Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 1.
Published in final edited form as: J Surg Oncol. 2017 Jun 12;116(1):7–15. doi: 10.1002/jso.24643

The pancreatic cancer microenvironment: A True Double Agent

Laleh Melstrom 1, Marcela D Salazar 2, Don J Diamond 3
PMCID: PMC5989710  NIHMSID: NIHMS970655  PMID: 28605029

Abstract

The tumor microenvironment in pancreatic cancer is a complex balance of pro- and anti-tumor components. The dense desmoplasia consists of immune cells, extracellular matrix, growth factors, cytokines, and cancer associated fibroblasts (CAF) or pancreatic stellate cells (PSC). There are a multitude of targets including hyaluronan, angiogenesis, focal adhesion kinase (FAK), connective tissue growth factor (CTGF), CD40, chemokine (C-X-C motif) receptor 4 (CXCR-4), immunotherapy and Vitamin D. The developing clinical therapeutics will be reviewed.

Keywords: tumor microenvironment, immunotherapy, hedgehog inhibitors, hyaluronan, vitamin D, Focal Adhesion Kinase

Introduction: Pancreas Cancer

Despite significant advances in the understanding of pancreatic cancer genetics, biology and clinical behavior, little has been accomplished in context of extending survival. The American Cancer Society estimates the incidence of pancreatic cancer to be 53,670 in 2017, with a concomitant mortality of 43,090.1 The five-year survival has been lengthened to a mere 8%.1 By 2030, the number of deaths from pancreatic cancer will surpass breast, prostate and colon cancer and become the second leading cause of death by cancer in the US2. In 2016, data from the ESPAC-4 trial was released comparing gemcitabine alone to gemcitabine plus capecitabine in the adjuvant post-resection setting, improving survival from 25.5 months to 28 months.3 Unfortunately, 80% of patients present with locally advanced (unresectable) or metastatic disease. The standard of care first line systemic therapy options for metastatic disease include gemcitabine alone, gemcitabine with nab-paclitaxel or FOLFIRINOX with a respective median survival of 6 months, 8 months and 11.1 months. The choice of regimen is dictated by the patients’ performance status.4,5 The majority of patients both treated with resection and chemotherapy or chemotherapy alone eventually succumb to the disease. There remains a great deal to be accomplished in improving outcomes.

There are inherent unique characteristics of pancreatic ductal adenocarcinoma (PDAC) that have made it particularly challenging to treat. Limitations in the management of PDAC include: 1) the lack of early detection modalities; 2) the occult nature of progression where patients at presentation have disseminated disease and 3) the presence of an exacerbated desmoplastic response that severely compromises tumor blood perfusion and drug delivery while imposing pressure for selection, further enabling the emergence of drug-resistant malignant clones69.

The desmoplastic microenvironment within the tumor is comprised of a complex dynamic milieu that both enables and contains the tumor. As more work is being done on this complex tumor-stroma relationship, the biology of PDAC is being further elucidated. The content of this review will focus on delineating these critical stromal components and how they may be utilized as therapeutic targets.

The unique microenvironment of pancreatic cancer

The microenvironment in pancreatic cancer is comprised of acellular stroma, cancer associated fibroblasts (CAF, also known as pancreas stellate cells (PSC)), immune cells and soluble factors such as cytokines, chemokines, growth and pro-angiogenic factors. The interaction of the stroma and tumor cells has best been studied extensively in the context of the molecular and cellular evolution of PDAC. The eventual development of PDAC is thought to stem from acinar ductal metaplasia (ADM), and subsequent morphologic changes of pancreatic intra-epithelial neoplasia (PanIN 1–3) that are in part regulated by oncogenic inflammation.1012 The mutational changes in this progression include the presence of an activating mutation in the K-ras-oncogene as a critical event in greater than 90% of PDAC.13 Additional subsequent somatic mutations in tumor suppressor genes include p16, p53, and DPC/SMAD4 which cumulatively lead to progressive epithelial transformation from PanIN1-3 and ultimately PDAC capable of metastasizing to lymph nodes and distant organs.1416 Along this continuum there is progressive evolution of the tumor microenvironment accompanied by an intense desmoplasmic response characterized by deposition of extracellular matrix proteins and proteases accounting for up to 80% of tumor volume9,1719

Cancer Associated Fibroblasts (CAF)/Pancreatic Stellate Cells (PSC)

In the normal pancreas, there exist resident fibroblasts and pancreatic stellate cells that function to maintain normal gland connective tissue architecture. In response to injury or tissue damage (e.g. chronic pancreatitis) there is local production of inflammatory cytokines and pro-angiogenic growth factors that increases recruitment of both adaptive and innate immune cells, promoting angiogenesis and PSC/CAF mediated-deposition of extracellular matrix (ECM).20,21 In the context of carcinogenesis, PSCs/CAFs acquire a myofibroblast-like phenotype accompanied by excessive secretion of ECM proteins including laminins, fibronectins, and collagens.21,22 In vitro and in vivo studies have demonstrated that PSC transformation is induced by signaling through growth factors and cytokines such as PEDF (pigment epithelium derived factor), ET-1 (endothelin 1), IGF-1 (insulin-like growth factor), TFF2 (trefoil factor 1) and PDGF (platelet derived growth factor).2328 Subsequently, intense signaling crosstalk between tumor cells and transformed α-SMA (alpha smooth muscle actin)-expressing PSCs leads to enhanced tumor cell proliferation and apoptosis resistance, facilitating migration and invasion of malignant cells.29,30

In a somewhat reciprocal fashion, PSCs supports PDAC cell proliferation via secretion of TGFβ (transforming growth factor beta), FGF2 (fibroblast growth factor 2), PDGF, epidermal growth factor (EGF), connective tissue growth factor (CTGF), AM (adrenomedullin) and Galectin-1. 29,3133 PSCs also stimulate invasion and migration of PDAC cells by the production of SDF-1 (stroma cell-derived factor-1), SPARC (secreted protein acidic and rich in cysteine), MMPs (matrix metalloproteinases).29,3436 PSCs contribute to PDAC chemoresistance in part via release of NO (nitric oxide), IL-1β (interleukin-1β), periostin and type 1 collagen signaling.23,3739

With the activation of PSCs, there is this perpetual propagation of fibrosis, which is a major limiting factor in the delivery of therapeutics and will be further addressed as a therapeutic target below.20 In addition, PSCs also control ECM turnover via production of tissue inhibitor proteases.40 Excessive deposition of ECM components leads to distortion of normal parenchyma, vascular compression and subsequent hypoxia. This hypoxia induces a host of changes including activation of PSCs leading to not only further ECM deposition and but neoangiogenesis as well.41

Tumor immune infiltrate

Changes in the composition of the immune infiltrate occur alongside the increase in mutational burden and progressive fibrosis during the transition from normal pancreas to pancreatic cancer.42 A small proportion of patients with chronic pancreatitis (4-6%), where there is persistent inflammation involving continuous activation of tissue damage and repair responses, will ultimately develop PDAC anywhere from 10–20 years from disease onset.43 Inflammatory mediators such as interleukin 1b (IL-1b), IL-6, and TNF released by altered acinar cells promote chronic inflammation due to persistent activation of an immune response.44 Chronic inflammation leads to profound changes in transcriptional programs in pre-neoplastic cells and quiescent PSCs within the injured tissue niche, ultimately promoting transformation into malignant phenotypes and myofribroblast-like cells, respectively.4547 Chronic inflammation also leads to a significant shift in the immune cell infiltrate towards an immunosuppressive phenotype, further facilitating immune evasion of transformed cells and tumor establishment/progression.48 Specifically, there is a significant increase in the tumor-infiltrating population of tumor-associated macrophages (TAMs), with an M2 phenotype, neutrophils with an N2 phenotype, regulatory T cells (Treg) cells as well as a Th1/Th2 cell shift.49

Tumor-Associated Macrophages (TAM)

TAMs reside within the TME in PDAC and are thought to serve as a potential therapeutic target in several solid tumors.50 There was a recent Phase II trial in PDAC targeting TAMs with the cytotoxic agent trabectedin (NCT01339754).45,51 Trabectedin targets mononuclear phagocytes selectively via caspase-8 dependent apoptosis and via differential expression of signaling and decoy TRAIL receptors in macrophages. TAMs have also had a role in PDAC progression via production of IL-6 and activation of the STAT3 pathway.10,52 Upon recruitment and activation, TAMS release IL-6 and thus activate the STAT3/SOCS pathway. This accelerates PDAC generation.

Myeloid cell infiltration and Polymorphonuclear neutrophils (PMN)

There is mounting evidence that infiltrating immune cells in the PDAC microenvironment are pro-tumorigenic.53 These cells are found early in the development of PDAC and can be found surrounding early PanIN I lesions.42 Local tumor derived production of granulocyte macrophage colony-stimulating factor (GM-CSF) recruits myeloid progenitor cells and induces subsequent differentiation of these into myeloid-derived suppressor cells (MDSCs).54 MDSCs in turn function to mitigate the CD8+ killer T cell immune surveillance functions. This prevents tumor-specific cytotoxic T cells from clearing transformed pancreatic ductal cells.55 Targeted depletion of granulocytic MDSC (Gr-MDSC) activates an endogenous antitumor T cell response indicating perhaps Gr-MDSC may be a promising therapeutic target in PDAC.56

In PDAC, the presence of TANs is associated with poor clinical outcome.57 PMN infiltrates can be observed near tumor cells or in the stroma and are associated with poor differentiation.58 Neutrophils in PDAC have been further characterized in solid tumors as anti-tumor N1 TAN (Tumor Associated Neutrophils) or pro-tumor N2 TANs.59 This polarization or phenotype has been attributed to the local concentration of TGF-β, whereby pro tumor N2 TANS are associated with higher concentrations of this cytokine.60,61 The release of neutrophil serine proteases (e.g. cathepsisn), metalloproteases (e.g. MMP8 and 9) and reactive oxygen species (ROS) also greatly contributes to this immunosuppressive milieu by promoting tumor matrix remodeling and cell mobility.62,63 Current strategies to target tumor-infiltrating neutrophils and MDSCs in pancreatic cancer in the preclinical setting involve the use of CXCR2 (C-X-C motif chemokine receptor 2, also known as interleukin-8 receptor beta) inhibitors or RNA interference against the immunosuppressive molecule indoleamine 2,3-dioxygenase 1 (IDO).6466 Overexpression of CXCR2 and IDO are independent markers of poor prognosis in pancreatic cancer, playing key roles in supporting immune evasion and establishment of metastatic processes.6770

Tumor infiltrating T cells

Upon examination of established PDAC there is an abundance of CD4+ T cells and a paucity of CD8+ T cells in the TME.42 Recent work has demonstrated that CD4+ T cells function to promote PanIN formation by intercepting antitumor immune responses by CD8+ T cells.71,72 The ratio for GATA3+ (Th2)/Tbet+ (Th1) tumor infiltrating lymphocytes was an independent predictive marker for survival after surgery in patients with (stage IIB/III) pancreatic cancer.73,74 T-cell-dependent antitumor immunity is thought to be in part via activation of the TNF receptor superfamily member CD40. Activation of CD40 via an agonist CD40 monoclonal antibody was adequate to overcome tumor induced immune suppression and induce macrophage and T cell dependent antitumor immunity in pancreatic cancer both in mice and in humans.75 CD4+ T cells can be categorized as regulatory T cells (Treg) or T helper 17 (TH17), and both were found in the PDAC TME in a Kras dependent fashion. CD4+ T cells and IL-17 are required for oncogenic Kras-driven pancreas cancer development. During Kras-driven PanIN development, CD4+ T cells suppress the antitumor activity of CD8+ T cells via secretion of IL-17.75 Additionally TH17 cells secrete IL-17A which via IL-17RA induces acinar-ductal metaplasia and PanINs, thus inducing tumor initiation and progression.72 CD4+T cells and their cytokine signaling has been an additional target of PDAC however, this will not be further addressed in this review.

Clinical targets: success and failure

Hyaluronan (HA)

HA is a glycosaminoglycan that is found in abundance in the PDAC TME. The fibrotic collagens and HA contribute to the ECM component of PDAC thus creating the desmoplasia that is so characteristic of these tumors.9 The desmoplasia and elevated interstitial fluid pressure (IFP) contribute to the barriers in the delivery of systemic therapies to this tumor by compressing/collapsing the tumor blood vessels.76 HA contributes to this IFP and functions as a core polymer whereby multiple hydrophilic matrix proteoglycans are formed. This allows for a gel like matrix that supports tumor growth and retains growth factors and cytokines.77

Functionally HA also binds to the CD44 and RHAMM cell surface receptors. This activates signaling pathways that contribute to cell invasion, migration, proliferation, adhesion and survival.78 Based on these roles, HA has been identified as a target to enhance delivery of therapeutics.79,80 PEGPH20 is a PEGylated recombinant hyaluronidase that degrades HA and has been found in both animal and human studies to transiently enhance penetration of systemic chemotherapy. In a Phase I/II clinical trial, gemcitabine when combined with PEGPH20 improved both overall survival and progression-free survival in advanced PDAC (NCT01453153). These findings were even more pronounced with patients that had high levels of HA expression. The limitations of this therapy include thromboembolic events and therefore anticoagulation is now administered in conjunction with PEGPH20 in active clinical trials (combination of PEGPH20/ gemcitabine/nab-paclitaxel-NCT02715804 and PEGPH20 and FOLFIRINOX-NCT01959139).

Hedgehog inhibitors

Targeting the stromal components has not always been a success. The Hedgehog pathway is hyperactivated in PDAC and is known to stimulate PSC’s to regulate stromal production.81 PDAC cells produce Hh ligand which binds to its receptor Patched2 located on PSCs. This results in signaling that removes the inhibitory effects of smoothened (Smo) and allows for Gli1 transcription factor translocation into the nucleus and the production of a host of genes including ECM proteins.82 There have been several strategies to interrupt Hh signaling and thereby create stromal depletion. IPI-926 is a small molecule inhibitor that inhibits the Hh pathway (Smo inhibitor) and in pre-clinical studies was shown to improve gemcitabine perfusion and survival.81 Unfortunately, clinical trials targeting this pathway were subject to early termination secondary to progression. There have been several proposed explanations for this lack of success. First, as predicted, depletion of the pro-stromal Shh pathway leads to fewer myofibroblasts and tumor stroma. However, there were more frequent ADM and PanIN lesions during early carcinogenesis and established tumors were undifferentiated, more likely to metastasize and with a higher proliferative index compared to littermates.83 Second, there is increased tumor vasculature and reduced autophagy.45 Lastly it has been postulated that with regards to gemcitabine chemotherapy, there is a transporter hENT1 and an activating enzyme deoxycytidine kinase that are associated with improved survival.84,85 This suggests that gemcitabine metabolism rather than biophysical delivery may matter more. Perhaps the stroma is contributing to some tumor control and therefore permanent stroma depletion leads to more aggressive disease.

This explanation has yet to be clearly elucidated but the disappointing findings of trials targeting the Hh pathway indicate that clearly the stroma may have a tumor insulating role.

Connective Tissue Growth Factor (CTGF)

CTGF is a pro-fibrotic mediator that is found in abundance in the stroma of PDAC and is the target of an ongoing trial (NCT02606136).86 Over expression of CTGF is regulated by chemokine signaling through CXC proteins which bind integrins and growth factors to promote fibrosis and collagen deposition, and subsequently cancer progression and metastasis.87,88 In preclinical models the monoclonal antibody FG-3019 resulted in higher rates of tumor cell apoptosis when combined with gemcitabine. Interestingly, the gross appearance of the cellular and acellular tumor stroma was unchanged. In contrast, there was significant molecular down regulation of pro-survival and anti-apoptotic proteins such as hypoxia inducible factor-1alpha (HIF1-α), Psen1, Ubqln2, Birc6, X-linked inhibitor of apoptosis (XIAP).45 In a recent Phase I trial, the human monoclonal antibody against CTGF (FG-3019) was tested with gemcitabine and erlotinib in Stage 3 or 4 pancreatic adenocarcinoma. Results were that with a high exposure (at least 15 days of treatment) and low baseline CTGF plasma levels there was improved PFS and OS. Currently there is an ongoing Phase I/II trial of gemcitabine and nab-paclitaxel with our without FG-3019 (pamrevilumab) in the neoadjuvant setting for locally advanced pancreatic cancer (NCT02210559).

Angiotensin Inhibitors

There is currently a clinical trial investigating the combination of losartan (angiotensin inhibitor) and FOLFIRINOX assessing if this inhibitor sensitizes pancreatic cancer to standard chemotherapy (NCT01821729). Angiotensin II may play a role in promoting tumor desmoplasia. Ang II is thought to promote PSC activation via inducing the protein kinase C and EGF-ERK pathways.89,90 However, there are some conflicting data whereby in mice with AT2 receptor deficiency in pancreatic fibroblasts, tumors grow significantly larger than controls in a syngeneic orthotopic model.91 Additionally, there are studies demonstrating that angiotensin inhibitors have stromal effects by targeting TGF-β signaling.92,93

Nearly 55% of PDACs have the tumor suppressor gene SMAD4 (DPC4) inactivated.94 SMAD4 is the primary transcriptional mediator of TGF-β effects on cell proliferation and morphology. Loss of SMAD4 leads to not only the loss of a tumor suppressor and but also no mitigation of TGF-β ligand abundance. This contributes to desmoplasia due to the downstream effects of TGF-β over- expression.9597

Pirfenidone

Similar to the downstream impact of angiotensin inhibitors, pirfenidone is a compound that mitigates the expression several mediators of fibrosis such as TGF-β and collagen. Treatment with pirfenidone inhibited PSC growth, migration, invasion, and ECM production. In preclinical studies pirfenidone has demonstrated some promising effects in combination with gemcitabine, with β-cyclodextrin MMP2 responsive liposome and with the antioxidant N-acetyl cysteine.98100 This compound has yet to be evaluated in human trials.

Angiogenesis

In the PDAC TME there is angiogenesis in response to chronic hypoxia, however the vessels appear compressed secondary in part to the abundance of extracellular matrix proteins. There has been work in designing a hypoxia-activated prodrug TH-302 that improved progression free survival of mice and is currently in Phase II/III clinical trials in advanced PDAC patients (NCT-01746979). Additionally the most well studied inhibitor of angiogenesis (Bevacizumuab; anti-VEGF antibody) when studied in pancreatic cancer patients did not translate into significant benefits in survival.101 Unlike its common use in metastatic colorectal cancer, bevacizumab has not materialized as the standard of care for PDAC.

Additional targets of angiogenesis in pancreas cancer have been the hepatocyte growth factor (HGF)-c-MET pathway. In the TME, PSC’s secrete HGF which binds to cMET on pancreatic cancer cells. This binding leads to cMET receptor phosphorylation and downstream signaling, inducing PDAC cell growth and migration. Blockage of HGF has been found to decrease angiogenesis. This may be a target of future investigation.

Immunotherapy and the Tumor Microenvironment

It is clear that the complex interaction between pancreatic cancer cells and the tumor microenvironment is heavily dependent on interactions with immune cell populations. Unlike some other solid tumors, such as lung cancer, melanoma and renal cell carcinoma, pancreatic cancer has not been able to gain advances in progression-free and overall survival with current immunotherapy even though following immunophenotyping of clinical specimens, tumors appear to be amenable to immunotherapy.102,103 However, the interdependent relationship between inflammation and tumor progression remains an area of intense interest and study.

There is progressive evidence that PSCs/CAFs not only modulate the stromal environment in PDAC but also play a significant role in mediating local immunosuppression and thus facilitate tumor progression and metastases.45,104107 The chemokine ligand CXCL12 is secreted by CAFs and binds to PDAC cells and leads to the depletion of CD8+ T cells. Depletion of CAFs that express fibroblast activation protein (FAP) and delivery of a CXCL-12 competitive blocker increased the accumulation of cytotoxic T cells in the TME and diminished PDAC tumor growth.108 Additional work evaluating a CXCR4 inhibitor (CXCR4 is the receptor for CXCL12 ligand) plerixafor induced a rapid T cell response and acted in synergy with PD-L1 antibody to inhibit tumor growth in KPC mice.108 This approach in combining a CXCR4 inhibitor with an immune checkpoint inhibitor is a current clinical trial (NCT02301130).

CD40 is an additional target that has been evaluated as it may have a role in T cell dependent and independent immune mediated mechanisms of tumor regression. CD40 is cell surface molecule and as a member of the TNF receptor family, partakes in immune regulation, and tumor apoptosis.109 CD40 was initially thought to be crucial in T cell dependent anti-tumor immunity, however, more recently it is thought that CD40 mediated tumor regression is more dependent on macrophages that facilitate stroma depletion. In a phase I trial, patients with advanced PDAC were treated with a monoclonal antibody that is a CD40 agonist (CP-870).110 Of the 22 patients, four (18%) achieved a partial response and in those tumors, there was a significant population of macrophages and minimal tumor infiltrating lymphocytes.

Both programmed cell death-1 receptor (PD-1) and PD-1 ligand expression are associated with poor prognosis. The expression of these, suppresses T cell activity and allows for immune escape in the TME. However, early clinical trials targeting either PD-1 or PD-L1 did not demonstrate a significant response as monotherapy for PDAC.111,112 There are currently several trials underway utilizing combination therapies with targeted therapies to PD-1 or PD-1 ligands.

Secreted protein acid and rich in cysteine (SPARC)

SPARC is over expressed in the PDAC TME and is associated with promoting tumor invasion and inhibiting angiogenesis.113 An additional strategy to better deliver therapeutics to PDAC is to modify chemotherapeutic agents such that the affinity for endogenous stromal receptors is increased. With this in mind, paclitaxel was conjugated with albumin-nanoparticles (nab-paclitaxel) with the hopes of better efficacy via albumin mediated absorption and binding by SPARC. However, the levels of SPARC expression did not appear to correlate with response to treatment.114,115 Additionally, there is work citing that the efficacy of nab-paclitaxel is actually dependent upon drug internalization by TAMs via macropinocytosis.116 Therefore, targeting the optimization of macropinocytosis may be an additional option to improving nab-paclitaxel efficacy. A Phase 3 clinical trial demonstrated a modest increase in overall survival when combined with gemcitabine. At present, the combination of gemcitabine and nab-paclitaxel is one of 2 regimens deemed as standard of care for metastatic PDAC.4

Vitamin D

Vitamin D has been more recently recognized as a stromal reprogramming agent. Activation of the vitamin D receptor (VDR) has been shown to induce PSCs into engaging in more dormant and less pro tumorigenic behavior.117 VDR activation decreases fibrosis and pro-inflammatory markers and increases intratumoral gemcitabine levels.118 Treatment with all-trans retinoic acid (ATRA) induced quiescence in CAFs leading to decreased proliferation, invasion and increased apoptosis. Vitamin D and several of its analogs have significant anti-inflammatory properties. Genetically engineered mouse models (GEMMS) of PDAC were treated with calcipotriol (Cal), a vitamin D analog in combination with gemcitabine. This treatment again induced stroma with less inflammation, enhanced drug delivery and improved survival of mice in a preclinical model.117 There are currently clinical trials in PDAC forthcoming utilizing vitamin D analogs as a stromal remodeling mechanism (NCT02930902, NCT02030860).

Focal Adhesion Kinase (FAK)

FAKs are non-receptor tyrosine kinases which include FAK1 and PYSK2 (FAK2). FAK1 has been implicated in cancer migration, proliferation and survival.119,120 Jiang et al demonstrated hyperactivated FAK activity in neoplastic PDAC cells as an important regulator of the fibrotic and immunosuppressive TME.121 FAK activity was elevated in human PDAC tissues and correlates with high levels of fibrosis and poor CD8+ cytotoxic T cell infiltration. Single agent FAK inhibition with VS-4718 limited tumor progress in a KPC mouse model of human PDAC.121 This correlated with reduced tumor fibrosis and decreased numbers of tumor infiltrating immunosuppressive cells. Additionally, the combination of FAK inhibition and PD-1 antagonists allowed for improved immune surveillance and a marked increase in survival in this preclinical model. Currently the clinical trial for VS-4718 is on hold (NCT02758587). However there are at least 2 other trials that are active targeting with FAK inhibition for PDAC (NCT02428270 and NCT025465310).

Conclusions

At present there are clearly multiple targets in PDAC. However, it has become clear that the TME plays a crucial role in both tumor containment and progression. The proper combinatorial approach in mitigating tumor desmoplasia and optimizing the systemic delivery of anti-cancer therapy to diminish the local tumor mediated immunosuppression will lead to success in this challenging disease.

Figure 1.

Figure 1

With progressive acquisition of somatic mutations, the TME in PDAC changes in a number of ways as summarized in part above.

Table 1.

Selected Clinical Trials Targeting PDAC Stroma

TARGET STRATEGY TRIAL
Hyaluronan PEGylated hyaluronidase NCT02715804
NCT01959139
Connective Tissue Growth Factor Monoclonal Ab to CTGF (pro-fibrotic mediator) NCT02606136
NCT02210559
Angiotensin Angiotensin Inhibitor losartan via preventing PKC and EGF-ERK pathway activation NCT01821729
Angiogenesis Hypoxia activated prodrug TH-302 NCT01746979
CXCR4 inhibitor Combination of CXCR4 inhibition AND checkpoint inhibition NCT02301130
Vitamin D analogs Vitamin D receptor activation decreases fibrosis NCT02930902
NCT02030860
Focal Adhesion Kinase (FAK) inhibition FAK implicated in cancer migration, proliferation and survival NCT02758587
NCT02428270
NCT025465310

Synopsis.

The tumor microenvironment in pancreatic cancer is thought to contribute to both carcinogenesis, tumor progression and immune evasion. Herein, we review the important components of the microenvironment and potential targets to overcome in treating this challenging disease.

Footnotes

Disclosures: none

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA: a cancer journal for clinicians. 2017;67:7–30. doi: 10.3322/caac.21387. [DOI] [PubMed] [Google Scholar]
  • 2.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–21. doi: 10.1158/0008-5472.CAN-14-0155. [DOI] [PubMed] [Google Scholar]
  • 3.Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017 doi: 10.1016/S0140-6736(16)32409-6. [DOI] [PubMed] [Google Scholar]
  • 4.Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. The New England journal of medicine. 2013;369:1691–703. doi: 10.1056/NEJMoa1304369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England journal of medicine. 2011;364:1817–25. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
  • 6.Hidalgo M, Cascinu S, Kleeff J, et al. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology. 2015;15:8–18. doi: 10.1016/j.pan.2014.10.001. [DOI] [PubMed] [Google Scholar]
  • 7.Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–13. doi: 10.1038/nature10762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–7. doi: 10.1038/nature09515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Whatcott CJ, Diep CH, Jiang P, et al. Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clin Cancer Res. 2015;21:3561–8. doi: 10.1158/1078-0432.CCR-14-1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fukuda A, Wang SC, Morris JPt, et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell. 2011;19:441–55. doi: 10.1016/j.ccr.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6:2969–72. [PubMed] [Google Scholar]
  • 12.Maniati E, Bossard M, Cook N, et al. Crosstalk between the canonical NF-kappaB and Notch signaling pathways inhibits Ppargamma expression and promotes pancreatic cancer progression in mice. J Clin Invest. 2011;121:4685–99. doi: 10.1172/JCI45797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–54. doi: 10.1016/0092-8674(88)90571-5. [DOI] [PubMed] [Google Scholar]
  • 14.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–3. doi: 10.1126/science.271.5247.350. [DOI] [PubMed] [Google Scholar]
  • 15.Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes & development. 2006;20:1218–49. doi: 10.1101/gad.1415606. [DOI] [PubMed] [Google Scholar]
  • 16.Pellegata NS, Sessa F, Renault B, et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res. 1994;54:1556–60. [PubMed] [Google Scholar]
  • 17.Yen TW, Aardal NP, Bronner MP, et al. Myofibroblasts are responsible for the desmoplastic reaction surrounding human pancreatic carcinomas. Surgery. 2002;131:129–34. doi: 10.1067/msy.2002.119192. [DOI] [PubMed] [Google Scholar]
  • 18.Gress TM, Muller-Pillasch F, Lerch MM, Friess H, Buchler M, Adler G. Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. Int J Cancer. 1995;62:407–13. doi: 10.1002/ijc.2910620409. [DOI] [PubMed] [Google Scholar]
  • 19.Erkan M, Michalski CW, Rieder S, et al. The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2008;6:1155–61. doi: 10.1016/j.cgh.2008.05.006. [DOI] [PubMed] [Google Scholar]
  • 20.Bachem MG, Schneider E, Gross H, et al. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115:421–32. doi: 10.1016/s0016-5085(98)70209-4. [DOI] [PubMed] [Google Scholar]
  • 21.Apte MV, Haber PS, Darby SJ, et al. Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis. Gut. 1999;44:534–41. doi: 10.1136/gut.44.4.534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Xu Z, Pothula SP, Wilson JS, Apte MV. Pancreatic cancer and its stroma: a conspiracy theory. World journal of gastroenterology. 2014;20:11216–29. doi: 10.3748/wjg.v20.i32.11216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Haqq J, Howells LM, Garcea G, Metcalfe MS, Steward WP, Dennison AR. Pancreatic stellate cells and pancreas cancer: current perspectives and future strategies. European journal of cancer. 2014;50:2570–82. doi: 10.1016/j.ejca.2014.06.021. [DOI] [PubMed] [Google Scholar]
  • 24.Samkharadze T, Erkan M, Reiser-Erkan C, et al. Pigment epithelium-derived factor associates with neuropathy and fibrosis in pancreatic cancer. The American journal of gastroenterology. 2011;106:968–80. doi: 10.1038/ajg.2010.479. [DOI] [PubMed] [Google Scholar]
  • 25.Klonowski-Stumpe H, Reinehr R, Fischer R, Warskulat U, Luthen R, Haussinger D. Production and effects of endothelin-1 in rat pancreatic stellate cells. Pancreas. 2003;27:67–74. doi: 10.1097/00006676-200307000-00010. [DOI] [PubMed] [Google Scholar]
  • 26.Schneider E, Schmid-Kotsas A, Zhao J, et al. Identification of mediators stimulating proliferation and matrix synthesis of rat pancreatic stellate cells. American journal of physiology Cell physiology. 2001;281:C532–43. doi: 10.1152/ajpcell.2001.281.2.C532. [DOI] [PubMed] [Google Scholar]
  • 27.Luttenberger T, Schmid-Kotsas A, Menke A, et al. Platelet-derived growth factors stimulate proliferation and extracellular matrix synthesis of pancreatic stellate cells: implications in pathogenesis of pancreas fibrosis. Laboratory investigation; a journal of technical methods and pathology. 2000;80:47–55. doi: 10.1038/labinvest.3780007. [DOI] [PubMed] [Google Scholar]
  • 28.Arumugam T, Brandt W, Ramachandran V, et al. Trefoil factor 1 stimulates both pancreatic cancer and stellate cells and increases metastasis. Pancreas. 2011;40:815–22. doi: 10.1097/MPA.0b013e31821f6927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Apte MV, Park S, Phillips PA, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004;29:179–87. doi: 10.1097/00006676-200410000-00002. [DOI] [PubMed] [Google Scholar]
  • 30.Vonlaufen A, Joshi S, Qu C, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68:2085–93. doi: 10.1158/0008-5472.CAN-07-2477. [DOI] [PubMed] [Google Scholar]
  • 31.Shek FW, Benyon RC, Walker FM, et al. Expression of transforming growth factor-beta 1 by pancreatic stellate cells and its implications for matrix secretion and turnover in chronic pancreatitis. The American journal of pathology. 2002;160:1787–98. doi: 10.1016/s0002-9440(10)61125-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tang D, Yuan Z, Xue X, et al. High expression of Galectin-1 in pancreatic stellate cells plays a role in the development and maintenance of an immunosuppressive microenvironment in pancreatic cancer. Int J Cancer. 2012;130:2337–48. doi: 10.1002/ijc.26290. [DOI] [PubMed] [Google Scholar]
