Google Scholar]
  • 133.Carew JS et al. Reolysin is a novel reovirus-based agent that induces endoplasmic reticular stress-mediated apoptosis in pancreatic cancer. Cell Death Dis. 4, e728 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Andtbacka RHI et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol 33, 2780–2788 (2015). [DOI] [PubMed] [Google Scholar]
  • 135.Christmas BJ et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol. Res 6, 1561–1577 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Dahlén E, Veitonmäki N & Norlén P Bispecific antibodies in cancer immunotherapy. Ther. Adv. Vaccines Immunother 6, 3–17 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Naing A et al. PEGylated IL-10 (Pegilodecakin) induces systemic immune activation, CD8+ T cell invigoration and polyclonal T cell expansion in cancer patients. Cancer Cell 34, 775–791 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Warburg O On the origin of cancer cells. Science 123, 309–314 (1956). [DOI] [PubMed] [Google Scholar]
  • 139.Altman BJ, Stine ZE & Dang CV From Krebs to clinic: glutamine metabolism to cancer therapy. Nat. Rev. Cancer 16, 619–634 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wang R et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Leone RD et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 366, 1013–1021 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Weigel PH Hyaluronan Synthase: The Mechanism of Initiation at the Reducing End and a Pendulum Model for Polysaccharide Translocation to the Cell Exterior. Int. J. Cell Biol 2015, 1–15 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Vigetti D, Viola M, Karousou E, De Luca G & Passi A Metabolic control of hyaluronan synthases. Matrix Biol. 35, 8–13 (2014). [DOI] [PubMed] [Google Scholar]
  • 144.Chanmee T et al. Hyaluronan production regulates metabolic and cancer stem-like properties of breast cancer cells via hexosamine biosynthetic pathway-coupled HIF-1 signaling. J. Biol. Chem 291, 24105–24120 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Sharma NS et al. Targeting tumor-intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti-PD1 therapy. J. Clin. Invest 130, 451–465 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wang W et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lemberg KM, Vornov JJ, Rais R & Slusher BS We’re not ‘don’ yet: optimal dosing and prodrug delivery of 6-diazo-5-oxo-L-norleucine. Mol. Cancer Therapeutics 17, 1824–1832 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Allard B, Longhi MS, Robson SC & Stagg J The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol. Rev 276, 121–144 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Loo JM et al. Extracellular metabolic energetics can promote cancer progression. Cell 160, 393–406 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kurth I et al. RGX-202, a first-in-class small-molecule inhibitor of the creatine transporter SLC6a8, is a robust suppressor of cancer growth and metastatic progression [abstract 5863]. Cancer Res. 10.1158/1538-7445.am2018-5863 (2018). [DOI] [Google Scholar]
  • 151.Blair AB et al. IDO1 inhibition potentiates vaccine-induced immunity against pancreatic adenocarcinoma. J. Clin. Invest 129, 1742–1755 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Jiang H et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med 22, 851–860 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Jiang H et al. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut 69, 122–132 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Mizuno S, Matsumoto K, Li MY & Nakamura T HGF reduces advancing lung fibrosis in mice: a potential role for MMP-dependent myofibroblast apoptosis. FASEB J. 19, 580–582 (2005). [DOI] [PubMed] [Google Scholar]
  • 155.Iekushi K et al. Hepatocyte growth factor attenuates renal fibrosis through TGF-b1 suppression by apoptosis of myofibroblasts. J. Hypertens 28, 2454–2461 (2010). [DOI] [PubMed] [Google Scholar]
  • 156.Zhao XK et al. Focal adhesion kinase regulates hepatic stellate cell activation and liver fibrosis. Sci. Rep 7, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wong VW et al. Focal adhesion kinase links mechanical force to skin fibrosis via inflammatory signaling. Nat. Med 18, 148–152 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Gates RE, King LE, Hanks SK & Nanney LB Potential role for focal adhesion kinase in migrating and proliferating keratinocytes near epidermal wounds and in culture. Cell Growth Differ. 5, 891–899 (1994). [PubMed] [Google Scholar]
  • 159.Lal H et al. Stretch-induced MAP kinase activation in cardiac myocytes: differential regulation through β1-integrin and focal adhesion kinase. J. Mol. Cell. Cardiol 43, 137–147 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kanteti R et al. Focal adhesion kinase a potential therapeutic target for pancreatic cancer and malignant pleural mesothelioma. Cancer Biol. Ther 19, 316–327 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Hochwald SN et al. A novel small molecule inhibitor of FAK decreases growth of human pancreatic cancer. Cell Cycle 8, 2435–2443 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Stokes JB et al. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol. Cancer Ther. 10, 2135–2145 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zheng D et al. A novel strategy to inhibit FAK and IGF-1R decreases growth of pancreatic cancer xenografts. Mol. Carcinog 49, 200–209 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Zhang J, He D-H, Zajac-Kaye M & Hochwald SN A small molecule FAK kinase inhibitor, GSK2256098, inhibits growth and survival of pancreatic ductal adenocarcinoma cells. Cell Cycle 13, 3143–3149 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Begum A et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS One 12, e0180181 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Vannini A et al. αvβ3-integrin regulates PD-L1 expression and is involved in cancer immune evasion. Proc. Natl Acad. Sci. USA 116, 20141–20150 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Serrels A et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell 163, 160–173 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Serrels B et al. IL-33 and ST2 mediate FAK-dependent antitumor immune evasion through transcriptional networks. Sci. Signal 10, eaan8355 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Shi Y-K, Hao XZ, Xing P, Hu B & Sun Y Phase I study of safety and pharmacokinetics for CT-707 in ALK-positive advanced non-small cell lung cancer. Ann. Oncol 28, x132 (2017). [Google Scholar]
  • 170.Mak G et al. A phase Ib dose-finding, pharmacokinetic study of the focal adhesion kinase inhibitor GSK2256098 and trametinib in patients with advanced solid tumours. Br. J. Cancer 120, 975–981 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Wang-Gillam A et al. Phase I study of defactinib combined with pembrolizumab and gemcitabine in patients with advanced cancer. J. Clin. Oncol 36, 380–380 (2018). [Google Scholar]
  • 172.Principe DR et al. TGF-β: duality of function between tumor prevention and carcinogenesis. J. Natl. Cancer Inst 106, djt369 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Mariathasan S et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Principe DR et al. TGFb blockade augments PD-1 inhibition to promote T-cell–mediated regression of pancreatic cancer. Mol. Cancer Ther 18, 613–620 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tauriello DVF et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554, 538–543 (2018). [DOI] [PubMed] [Google Scholar]
  • 176.Soares KC et al. TGF-β blockade depletes T regulatory cells from metastatic pancreatic tumors in a vaccine dependent manner. Oncotarget 6, 43005–43015 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Melisi D et al. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br. J. Cancer 119, 1208–1214 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Melisi D et al. A phase Ib dose-escalation and cohort-expansion study of safety and activity of the transforming growth factor (TGF) β receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer. J. Clin. Oncol 37, 4124–4124 (2019). [Google Scholar]
  • 179.Taylor NP Lilly puts two-thirds of midphase cancer pipeline up for sale in major shake-up of R&D priorities. FierceBiotech https://www.fiercebiotech.com/biotech/lilly-puts-two-thirds-mid-phase-cancer-pipeline-up-for-sale-major-shake-up-r-d-priorities (2017).
  • 180.Akhurst RJ Targeting TGF-β signaling for therapeutic gain. Cold Spring Harb. Perspect. Biol 9, a022301 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Ravi R et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ enhance the efficacy of cancer immunotherapy. Nat. Commun 9, 741 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y & Jain RK Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc. Natl Acad. Sci. USA 108, 2909–2914 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Cohn RD et al. Angiotensin II type 1 receptor blockade attenuates TGF-β-induced failure of muscle regeneration in multiple myopathic states. Nat. Med 13, 204–210 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Chauhan VP et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat. Commun 4, 1–11 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Murphy JE et al. Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer. JAMA Oncol. 5, 1020 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Corbett TH et al. Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Res. 44, 717–726 (1984). [PubMed] [Google Scholar]
  • 187.Bhadury J, López MD, Muralidharan SV, Nilsson LM & Nilsson JA Identification of tumorigenic and therapeutically actionable mutations in transplantable mouse tumor cells by exome sequencing. Oncogenesis 2, e44 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Raphael BJ et al. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32, 185–203 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Hingorani SR et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005). [DOI] [PubMed] [Google Scholar]
  • 190.Soares KC et al. A preclinical murine model of hepatic metastases. J. Vis. Exp 10.3791/51677 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES