Skip to main content
Annals of Gastroenterological Surgery logoLink to Annals of Gastroenterological Surgery
. 2018 Jun 22;2(4):274–281. doi: 10.1002/ags3.12176

Immunotherapy for pancreatic cancer: Barriers and breakthroughs

Robert J Torphy 1, Yuwen Zhu 1, Richard D Schulick 1,
PMCID: PMC6036358  PMID: 30003190

Abstract

Immunotherapy is a rapidly growing field and represents a paradigm shift in the treatment of malignancies as it offers a new therapeutic approach beyond surgery, conventional chemotherapy, and radiation treatment. Targeting immune checkpoints, such as cytotoxic T‐lymphocyte‐associated antigen 4 and programmed death 1/programmed death ligand 1 has had immense clinical success resulting in sustained treatment response for a subset of patients with certain malignancies such as melanoma, non‐small‐cell lung cancer, urothelial carcinoma, squamous cell carcinoma of the head and neck, renal cell cancer, hepatocellular cancer, and metastatic colorectal cancer. Importantly, there has been limited success in the use of immunotherapy in the treatment of pancreatic cancer. Investigation into the complex tumor microenvironment of pancreatic cancer that is composed of immune cells, stromal cells, and extracellular matrix proteins has begun to shed light on important attributes of this microenvironment that act as barriers to the effective use of immunotherapy. In this review, we will discuss the progress that has been made in treating pancreatic cancer with immunotherapy, the barriers that have limited treatment success, and breakthroughs with combination treatments that hold promise for the future.

Keywords: cancer vaccine, immune checkpoint, immunotherapy, pancreatic cancer, tumor microenvironment

1. INTRODUCTION

Pancreatic cancer is projected to be the third leading cause of cancer‐related death in the USA in 2018.1 Significant improvements in the treatment of pancreatic cancer have been made over the past two decades, with a median survival of 28.0 months now reported for patients with resectable disease who receive adjuvant gemcitabine plus capecitabine.2 Although outcomes have improved dramatically for patients with resectable disease who receive adjuvant chemotherapy, there has been very limited advancement in treating metastatic pancreatic cancer since gemcitabine was shown to be superior to 5‐fluorouracil in 1997.3 Since that time, the addition of nab‐paclitaxel to gemcitabine therapy and FOLFIRINOX have been shown to be alternative treatment strategies, but survival benefits with these treatments are modest and overall survival remains poor.4

Although the treatment of pancreatic cancer has evolved slowly over the past two decades, treatment of other malignancies has experienced immense progress through the breakthrough of cancer immunotherapy. However, the success of immunotherapy has not translated to the treatment of pancreatic cancer which has been shown to be unresponsive to anti‐programmed death 1 (anti‐PD‐1) and anti‐cytotoxic T‐lymphocyte‐associated antigen 4 (anti‐CTLA‐4).5, 6

The ineffectiveness of immunotherapy in pancreatic cancer may be explained by these tumors being non‐immunogenic.7 One explanation for the non‐immunogenic nature of these tumors is their poor antigenicity. A key initial step of launching effective anti‐tumor immunity is tumor antigenicity; T cells must be able to recognize malignant cells as foreign to elicit their destruction. The antigenicity of tumors can be, in part, inferred by their mutational landscape. Solid tumors such as pancreatic cancer, colon cancer, and breast cancer contain an average of 33 to 66 somatic mutations that result in altered protein production and, in turn, could lead to the expression of “foreign” antigens. Conversely, melanoma and lung cancer have approximately twice as many somatic mutations making them relatively more antigenic.8 Despite the decreased antigenicity of pancreatic cancer when compared to melanoma and lung cancer, multiple studies have indicated that the human immune system can develop an immune response to pancreatic cancer and generate functional anti‐tumor T cells.9, 10 This suggests that other mechanisms are likely contributing to the non‐immunogenic nature of pancreatic cancer.

It is well known that the interaction of the immune system and cancer cells is a dynamic process that can eliminate antigenic cancer cells, but can also evolve to a phase where cancers escape from immune surveillance.11 The process of immune escape is multifactorial but involves reduced immune recognition of cancer cells and the development of an immunosuppressive microenvironment. There is an increasing body of knowledge that suggests the tumor microenvironment of pancreatic cancer is very effective in promoting immune escape, rendering the immune system unable to mount an effective anti‐tumor response. Understanding the role of the tumor microenvironment in facilitating immune escape in pancreatic cancer holds great potential for improving the success of immunotherapy for pancreatic cancer in the future.

In this review we will briefly describe the limited success that has been seen with immune checkpoint blockade and pancreatic cancer vaccines and explore the unique tumor microenvironment of pancreatic cancer that acts to limit the effectiveness of immunotherapy. Last, we will highlight how the tumor microenvironment can be targeted in combination with immunotherapy to unleash the potential of immunotherapy as an effective treatment modality in pancreatic cancer.

2. LIMITED SUCCESS OF TRADITIONAL IMMUNOTHERAPY

2.1. Immune checkpoint blockade

Cytotoxic T‐lymphocyte‐associated antigen 4 is a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells and competes with CD28 for ligands B7‐1 and B7‐2. Blockade of CTLA‐4 can induce anti‐tumor immunity.12 Ipilimumab is a fully human monoclonal antibody (mAb) that improves overall survival in patients with metastatic melanoma and elicits a long‐term survival benefit in a subset of patients.13 Ipilimumab became the first immune checkpoint‐targeted therapy to receive approval for clinical use in the USA and Europe in 2011. Ipilimumab treatment was evaluated in a phase II study in patients with advanced pancreatic cancer in 27 patients and showed a delayed response in one patient only, indicating that single‐agent ipilimumab was not an effective therapy in advanced pancreatic cancer.5

Programmed death 1 was the second coinhibitory receptor to come to the forefront of cancer immunotherapy. PD‐1 is expressed by effector T cells, regulatory T (Treg) cells, B cells and natural killer (NK) cells and binds to the ligands programmed death ligand 1 (PD‐L1; B7‐H1) and programmed death ligand 2 (PD‐L2; B7‐DC). Upon binding to these ligands, PD‐1 acts to inhibit T cells.14 PD‐L1 is expressed both by cancer cells and tumor‐infiltrating lymphocytes and upregulation of PD‐L1 on tumor cells is an adaptive mechanism to facilitate immune evasion. PD‐1 mAb therapy with pembrolizumab and nivolumab have had great clinical success for a variety of solid tumors and have received Food and Drug Administration (FDA) approval for the treatment of multiple solid tumors, including melanoma, non‐small‐cell lung cancer, urothelial carcinoma, and renal cell cancer.15, 16 Atezolizumab is an anti‐PD‐L1 monoclonal antibody that can also disrupt the interaction between PD‐1 and its ligands,17 and has been approved for the treatment of non‐small‐cell lung cancer and urothelial carcinoma.18

Multiple human studies have indicated that high PD‐L1 expression in pancreatic cancer tumors is associated with worse outcomes suggesting that targeting the PD‐1/PD‐L1 interaction may have therapeutic benefit in these patients.19, 20, 21 An early preclinical study in a mouse tumor transplant model showed that PD‐1 or PD‐L1 blockade had an anti‐tumor effect which was enhanced when given together with gemcitabine.19 Unfortunately, these preclinical findings have not translated to clinical success. In a phase I trial of anti‐PD‐L1 therapy, no patients with pancreatic cancer showed a clinical response.6

2.2. Vaccine therapy

Cancer vaccines are designed to augment antigen presentation and activate antigen‐specific effector and memory T cells.22 When vaccines containing target tumor antigens are given, host antigen‐presenting cells (APC) are tasked with taking up these antigens for presentation to effector T cells which are then primed to kill tumor cells expressing these antigens. The desired result is the development of anti‐tumor immunity. Several pancreatic cancer antigens have been identified that are shared by the majority of pancreatic tumors and include carcinoembryonic antigen (CEA), mucin‐1 (MUC‐1), and the product of mutated KRAS.23 All these antigens have the potential to be used as vaccines for pancreatic cancer. In addition, allogenic tumor cell vaccination, using tumor cell lines, can induce effective and tumor‐specific immune responses in mouse tumor models.24

GVAX is a whole‐cell vaccine in which pancreatic cancer cells are engineered to express the proinflammatory cytokine granulocyte monocyte‐colony stimulating factor (GM‐CSF) to further stimulate APC antigen uptake and T‐cell priming.25 A phase I study evaluated the safety and efficacy of GVAX as an adjuvant therapy given in series with chemoradiation therapy in patients with resected pancreatic cancer. This phase I study showed that vaccination with GVAX was safe. Data from this trial also provided evidence that GVAX was effective in promoting the development of anti‐tumor immunity, as vaccinated patients developed a dose‐dependent delayed‐type hypersensitive reaction against autologous tumor cells.23 This immunotherapy approach was further evaluated in a phase II study of 60 patients and their outcomes were compared to historical controls. Patients who received adjuvant vaccine therapy in conjunction with chemoradiation did not show improved 1‐year or overall survival compared to historical controls treated with adjuvant chemoradiation therapy alone. However, a subgroup of patients with prolonged disease‐free survival had increased tumor antigen‐specific CD8+ T cells after their final vaccination suggesting this treatment approach may be effective for certain patients.26

An alternative to whole‐cell vaccines are peptide‐based vaccines, an example of which is the peptide product of mutated KRAS. The KRAS gene is mutated in over 90% of pancreatic adenocarcinomas, making it a vaccine target that would be broadly applicable for the treatment of pancreatic cancer.27 A phase I/II trial of a mutated RAS peptide vaccine in conjunction with GM‐CSF in patients with resected or advanced pancreatic cancer showed that this treatment strategy elicited an anti‐RAS immune response in 58% of patients. Further, median survival in those who developed an anti‐RAS immune response was more than double those who did not develop a response.28 A recent phase II study of 30 patients from Japan also indicated safety and efficacy of an adjuvant multipeptide vaccine and chemotherapy regimen following surgical resection. The multipeptide vaccine, OCV‐C01, contained peptides derived from vascular endothelial growth factor receptor (VEGFR)1, VEGFR2 and a kinesin‐family protein (KIF20A). This regimen was well tolerated and, importantly, 58.6% of patients developed a cytotoxic lymphocyte response against KIF20A. In the per‐protocol analysis, patients who developed a KIF20A‐specific cytotoxic lymphocyte response had a significantly improved disease‐free survival.29 These promising results indicate that vaccine therapy can be a useful immunotherapy in the treatment of pancreatic cancer, but more work is needed to identify key biomarkers that predict response, and to optimize combinatory therapies to increase the effectiveness for all patients.

2.3. Vaccine and immune checkpoint blockade in combination

A phase I study compared ipilimumab and ipilimumab in combination with GVAX in locally advanced and metastatic pancreatic cancer. This study included 30 patients, randomized 1:1, and showed that combination therapy was safe with evidence favoring increased efficacy with combination therapy. Two patients in the ipilimumab‐alone arm showed stable disease at 7 and 22 weeks, whereas three patients in the combination arm had prolonged disease stabilization at 31, 71, and 81 weeks. Patients from both treatment arms who showed prolonged overall survival had higher levels of mesothelin‐specific CD8+ T cells in peripheral blood samples after treatment indicating an improved anti‐cancer T‐cell response.30

Preclinical studies in melanoma indicated that GM‐CSF‐producing vaccines in combination with immune checkpoint blockade was able to effectively treat a non‐immunogenic melanoma cancer cell line that did not respond to immune checkpoint blockade therapy alone.31 A similar treatment modality was tested in preclinical models of pancreatic cancer using GVAX in combination with anti‐PD‐1 therapy. Combination therapy was found to significantly increase median overall survival compared to PD‐1 monotherapy with a trend towards improved overall survival. Tumor infiltrating lymphocytes (TIL) collected from the tumor microenvironment of mice that received combination therapy had an increase in the percentage of interferon (IFN)‐gamma‐producing CD8+ T cells compared with single‐agent therapy, indicating a synergistic effect on anti‐tumor immunity.32 A possible mechanism for this synergy is that vaccination induces some anti‐tumor immunity that is overcome by upregulation of PD‐L1 on tumors. Patients who received the GVAX vaccine 2 weeks prior to surgical resection had an increased frequency of tumors positive for PD‐L1 (12.5% of resected tumors in the unvaccinated group vs 25% of resected tumors in the GVAX group), suggesting this as a likely mechanism of immune resistance.32 Building on these results, GVAX is currently being studied in a clinical trial with or without nivolumab for patients with resectable pancreatic cancer at Johns Hopkins University (NCT02451982; clinicaltrials.gov).

3. TUMOR MICROENVIRONMENT AS A BARRIER TO IMMUNOTHERAPY

Pancreatic cancer tumors are composed of malignant cancer cells surrounded by an abundant desmoplastic stroma (Figure 1A). This desmoplastic stroma forms the tumor microenvironment of pancreatic cancer and is composed of fibroblasts, pancreatic stellate cells, immune cells, blood vessels, and extracellular matrix proteins.33 This abundant stroma has been implicated as a physical barrier in pancreatic cancer that plays a role in preventing effective delivery of standard chemotherapies to tumors.34 Depleting stroma in preclinical mouse models of pancreatic cancer through inhibiting the Hedgehog cellular signaling pathway was shown to improve delivery of gemcitabine to tumors and resulted in improved survival.35 However, these results did not translate to clinical success. A phase II clinical trial of gemcitabine with vismodegib, a Hedgehog antagonist used to deplete stroma, showed no survival benefit in pancreatic cancer patients.36 Subsequent studies have reported stroma depletion may actually enhance tumor growth underscoring the complex role that stroma plays in tumor biology.37 Despite the conflicting results of tumor response to stroma‐depleting therapies, the tumor microenvironment plays a significant role in tumor biology and in modulating the immune recognition of pancreatic cancer (Figure 1B).

Figure 1.

Figure 1

Abundant desmoplastic stroma of pancreatic cancer. A, Hematoxylin and eosin‐stained primary pancreatic ductal adenocarcinoma (10×), showing abundant desmoplastic stroma surrounding tumor epithelial cells. B, Interaction of tumor epithelial cells and stromal components can impede drug delivery, enhance or impair tumor growth and metastasis, and alter immune surveillance of tumors

3.1. Immunosuppressive immune cells in the tumor microenvironment

Genetically modified mouse models that develop spontaneous pancreatic tumors are an important model for studying the effects of the tumor microenvironment on response to immunotherapy. One such model is the KPC mouse, which is genetically engineered to express mutant Kras and mutant p53 in the pancreas.38 This model develops pre‐invasive lesions (pancreatic intraepithelial neoplasia) which progress to invasive and metastatic disease, recapitulating the progression of human disease. These spontaneous tumors also have an abundant desmoplastic stroma reaction like that found in human tumors.39 Evaluation of the immune response in pre‐invasive and invasive lesions from these mouse models showed that leukocyte invasion is present throughout the progression of disease.40, 41, 42 However, this leukocyte invasion was dominated by immunosuppressive cell types (tumor‐associated macrophages, myeloid‐derived suppressor cells [MDSC], and Treg cells) and lacked effector T cells.40 These findings are significant as they indicate a suppressed immune response from the earliest development of pre‐invasive lesions as opposed to an activated immune response that is overcome by the tumor developing resistance mechanisms.

The prevalence of these immunosuppressive cell types and their prognostic significance has also been shown in humans. Patients with pancreatic cancer have elevated levels of Treg and MDSC in peripheral blood samples when compared to healthy controls and elevated levels of these immunosuppressive cells are associated with a poor prognosis.43, 44 In the tumor microenvironment of the pancreas, Treg, MDSC, and M2‐polarized tumor‐associated macrophages are also increased and are associated with a poor prognosis.44, 45, 46, 47

3.2. Immunosuppressive function of carcinoma‐associated fibroblasts

Along with the immunosuppressive immune cells found in the tumor microenvironment of pancreatic cancer, these tumors contain an abundant stroma of mesenchymal origin rich in fibroblasts. Carcinoma‐associated fibroblasts of epithelial malignancies have been shown to express fibroblast activation protein‐α (FAP).34 In pancreatic cancer, high FAP expression in tumors correlates with worse prognosis.34, 48 Kraman et al49 further investigated the potential of FAP to have immunosuppressive properties in the tumor microenvironment using a transgenic mouse model depleted of FAP‐positive cells. Mice that were depleted of FAP+ cells and harboring subcutaneous tumors showed tumor regression after treatment with a vaccine‐based immunotherapy. In contrast, wild‐type mice did not show tumor regression, suggesting depletion of FAP+ cells enabled the efficacy of this vaccine‐based immunotherapy. In a separate study using KPC mice, depletion of FAP+ stromal cells was synergistic with anti‐CTLA‐4 or anti‐PD‐L1 therapy, further demonstrating the role of stroma in suppressing anti‐tumor immunity.50

4. BREAKTHROUGHS WITH COMBINATION THERAPIES

The tumor microenvironment of pancreatic cancer presents a therapeutic barrier in treating pancreatic cancer with traditional immunotherapies. However, treating pancreatic cancer with combination approaches that reprogram the tumor microenvironment and ultimately unleash the potential benefits of immunotherapy are now being pursued with promising preliminary results.

4.1. Colony‐stimulating factor 1 receptor blockade to reprogram the immunosuppressive microenvironment

In the tumor microenvironment, colony‐stimulating factor 1 receptor (CSF1R) is expressed on tumor associated macrophages and MDSC which can play critical roles suppressing the cytotoxic immune response. Investigation of the expression of CSF1R and its ligand, CSF1, indicating that CSF1 is frequently expressed by pancreatic cancer cells whereas CSF1R expression is limited to cells in the tumor microenvironment, suggesting a tumor cell/tumor microenvironment interaction. Blockade of CSF1R has been shown to improve chemotherapy‐induced antitumor immunity in mouse cancer models leading to the hypothesis that targeting the CSF1R/CSF1 interaction in combination with immune checkpoint blockade could have a synergistic response.51, 52

In mouse pancreatic cancer tumor transplant models, treatment with CSF1R blockade resulted in a reprogramming of the immunosuppressive tumor microenvironment. Following treatment with CSF1R blockade by a CSF1R tyrosine kinase inhibitor tumor, infiltrating macrophages and MDSC were depleted. Treatment of tumor‐bearing mice with CSF1R blockade did result in enhanced effector T‐cell tumor infiltration and reduced tumor growth. However, following treatment, tumor cells had increased expression of PD‐L1 and effector T cells had increased expression of CTLA4, suggesting immune checkpoint blockade in conjunction with CSF1R blockade may have a synergistic effect. Combination therapy with gemcitabine, CSF1R blockade and either anti‐CTLA4 or anti‐PD1 therapy resulted in a synergistic response that was further enhanced with co‐blockade of both PD‐1 and CTLA‐4 with complete tumor regression in 30% of animals and an average tumor regression of 85%.52

IMC‐CS4 is a CSF1R antibody currently in phase I clinical trial in conjunction with GVAX and anti‐PD1 therapy for borderline resectable pancreatic cancer. PLX‐3397, commercially named Pexidartinib (Plexxikon Inc., Berkeley, CA, USA), is another anti‐CSF1R agent that is currently in phase I clinical trial in conjunction with anti‐PD‐L1 for patients with advanced pancreatic and colorectal cancer (Table 1).

Table 1.

Active clinical trials of novel or combination immunotherapy approaches for the treatment of pancreatic cancer

Treatment Phase Patient population ClinicalTrials.gov identifier
GVAX
CSF1R antibody (IMC‐CS4)
Anti‐PD1 (Pembrolizumab)
Cyclophosphamide
1 Borderline resectable pancreatic cancer NCT03153410
Anti‐PD‐L1 (Durvalumab)
CSF1R inhibitor (Pexidartinib)
1 Advanced pancreatic and colorectal cancer NCT02777710
CXCR4 antagonist (Plerixafor) 1 Advanced pancreatic cancer NCT03277209
CD40 agonist (R07009789)
Nab‐Paclitaxel
Gemcitabine
1 Resectable pancreatic cancer NCT02588443

CSF1R, colony‐stimulating factor 1 receptor; PD‐1, programmed death 1; PD‐L1, programmed death ligand 1.

4.2. Targeting CXCL12/CXCR4 to reverse carcinoma‐associated fibroblast immunosuppression

Carcinoma‐associated fibroblasts are an abundant component of the tumor microenvironment and are no longer considered an innocent bystander in cancer progression. Depleting FAP+ carcinoma‐associated fibroblasts in mouse models was shown to be synergistic with vaccine‐based or immune checkpoint‐based immunotherapies in eliciting anti‐tumor immunity and tumor regression.49, 50 However, therapeutically targeting the stroma of pancreatic cancer is challenging in humans as the cells that comprise this tissue compartment are present throughout the human body and play important roles in normal homeostasis. Thus, pan‐depletion of FAP+ fibroblasts in humans is not a safe or feasible treatment approach.

Feig et al50 showed that FAP+ stromal cells produce C‐X‐C motif chemokine ligand 12 (CXCL12), also referred to as stromal‐derived factor‐1. CXCL12 binds to the C‐X‐C chemokine receptor type 4 (CXCR4) and functions to promote cellular chemotaxis.53 In pancreatic cancer, CXCL12 is abundant in the tumor microenvironment, whereas CXCR4 has been shown to be expressed on cancer and endothelial cells.54 CXCL12 was shown to promote chemotaxis of pancreatic cancer cells expressing CXCR4 in vitro which could be inhibited with AMD3100, an antagonist anti‐CXCR4 mAb, suggesting a mechanism of tumor‐stromal cross‐talk.55 CXCR4 is also expressed on hematopoietic cells including macrophages and T cells, and CXCL12 acts as a chemoattractant for these immune cells.56

To evaluate whether the interaction of CXCL12 and CXCR4 is significant in the mechanism of FAP+ stromal‐mediated immunosuppression, tumor‐bearing mice were treated with AMD3100, resulting in a T‐cell mediated reduction in tumor growth. When AMD3100 was combined with immune checkpoint blockade with anti‐PD‐L1 therapy, tumor regression occurred with combination treatment but not with checkpoint blockade alone. Mice treated with the combination therapy had increased numbers of intratumoral T cells, suggesting a mechanism behind this treatment effect is improved T‐cell trafficking into the tumor microenvironment.50

AMD3100, commercially named Mozobil (Plerixafor injection; Sanofi Oncology, Bridgewater, NJ, USA), is in phase I clinical trial to evaluate its effect on T‐cell infiltration in the tumor microenvironment of patients with advanced pancreatic cancer (Table 1).

4.3. Immune checkpoint blockade with agonist CD40 mAb to induce T‐cell immunity

CD40 is a cell surface molecule expressed by immune cells and plays a role in both cellular and humoral immunity. The binding of CD40 to the CD40 ligand expressed on CD4+ helper T cells results in activation of antigen‐presenting cells.57 Furthermore, triggering CD40 has been shown to enhance the efficacy of vaccines in promoting anti‐tumor immunity.57 These findings led to the hypothesis that anti‐CD40 agonist mAb therapy could be used in conjunction with chemotherapy as a novel treatment for pancreatic cancer.58

In preclinical studies, CD40 agonist therapy has elicited tumor regression when used in combination with chemotherapy.59, 60, 61 In the KPC model of pancreatic adenocarcinoma, combination treatment with gemcitabine and an agonist CD40 antibody resulted in regression of spontaneous tumors in 30% of mice.59 Agonist CD40 treatment in combination with gemcitabine and nab‐paclitaxel also resulted in tumor regression of subcutaneously implanted KPC tumor cells and prolonged overall survival compared to anti‐CD40 therapy or chemotherapy alone.59, 61

In a phase I clinical trial, CD40 agonist mAb therapy in combination with gemcitabine resulted in tumor response in 19% of patients with unresectable chemotherapy naïve pancreatic cancer.62 Tumor biopsies from these patients showed a dearth of TIL and an abundance of tumor infiltrating macrophages.62 A second phase I study is aimed at studying the combination of anti‐CD40 therapy with nab‐paclitaxel and gemcitabine in pancreatic cancer and is currently enrolling (Table 1).

In mice with subcutaneous KPC tumors, treatment with anti‐PD‐1 or anti‐CTLA‐4 enhanced the anti‐tumor effects of chemotherapy and agonist CD40 treatment. Thirty‐nine percent of mice treated with anti‐PD‐1, anti‐CTLA4 and an agonist CD40 antibody had long‐term complete tumor remission and prolonged survival. This remarkable tumor response was shown to be T‐cell dependent. CD40 and chemotherapy treatment resulted in changes in the immune microenvironment with a reduction in Treg and an increase in CD8+ T cells which was further augmented with immune checkpoint blockade. Importantly, when mice that had a complete response were challenged with tumor cells in the opposite flank, the majority of mice rejected this challenge without additional therapy suggesting developed lasting anti‐tumor immune memory. KPC mice with spontaneous pancreatic tumors who received the combination treatment also had an improved median overall survival compared to control‐treated mice, anti‐PD‐1‐alone treated mice, or CD40/chemotherapy‐treated mice.7 Currently, R07009789 (a CD40 agonist mAb) is in phase I clinical trial in conjunction with gemcitabine and nabpaclitaxel for patients with resectable pancreatic cancer (Table 1).

5. CONCLUSIONS

  1. Patients with pancreatic cancer show poor response to checkpoint blockade with anti‐CTLA‐4 and anti‐PD1/anti‐PD‐L1 immunotherapies.

  2. Pancreatic cancer has an abundant desmoplastic stroma which is composed of fibroblasts, immune cells, endothelial cells, and extracellular matrix proteins.

  3. The tumor microenvironment is dominated by immunosuppressive cell types (tumor‐associated macrophages, MDSC, and Treg cells) and lacks effector T cells.

  4. Carcinoma‐associated fibroblasts have important immunosuppressive effects. Fibroblast activation protein‐α‐positive cells have been shown to be an important mediator in promoting an immunosuppressive microenvironment.

  5. Combination treatment strategies that act to stimulate the immune response and break down the barriers of the tumor microenvironment in conjunction with immunotherapy hold promise for improving care for pancreatic cancer patients.

DISCLOSURE

Conflicts of interest: R.J. Torphy and Y. Zhu declare no conflicts of interest for this article. R.D. Schulick declares patents licensed to Aduro Biotech and GlaxoSmithKline.

Torphy RJ, Zhu Y, Schulick RD. Immunotherapy for pancreatic cancer: Barriers and breakthroughs. Ann Gastroenterol Surg. 2018;2:274–281. https://doi.org/10.1002/ags3.12176

REFERENCES

  • 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30. [DOI] [PubMed] [Google Scholar]
  • 2. 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 (London, England). 2017;389:1011–24. [DOI] [PubMed] [Google Scholar]
  • 3. Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first‐line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13. [DOI] [PubMed] [Google Scholar]
  • 4. Manji GA, Olive KP, Saenger YM, Oberstein P. Current and emerging therapies in metastatic pancreatic cancer. Clin Cancer Res. 2017;23:1670–8. [DOI] [PubMed] [Google Scholar]
  • 5. Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti‐CTLA‐4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti‐PD‐L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Winograd R, Byrne KT, Evans RA, et al. Induction of T‐cell immunity overcomes complete resistance to PD‐1 and CTLA‐4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol Res. 2015;3:399–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW. Cancer genome landscapes. Science (New York, NY). 2013;339:1546–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Fukunaga A, Miyamoto M, Cho Y, et al. CD8+  tumor‐infiltrating lymphocytes together with CD4+  tumor‐infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 2004;28:e26–31. [DOI] [PubMed] [Google Scholar]
  • 10. Schmitz‐Winnenthal FH, Volk C, Z'Graggen K, et al. High frequencies of functional tumor‐reactive T cells in bone marrow and blood of pancreatic cancer patients. Cancer Res. 2005;65:10079–87. [DOI] [PubMed] [Google Scholar]
  • 11. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases–elimination, equilibrium and escape. Curr Opin Immunol. 2014;27:16–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA‐4 blockade. Science. 1996;271:1734–6. [DOI] [PubMed] [Google Scholar]
  • 13. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD‐1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–30. [DOI] [PubMed] [Google Scholar]
  • 16. Reck M, Rodriguez‐Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD‐L1‐Positive Non‐Small‐Cell Lung Cancer. N Engl J Med. 2016;375:1823–33. [DOI] [PubMed] [Google Scholar]
  • 17. Hoffman‐Censits JH, Grivas P, Van Der Heijden MS, et al. IMvigor 210, a phase II trial of atezolizumab (MPDL3280A) in platinum‐treated locally advanced or metastatic urothelial carcinoma (mUC). J Clin Oncol. 2016;34(2_suppl):355. [Google Scholar]
  • 18. Krishnamurthy A, Jimeno A. Atezolizumab: A novel PD‐L1 inhibitor in cancer therapy with a focus in bladder and non‐small cell lung cancers. Drugs Today. 2017;53:217–37. [DOI] [PubMed] [Google Scholar]
  • 19. 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] [PubMed] [Google Scholar]
  • 20. Yamaki S, Yanagimoto H, Tsuta K, Ryota H, Kon M. PD‐L1 expression in pancreatic ductal adenocarcinoma is a poor prognostic factor in patients with high CD8(+) tumor‐infiltrating lymphocytes: highly sensitive detection using phosphor‐integrated dot staining. Int J Clin Oncol. 2017;22:726–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Zhuan‐Sun Y, Huang F, Feng M, et al. Prognostic value of PD‐L1 overexpression for pancreatic cancer: evidence from a meta‐analysis. Onco Targets Therapy. 2017;10:5005–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Soares KC, Zheng L, Edil B, Jaffee EM. Vaccines for pancreatic cancer. Cancer J. 2012;18:642–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Jaffee EM, Hruban RH, Biedrzycki B, 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. 2001;19:145–56. [DOI] [PubMed] [Google Scholar]
  • 24. Greten TF, Jaffee EM. Cancer vaccines. J Clin Oncol. 1999;17:1047–60. [DOI] [PubMed] [Google Scholar]
  • 25. Jaffee EM, Schutte M, Gossett J, et al. Development and characterization of a cytokine‐secreting pancreatic adenocarcinoma vaccine from primary tumors for use in clinical trials. Cancer J Sci Am. 1998;4:194–203. [PubMed] [Google Scholar]
  • 26. Lutz E, Yeo CJ, Lillemoe KD, 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. 2011;253:328–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. 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] [PubMed] [Google Scholar]
  • 28. Gjertsen MK, Buanes T, Rosseland AR, 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. 2001;92:441–50. [DOI] [PubMed] [Google Scholar]
  • 29. Miyazawa M, Katsuda M, Maguchi H, et al. Phase II clinical trial using novel peptide cocktail vaccine as a postoperative adjuvant treatment for surgically resected pancreatic cancer patients. Int J Cancer. 2017;140:973–82. [DOI] [PubMed] [Google Scholar]
  • 30. Le DT, Lutz E, Uram JN, et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM‐CSF gene in previously treated pancreatic cancer. J Immunother. 2013;36:382–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti‐cytotoxic T lymphocyte‐associated antigen 4 (CTLA‐4) and granulocyte/macrophage colony‐stimulating factor (GM‐CSF)‐producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med. 1999;190:355–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Soares KC, Rucki AA, Wu AA, et al. PD‐1/PD‐L1 blockade together with vaccine therapy facilitates effector T‐cell infiltration into pancreatic tumors. J Immunother. 2015;38:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Neesse A, Algul H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–84. [DOI] [PubMed] [Google Scholar]
  • 34. Garin‐Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proc Natl Acad Sci USA. 1990;87:7235–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. 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] [PMC free article] [PubMed] [Google Scholar]
  • 36. Catenacci DV, Junttila MR, Karrison T, et al. Randomized Phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Özdemir BC, Pentcheva‐Hoang T, Carstens JL, et al. Depletion of Carcinoma‐Associated Fibroblasts and Fibrosis Induces Immunosuppression and Accelerates Pancreas Cancer with Diminished Survival. Cancer Cell. 2014;25:719–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Hingorani SR, Wang L, Multani AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–83. [DOI] [PubMed] [Google Scholar]
  • 39. Hruban RH, Adsay NV, Albores‐Saavedra J, et al. Pathology of genetically engineered mouse models of pancreatic exocrine cancer: consensus report and recommendations. Cancer Res. 2006;66:95–106. [DOI] [PubMed] [Google Scholar]
  • 40. 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] [PubMed] [Google Scholar]
  • 41. Clark CE, Beatty GL, Vonderheide RH. Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer. Cancer Lett. 2009;279:1–7. [DOI] [PubMed] [Google Scholar]
  • 42. Hingorani SR, Petricoin EF, Maitra A, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4:437–50. [DOI] [PubMed] [Google Scholar]
  • 43. Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid‐derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin‐13. Cancer Immunol Immunother. 2011;60:1419–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Liyanage UK, Moore TT, Joo HG, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol. 2002;169:2756–61. [DOI] [PubMed] [Google Scholar]
  • 45. Kurahara H, Shinchi H, Mataki Y, et al. Significance of M2‐polarized tumor‐associated macrophage in pancreatic cancer. J Surg Res. 2011;167:e211–9. [DOI] [PubMed] [Google Scholar]
  • 46. 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. 2006;12:5423–34. [DOI] [PubMed] [Google Scholar]
  • 47. Hutcheson J, Balaji U, Porembka MR, et al. Immunological and metabolic features of pancreatic ductal adenocarcinoma define prognostic subtypes of disease. Clin Cancer Res. 2016;22:3606–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Shi M, Yu D‐H, Chen Y, et al. Expression of fibroblast activation protein in human pancreatic adenocarcinoma and its clinicopathological significance. World J Gastroenterol. 2012;18:840–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kraman M, Bambrough PJ, Arnold JN, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein‐alpha. Science. 2010;330:827–30. [DOI] [PubMed] [Google Scholar]
  • 50. 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. Proc Natl Acad Sci USA. 2013;110:20212–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Mitchem JB, Brennan DJ, Knolhoff BL, et al. Targeting tumor‐infiltrating macrophages decreases tumor‐initiating cells, relieves immunosuppression and improves chemotherapeutic responses. Can Res. 2013;73:1128–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zhu Y, Knolhoff BL, Meyer MA, et al. CSF1/CSF1R blockade reprograms tumor‐infiltrating macrophages and improves response to T‐cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Sun X, Cheng G, Hao M, et al. CXCL12/CXCR4/CXCR7 Chemokine Axis and Cancer Progression. Cancer Metastasis Rev. 2010;29:709–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. 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]
  • 55. Marchesi F, Monti P, Leone BE, et al. Increased survival, proliferation, and migration in metastatic human pancreatic tumor cells expressing functional CXCR4. Cancer Res. 2004;64:8420–7. [DOI] [PubMed] [Google Scholar]
  • 56. Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez‐Ramos JC. The chemokine SDF‐1 is a chemoattractant for human CD34+  hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+  progenitors to peripheral blood. J Exp Med. 1997;185:111–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Diehl L, den Boer AT, Schoenberger SP, et al. CD40 activation in vivo overcomes peptide‐induced peripheral cytotoxic T‐lymphocyte tolerance and augments anti‐tumor vaccine efficacy. Nat Med. 1999;5:774–9. [DOI] [PubMed] [Google Scholar]
  • 58. Vonderheide RH, Bajor DL, Winograd R, Evans RA, Bayne LJ, Beatty GL. CD40 immunotherapy for pancreatic cancer. Cancer Immunol Immunother. 2013;62:949–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. 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] [PMC free article] [PubMed] [Google Scholar]
  • 60. Beatty GL, Winograd R, Evans RA, et al. Exclusion of T Cells From Pancreatic Carcinomas in Mice is Regulated by Ly6C(low) F4/80(+) Extra‐tumor Macrophages. Gastroenterology. 2015;149:201–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Byrne KT, Vonderheide RH. CD40 stimulation obviates innate sensors and drives T cell immunity in cancer. Cell Rep. 2016;15:2719–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. 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] [PMC free article] [PubMed] [Google Scholar]

Articles from Annals of Gastroenterological Surgery are provided here courtesy of Wiley

RESOURCES