  • 33.Ramachandran V, Arumugam T, Langley R, et al. The ADMR receptor mediates the effects of adrenomedullin on pancreatic cancer cells and on cells of the tumor microenvironment. PloS one. 2009;4:e7502. doi: 10.1371/journal.pone.0007502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Koshiba T, Hosotani R, Miyamoto Y, et al. Expression of stromal cell-derived factor 1 and CXCR4 ligand receptor system in pancreatic cancer: a possible role for tumor progression. Clin Cancer Res. 2000;6:3530–5. [PubMed] [Google Scholar]
  • 35.Mantoni TS, Schendel RR, Rodel F, et al. Stromal SPARC expression and patient survival after chemoradiation for non-resectable pancreatic adenocarcinoma. Cancer biology & therapy. 2008;7:1806–15. doi: 10.4161/cbt.7.11.6846. [DOI] [PubMed] [Google Scholar]
  • 36.Schneiderhan W, Diaz F, Fundel M, 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. Journal of cell science. 2007;120:512–9. doi: 10.1242/jcs.03347. [DOI] [PubMed] [Google Scholar]
  • 37.Muerkoster S, Wegehenkel K, Arlt A, et al. Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin-1beta. Cancer Res. 2004;64:1331–7. doi: 10.1158/0008-5472.can-03-1860. [DOI] [PubMed] [Google Scholar]
  • 38.Erkan M, Kleeff J, Gorbachevski A, et al. Periostin creates a tumor-supportive microenvironment in the pancreas by sustaining fibrogenic stellate cell activity. Gastroenterology. 2007;132:1447–64. doi: 10.1053/j.gastro.2007.01.031. [DOI] [PubMed] [Google Scholar]
  • 39.Armstrong T, Packham G, Murphy LB, et al. Type I collagen promotes the malignant phenotype of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2004;10:7427–37. doi: 10.1158/1078-0432.CCR-03-0825. [DOI] [PubMed] [Google Scholar]
  • 40.Wehr AY, Furth EE, Sangar V, Blair IA, Yu KH. Analysis of the human pancreatic stellate cell secreted proteome. Pancreas. 2011;40:557–66. doi: 10.1097/MPA.0b013e318214efaf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Masamune A, Kikuta K, Watanabe T, Satoh K, Hirota M, Shimosegawa T. Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer. American journal of physiology Gastrointestinal and liver physiology. 2008;295:G709–17. doi: 10.1152/ajpgi.90356.2008. [DOI] [PubMed] [Google Scholar]
  • 42.Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007;67:9518–27. doi: 10.1158/0008-5472.CAN-07-0175. [DOI] [PubMed] [Google Scholar]
  • 43.Bang UC, Benfield T, Hyldstrup L, Bendtsen F, Beck Jensen JE. Mortality, cancer, and comorbidities associated with chronic pancreatitis: a Danish nationwide matched-cohort study. Gastroenterology. 2014;146:989–94. doi: 10.1053/j.gastro.2013.12.033. [DOI] [PubMed] [Google Scholar]
  • 44.Weber CK, Adler G. From acinar cell damage to systemic inflammatory response: current concepts in pancreatitis. Pancreatology. 2001;1:356–62. doi: 10.1159/000055834. [DOI] [PubMed] [Google Scholar]
  • 45.Neesse A, Algul H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–84. doi: 10.1136/gutjnl-2015-309304. [DOI] [PubMed] [Google Scholar]
  • 46.Farrow B, Sugiyama Y, Chen A, Uffort E, Nealon W, Mark Evers B. Inflammatory mechanisms contributing to pancreatic cancer development. Annals of surgery. 2004;239:763–9. doi: 10.1097/01.sla.0000128681.76786.07. discussion 9-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Andoh A, Takaya H, Saotome T, et al. Cytokine regulation of chemokine (IL-8, MCP-1, and RANTES) gene expression in human pancreatic periacinar myofibroblasts. Gastroenterology. 2000;119:211–9. doi: 10.1053/gast.2000.8538. [DOI] [PubMed] [Google Scholar]
  • 48.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
  • 49.Protti MP, De Monte L. Immune infiltrates as predictive markers of survival in pancreatic cancer patients. Frontiers in physiology. 2013;4:210. doi: 10.3389/fphys.2013.00210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Georgoudaki AM, Prokopec KE, Boura VF, et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell reports. 2016;15:2000–11. doi: 10.1016/j.celrep.2016.04.084. [DOI] [PubMed] [Google Scholar]
  • 51.Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell. 2013;23:249–62. doi: 10.1016/j.ccr.2013.01.008. [DOI] [PubMed] [Google Scholar]
  • 52.Lesina M, Kurkowski MU, Ludes K, et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19:456–69. doi: 10.1016/j.ccr.2011.03.009. [DOI] [PubMed] [Google Scholar]
  • 53.Wormann SM, Diakopoulos KN, Lesina M, Algul H. The immune network in pancreatic cancer development and progression. Oncogene. 2014;33:2956–67. doi: 10.1038/onc.2013.257. [DOI] [PubMed] [Google Scholar]
  • 54.Bayne LJ, Beatty GL, Jhala N, et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012;21:822–35. doi: 10.1016/j.ccr.2012.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010;70:68–77. doi: 10.1158/0008-5472.CAN-09-2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stromnes IM, Brockenbrough JS, Izeradjene K, et al. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut. 2014;63:1769–81. doi: 10.1136/gutjnl-2013-306271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gregory AD, Houghton AM. Tumor-associated neutrophils: new targets for cancer therapy. Cancer Res. 2011;71:2411–6. doi: 10.1158/0008-5472.CAN-10-2583. [DOI] [PubMed] [Google Scholar]
  • 58.Reid MD, Basturk O, Thirabanjasak D, et al. Tumor-infiltrating neutrophils in pancreatic neoplasia. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2011;24:1612–9. doi: 10.1038/modpathol.2011.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Felix K, Gaida MM. Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression. International journal of biological sciences. 2016;12:302–13. doi: 10.7150/ijbs.14996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Piccard H, Muschel RJ, Opdenakker G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Critical reviews in oncology/hematology. 2012;82:296–309. doi: 10.1016/j.critrevonc.2011.06.004. [DOI] [PubMed] [Google Scholar]
  • 61.Fridlender ZG, Sun J, Kim S, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16:183–94. doi: 10.1016/j.ccr.2009.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tazzyman S, Lewis CE, Murdoch C. Neutrophils: key mediators of tumour angiogenesis. International journal of experimental pathology. 2009;90:222–31. doi: 10.1111/j.1365-2613.2009.00641.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.De Larco JE, Wuertz BR, Furcht LT. The potential role of neutrophils in promoting the metastatic phenotype of tumors releasing interleukin-8. Clin Cancer Res. 2004;10:4895–900. doi: 10.1158/1078-0432.CCR-03-0760. [DOI] [PubMed] [Google Scholar]
  • 64.Steele CW, Karim SA, Leach JD, et al. CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma. Cancer Cell. 2016;29:832–45. doi: 10.1016/j.ccell.2016.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ijichi H, Chytil A, Gorska AE, et al. Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma. J Clin Invest. 2011;121:4106–17. doi: 10.1172/JCI42754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Manuel ER, Chen J, D’Apuzzo M, et al. Salmonella-Based Therapy Targeting Indoleamine 2,3-Dioxygenase Coupled with Enzymatic Depletion of Tumor Hyaluronan Induces Complete Regression of Aggressive Pancreatic Tumors. Cancer immunology research. 2015;3:1096–107. doi: 10.1158/2326-6066.CIR-14-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kuwada Y, Sasaki T, Morinaka K, Kitadai Y, Mukaida N, Chayama K. Potential involvement of IL-8 and its receptors in the invasiveness of pancreatic cancer cells. International journal of oncology. 2003;22:765–71. [PubMed] [Google Scholar]
  • 68.Wang S, Wu Y, Hou Y, et al. CXCR2 macromolecular complex in pancreatic cancer: a potential therapeutic target in tumor growth. Translational oncology. 2013;6:216–25. doi: 10.1593/tlo.13133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Witkiewicz A, Williams TK, Cozzitorto J, et al. Expression of indoleamine 2,3-dioxygenase in metastatic pancreatic ductal adenocarcinoma recruits regulatory T cells to avoid immune detection. Journal of the American College of Surgeons. 2008;206:849–54. doi: 10.1016/j.jamcollsurg.2007.12.014. discussion 54-6. [DOI] [PubMed] [Google Scholar]
  • 70.Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nature medicine. 2003;9:1269–74. doi: 10.1038/nm934. [DOI] [PubMed] [Google Scholar]
  • 71.Zhang J, Han S, Zhang B, Zhang Y. Cancer immunology and cancer immunodiagnosis. Journal of immunology research. 2014;2014:725691. doi: 10.1155/2014/725691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.McAllister F, Bailey JM, Alsina J, et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell. 2014;25:621–37. doi: 10.1016/j.ccr.2014.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tassi E, Gavazzi F, Albarello L, et al. Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. Journal of immunology. 2008;181:6595–603. doi: 10.4049/jimmunol.181.9.6595. [DOI] [PubMed] [Google Scholar]
  • 74.De Monte L, Reni M, Tassi E, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. The Journal of experimental medicine. 2011;208:469–78. doi: 10.1084/jem.20101876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–6. doi: 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418–29. doi: 10.1016/j.ccr.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Thompson CB, Shepard HM, O’Connor PM, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Molecular cancer therapeutics. 2010;9:3052–64. doi: 10.1158/1535-7163.MCT-10-0470. [DOI] [PubMed] [Google Scholar]
  • 78.Sohr S, Engeland K. RHAMM is differentially expressed in the cell cycle and downregulated by the tumor suppressor p53. Cell cycle. 2008;7:3448–60. doi: 10.4161/cc.7.21.7014. [DOI] [PubMed] [Google Scholar]
  • 79.Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20. doi: 10.1136/gutjnl-2012-302529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Michl P, Gress TM. Improving drug delivery to pancreatic cancer: breaching the stromal fortress by targeting hyaluronic acid. Gut. 2012;61:1377–9. doi: 10.1136/gutjnl-2012-302604. [DOI] [PubMed] [Google Scholar]
  • 81.Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–61. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rucki AA, Zheng L. Pancreatic cancer stroma: understanding biology leads to new therapeutic strategies. World journal of gastroenterology. 2014;20:2237–46. doi: 10.3748/wjg.v20.i9.2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–47. doi: 10.1016/j.ccr.2014.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Marechal R, Bachet JB, Mackey JR, et al. Levels of gemcitabine transport and metabolism proteins predict survival times of patients treated with gemcitabine for pancreatic adenocarcinoma. Gastroenterology. 2012;143:664–74. e1–6. doi: 10.1053/j.gastro.2012.06.006. [DOI] [PubMed] [Google Scholar]
  • 85.Greenhalf W, Ghaneh P, Neoptolemos JP, et al. Pancreatic cancer hENT1 expression and survival from gemcitabine in patients from the ESPAC-3 trial. Journal of the National Cancer Institute. 2014;106 doi: 10.1093/jnci/djt347. djt347. [DOI] [PubMed] [Google Scholar]
  • 86.Kota J, Hancock J, Kwon J, Korc M. Pancreatic cancer: Stroma and its current and emerging targeted therapies. Cancer letters. 2017;391:38–49. doi: 10.1016/j.canlet.2016.12.035. [DOI] [PubMed] [Google Scholar]
  • 87.Heng EC, Huang Y, Black SA, Jr, Trackman PC. CCN2, connective tissue growth factor, stimulates collagen deposition by gingival fibroblasts via module 3 and alpha6- and beta1 integrins. Journal of cellular biochemistry. 2006;98:409–20. doi: 10.1002/jcb.20810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Abreu JG, Ketpura NI, Reversade B, De Robertis EM. Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-beta. Nature cell biology. 2002;4:599–604. doi: 10.1038/ncb826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hama K, Ohnishi H, Yasuda H, et al. Angiotensin II stimulates DNA synthesis of rat pancreatic stellate cells by activating ERK through EGF receptor transactivation. Biochemical and biophysical research communications. 2004;315:905–11. doi: 10.1016/j.bbrc.2004.01.155. [DOI] [PubMed] [Google Scholar]
  • 90.Hama K, Ohnishi H, Aoki H, et al. Angiotensin II promotes the proliferation of activated pancreatic stellate cells by Smad7 induction through a protein kinase C pathway. Biochemical and biophysical research communications. 2006;340:742–50. doi: 10.1016/j.bbrc.2005.12.069. [DOI] [PubMed] [Google Scholar]
  • 91.Doi C, Egashira N, Kawabata A, et al. Angiotensin II type 2 receptor signaling significantly attenuates growth of murine pancreatic carcinoma grafts in syngeneic mice. BMC cancer. 2010;10:67. doi: 10.1186/1471-2407-10-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Arnold SA, Rivera LB, Carbon JG, et al. Losartan slows pancreatic tumor progression and extends survival of SPARC-null mice by abrogating aberrant TGFbeta activation. PloS one. 2012;7:e31384. doi: 10.1371/journal.pone.0031384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Masamune A, Hamada S, Kikuta K, et al. The angiotensin II type I receptor blocker olmesartan inhibits the growth of pancreatic cancer by targeting stellate cell activities in mice. Scandinavian journal of gastroenterology. 2013;48:602–9. doi: 10.3109/00365521.2013.777776. [DOI] [PubMed] [Google Scholar]
  • 94.Biankin AV, Waddell N, Kassahn KS, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399–405. doi: 10.1038/nature11547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kano MR, Bae Y, Iwata C, et al. Improvement of cancer-targeting therapy, using nanocarriers for intractable solid tumors by inhibition of TGF-beta signaling. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:3460–5. doi: 10.1073/pnas.0611660104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Medicherla S, Li L, Ma JY, et al. Antitumor activity of TGF-beta inhibitor is dependent on the microenvironment. Anticancer research. 2007;27:4149–57. [PubMed] [Google Scholar]
  • 97.Gore AJ, Deitz SL, Palam LR, Craven KE, Korc M. Pancreatic cancer-associated retinoblastoma 1 dysfunction enables TGF-beta to promote proliferation. J Clin Invest. 2014;124:338–52. doi: 10.1172/JCI71526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kozono S, Ohuchida K, Eguchi D, et al. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Cancer Res. 2013;73:2345–56. doi: 10.1158/0008-5472.CAN-12-3180. [DOI] [PubMed] [Google Scholar]
  • 99.Li Ji T, Zhang SY, et al. An MMP-2 Responsive Liposome Integrating Antifibrosis and Chemotherapeutic Drugs for Enhanced Drug Perfusion and Efficacy in Pancreatic Cancer. ACS applied materials & interfaces. 2016;8:3438–45. doi: 10.1021/acsami.5b11619. [DOI] [PubMed] [Google Scholar]
  • 100.Suklabaidya S, Das B, Ali SA, et al. Characterization and use of HapT1-derived homologous tumors as a preclinical model to evaluate therapeutic efficacy of drugs against pancreatic tumor desmoplasia. Oncotarget. 2016;7:41825–42. doi: 10.18632/oncotarget.9729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303) Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28:3617–22. doi: 10.1200/JCO.2010.28.1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Chen L, Zhang X. Primary analysis for clinical efficacy of immunotherapy in patients with pancreatic cancer. Immunotherapy. 2016;8:223–34. doi: 10.2217/imt.15.105. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang Y, Choi M. Immune Therapy in Pancreatic Cancer: Now and the Future? Reviews on recent clinical trials. 2015;10:317–25. doi: 10.2174/1574887110666150916142537. [DOI] [PubMed] [Google Scholar]
  • 104.Watt J, Kocher HM. The desmoplastic stroma of pancreatic cancer is a barrier to immune cell infiltration. Oncoimmunology. 2013;2:e26788. doi: 10.4161/onci.26788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell. 2010;17:135–47. doi: 10.1016/j.ccr.2009.12.041. [DOI] [PubMed] [Google Scholar]
  • 107.Kalluri R, Zeisberg M. Fibroblasts in cancer. Nature reviews Cancer. 2006;6:392–401. doi: 10.1038/nrc1877. [DOI] [PubMed] [Google Scholar]
  • 108.Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:20212–7. doi: 10.1073/pnas.1320318110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunological reviews. 2009;229:152–72. doi: 10.1111/j.1600-065X.2009.00782.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res. 2013;19:6286–95. doi: 10.1158/1078-0432.CCR-13-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. The New England journal of medicine. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Nomi T, Sho M, Akahori T, et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res. 2007;13:2151–7. doi: 10.1158/1078-0432.CCR-06-2746. [DOI] [PubMed] [Google Scholar]
  • 113.Neuzillet C, Tijeras-Raballand A, Cros J, Faivre S, Hammel P, Raymond E. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer metastasis reviews. 2013;32:585–602. doi: 10.1007/s10555-013-9439-3. [DOI] [PubMed] [Google Scholar]
  • 114.Hidalgo M, Plaza C, Musteanu M, et al. SPARC Expression Did Not Predict Efficacy of nab-Paclitaxel plus Gemcitabine or Gemcitabine Alone for Metastatic Pancreatic Cancer in an Exploratory Analysis of the Phase III MPACT Trial. Clin Cancer Res. 2015;21:4811–8. doi: 10.1158/1078-0432.CCR-14-3222. [DOI] [PubMed] [Google Scholar]
  • 115.Schneeweiss A, Seitz J, Smetanay K, et al. Efficacy of nab-paclitaxel does not seem to be associated with SPARC expression in metastatic breast cancer. Anticancer research. 2014;34:6609–15. [PubMed] [Google Scholar]
  • 116.Cullis J, Siolas D, Avanzi A, Barui S, Maitra A, Bar-Sagi D. Macropinocytosis of Nab-paclitaxel Drives Macrophage Activation in Pancreatic Cancer. Cancer immunology research. 2017;5:182–90. doi: 10.1158/2326-6066.CIR-16-0125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sherman MH, Yu RT, Engle DD, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell. 2014;159:80–93. doi: 10.1016/j.cell.2014.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Froeling FE, Feig C, Chelala C, et al. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to slow tumor progression. Gastroenterology. 2011;141:1486–97. 97 e1–14. doi: 10.1053/j.gastro.2011.06.047. [DOI] [PubMed] [Google Scholar]
  • 119.Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications. Nature reviews Cancer. 2014;14:598–610. doi: 10.1038/nrc3792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Serrels A, Lund T, Serrels B, et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell. 2015;163:160–73. doi: 10.1016/j.cell.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Jiang H, Hegde S, Knolhoff BL, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nature medicine. 2016;22:851–60. doi: 10.1038/nm.4123. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES