Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 1.
Published in final edited form as: Gastroenterology. 2019 Jan 18;156(7):2056–2072. doi: 10.1053/j.gastro.2018.12.038

Broadening the impact of immunotherapy to pancreatic cancer: Challenges and opportunities

Vinod P Balachandran 1,5,*, Gregory L Beatty 2,3,5,*, Stephanie K Dougan 4,5,*
PMCID: PMC6486864  NIHMSID: NIHMS1526872  PMID: 30660727

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second deadliest cancer in the US by 2025 1, with 5-year survival at less than 10% 2. In other recalcitrant cancers, immunotherapy has shown unprecedented response rates, including durable remissions after drug discontinuation. However, responses to immunotherapy in PDAC are rare. Accumulating evidence in mice and humans suggests that this remarkable resistance is linked to the complex, dueling role of the immune system in simultaneously promoting and restraining PDAC. In this review, we highlight the rationale that supports pursuing immunotherapy in PDAC, outline the key barriers that limit immunotherapy efficacy, and summarize the primary preclinical and clinical efforts to sensitize PDAC to immunotherapy.

Keywords: PDAC, immunity, clinical trials, immunotherapy, pancreatic cancer

1. Introduction

The most successful immunotherapies to date have leveraged the fundamental ability of T cells to recognize and kill tumor cells based on their aberrant expression of unique antigens. Immunotherapies harness this central tenant through multiple strategies, including boosting endogenous anti-tumor T cell activity with adoptive transfer of endogenous or genetically modified tumor-specific T cells, disrupting immune suppressive mechanisms restraining productive T cell immunosurveillance, and enhancing tumor antigen delivery with vaccines. Despite these varied approaches, PDAC has remained almost completely resistant to immunotherapy.

2. Barriers to immunotherapy in PDAC

A hallmark of the PDAC tumor microenvironment (TME) in both mice and humans is an abundance of non-cancer cell components in the tumor mass, collectively designated as the stroma. In PDAC, stroma can account for up to 50% of the total tumor mass and has been shown to inhibit both spontaneous and therapeutically induced anti-tumor immunity. Stromal barriers to immunotherapy in PDAC include both cellular and molecular suppressive components (Figure 1).

Figure 1. The pancreatic tumor microenvironment is highly immunosuppressive.

Figure 1.

Malignant cells (purple), fibroblasts, and myeloid cells in the tumor microenvironment secrete a variety of immunosuppressive cytokines and chemokines (pink) that prevent effective anti-tumor CD8 T cell responses. TAM, tumor-associated macrophage; Treg, Foxp3+ regulatory T cell; MoMDSC, monocytic myeloid-derived suppressor cell; GrMDSC, granulocytic myeloid-derived suppressor cell; iCAF, inflammatory cancer-associated fibroblast; myCAF, myofibroblastic cancer-associated fibroblasts.

Cellular components of immunosuppression in PDAC

Myeloid cells

A widespread strategy of immune escape used by cancer cells is induction of an altered state of myelopoiesis via secretion of metabolites, cytokines, and chemokines. These factors promote the accumulation of suppressive myeloid cells in the TME that are capable of inhibiting anti-tumor T cell responses 3, 4. Intratumoral suppressive myeloid cells are broadly classified as (i) myeloid-derived suppressor cells (MDSCs; defined as CD11b+ Gr-1+ in mice and CD11b+CD33+ in humans), which are comprised of myeloid progenitors and immature mononuclear cells, and (ii) tumor-associated macrophages (TAMs; defined as CD11bLow F4/80+ Gr-1 in mice and HLA-DR+ CD68+ in humans) that in contrast are mature and can be derived from either embryonic- or hematologic-origins 57.

In mice, intratumoral myeloid cells are the most common suppressive cells and therefore are an important therapeutic target. In both genetically-engineered mouse models (GEMM) and orthotopic mouse models of PDAC, TAMs account for roughly 15–20% of the total cellular tumor mass, both in primary tumors and metastatic sites, while MDSCs account for 5–10% 8, 9. Tumor cell-derived secreted factors, namely GM-CSF and CCL2 for MDSCs 8, 9 and CSF1 8 and BAG3 10 for TAMs, are critical determinants of the recruitment, polarization, and induction of intratumoral myeloid cell suppressive function. GM-CSF production has been shown to be dependent on oncogenic Kras, thereby directly linking oncogenic driver-dependency to myeloid cell suppression 11. GM-CSF can also be produced by cancer-associated mesenchymal cells to drive macrophage polarization within tumors and to support tumor growth, invasion, and metastatic potential 12. Both MDSCs and TAMs have been shown to directly induce T cell suppression through secreted factors, as well as indirectly by inducing tumor cell-specific PDL1 expression to inhibit spontaneous anti-tumor immunity 13.

While myeloid cells have demonstrated clear suppressive potential within tumors, their role in regulating immune suppression in PDAC may not be restricted to the tumor microenvironment. In support of this, depletion of macrophages residing outside of tumors has been shown to promote CD4+ T cell infiltration and, in combination with immunotherapy, to induce CD8+ T cell-dependent immune responses 14. As selective depletion of macrophages within tumors is technically challenging, it remains unclear from preclinical modeling whether myeloid cells disrupt productive T cell immunosurveillance in PDAC through suppressive mechanisms that are solely active within tumors, within secondary lymphoid organs, or both. Nonetheless, preclinical findings support the notion that targeted myeloid cell depletion can unmask endogenous T cell immunity 8, 15. Additionally, myeloid cell depletion has been shown to sensitize tumors to systemic therapies, including chemotherapy 8, radiation 16, checkpoint blockade 17, and chemoimmunotherapy 14.

Comparable roles for myeloid cells in PDAC tumor biology have been implicated in humans. In PDAC patients, mobilization of bone marrow (BM) derived inflammatory monocytes is increased, with lower inflammatory monocyte frequencies detected in the BM and greater frequencies observed in the periphery as compared to healthy controls 18. Furthermore, both CD66b+ MDSCs and CD68+ TAMs infiltrate human tumors similar to what is seen in mice, and the intratumoral density of both cell types correlates with poor survival 19. Similar to murine data, GM-CSF is also implicated as a key driver of tumor cell-derived altered myelopoiesis in humans. For instance, GM-CSF induces monocyte differentiation into MDSCs, thereby inhibiting T cell function in vitro 20. Collectively, these data demonstrate that altered myelopoiesis and myeloid cell-derived immunosuppression are key immune regulatory pathways operative in PDAC.

T cells

PDAC has been classically described as a “cold tumor” in both mice and humans, as it is characterized by a relative paucity of intratumoral CD8+ T cells. However, emerging evidence has now shown significant heterogeneity in intratumoral T cell infiltrates detected in both mice 21 and humans 19, 22, with the clear association between intratumoral effector T cell densities and improved outcomes bolstering the rationale to pursue immunotherapy in PDAC. Despite this, the majority of PDAC tumors show T cell populations skewed toward suppressive CD4+ T cell lineages. In GEMM of PDAC, CD4+ T cells account for <5% of all intratumoral cells 23. Foxp3+ regulatory T cells (Tregs) are recruited early during PDAC development and account for approximately 25% of all CD4+ T cells in both pancreatic intraepithelial neoplasia (PanIN) as well as invasive PDAC 23. In orthotopic models of PDAC, Treg depletion elicits effective CD8+ T cell-dependent anti-tumor immunity 24. However, Treg depletion in spontaneously arising PDAC induces only a modest infiltration of conventional CD4+ T cells without impacting CD8+ T cell recruitment and, as such, is insufficient to restore productive T cell immunosurveillance 25. Thus, while these data suggest a role for Tregs in shaping the immune response to PDAC, they also imply that additional immunoregulatory mechanisms exist to inhibit productive immunosurveillance.

In treatment-naïve stage I-III human PDAC, Tregs are expanded nearly two-fold in the peripheral blood, accounting for 13% of all CD4+ T cells 26. Similarly, Tregs accumulate in neoplastic pancreatic tissue as compared to both inflammatory and non-inflammatory pancreatic tissues 19, 27, ultimately comprising nearly 20% of intratumoral CD4+ T cells 26. Intratumoral Tregs secrete suppressive cytokines, including TGFβ and IL-10, and can suppress CD8+ T cell proliferation and IFNγ production. Moreover, intratumoral Treg density correlates with lymph node metastases and poor survival 19, 27.

Other T cell lineages have also been detected in PDAC, with predominantly protumorigenic effects. Gamma delta T cells can inhibit classical CD3+ alpha-beta T cells through secreted and ligand-dependent mechanisms 28, 29. Th17 cells have been shown to have the potential for both pro- and anti-tumorigenic effects, differences which are likely model-dependent. Th17 cell infiltration into subcutaneous murine PDAC tumors (Pan02) was associated with delayed tumor growth and survival in the setting of tumor-derived IL-6 30. In contrast, oncogenic Kras has been shown to induce Th17 cell infiltration into PanIN lesions and promote PDAC progression in an IL17-dependent manner 28. IL17-dependent PDAC progression has similarly been reported in other preclinical studies 31, and pharmacologic blockade of IL17 has demonstrated anti-tumor activity 28, 31. Together, these findings illustrate the remarkable ability of PDAC to orchestrate a microenvironment enriched in suppressive T cell populations, with few effector T cells.

B cells

B cells are a more recently studied cell type in PDAC and have been linked to the suppressive cellular PDAC TME. CD19+ B cells account for nearly 20% of CD45+ immune cells in PanINs 32 and 7% in PDAC 33. B cell recruitment to PDAC tumors is dependent on tumor cell production of CXCL13 34 as well as the B cell and myeloid cell-specific kinase Bruton’s Tyrosine Kinase (BTK) 33. Pancreatic ductal cell-specific production of the hypoxia-inducible factor (HIF) 1α can restrict B cell infiltration into tumors 32. Functionally, studies using genetic and pharmacologic depletion 33, 34 and augmentation 32 have shown that B cells promote PDAC tumorigenesis. Similar to mice, human PDAC tumors can also be infiltrated by B cells 22. However, the clinical significance of B cells in human PDAC remains ill-defined.

Stromal cells

PDAC is characterized by dense desmoplasia, which stems from activated pancreatic stellate cells (PSCs) that produce collagens, laminin, and fibronectin. These desmoplastic components of the stroma are classically thought to be immunosuppressive. For instance, PSCs can promote MDSC differentiation in humans 35. Consistent with this, Fibroblast Activation Protein-α (FAP) expressing stromal cells were found to inhibit immune-dependent PDAC growth in a subcutaneous model 36. PSCs were subsequently shown to induce CD8+ T cell sequestration in a CXCL12-dependent manner 37, 38. Interrupting the CXCL12-CXCR4 axis results in greater intratumoral densities of CD8+ T cells and rapid treatment responses in combination with immune checkpoint blockade 37. However, elements of the stroma may also act to restrain, rather than support PDAC growth 39. Depletion of α-SMA+ myofibroblasts in GEMM mice decreases effector T cell frequency while increasing regulatory T cell frequency. This finding suggests that myofibroblasts may have some immunostimulatory capacity 40. However, paradoxically, myofibroblast depletion was reported to sensitize GEMM mice to CTLA-4 blockade 40. These studies demonstrate that the immunomodulatory effects of stromal cells in the PDAC TME are complex, with cell-type and context-specific biology. Understanding these cellular networks remains an active area of investigation.

Molecular components of immunosuppression in PDAC

Beyond cellular immunosuppression, tumor cell-derived molecular factors have also been shown to suppress spontaneous anti-tumor immunity. Human PDAC cells can hyperactivate Focal Adhesion Kinase (FAK), a non-receptor tyrosine kinase whose activation is correlated with higher fibrosis and poor CD8+ T cell infiltration 41. In GEMM mice, inhibition of FAK activation reduces intratumoral fibrosis, decreases the accumulation of intratumoral myeloid suppressive cells, limits tumor progression, and sensitizes PDAC to PD-1 inhibition 41. Additional networks of tumor cell-derived, kinase-mediated suppression of adaptive immunity through macrophages have also been reported. For example, Receptor Interacting Protein (RIP)1 and RIP3 are key mediators of necroptosis, a form of caspase-8 independent, non-apoptotic, organized cellular necrosis seen in PDAC. Intact RIP1/RIP3 signaling can promote PDAC oncogenesis through CXCL1 ligation of its receptor, Mincle, which is expressed on tumor-infiltrating myeloid cells 42.

Extracellular matrix components such as hyaluronic acid can collapse intratumoral vasculature due to elevated interstitial fluid pressures, potentially creating a physical barrier to the delivery of chemotherapy and immunotherapeutics 43, 44. It is notable, though, that monocytes and neutrophils effectively infiltrate pancreatic tumors. While some intratumoral myeloid cells are derived from local proliferation of embryonically derived macrophages, more than half are continually replenished from the circulation 7. Post-capillary venules, where immune cells can extravasate, tend to be located on the periphery of pancreatic tumors, suggesting that myeloid cells are capable of penetrating the tumor microenvironment and do so more effectively than lymphocytes 45. T cell accumulation in pancreatic tumors is impeded by numerous factors, including extratumoral macrophages, expression of FASL on vascular endothelial cells, and TGFβ signaling 14, 46, 47. T cell function and accumulation can also be augmented by angiotensin system inhibitors, which result in increased blood vessel diameter and improved perfusion of pancreatic tumors 4850. Increased perfusion likely increases oxygen and glucose availability to T cells in pancreatic tumors.

Several metabolic pathways can also impede T cell function in tumors. For example, intratumoral myeloid cells produce the enzyme IDO, which converts tryptophan to kynurenine 51. Tryptophan is an essential amino acid for T cell function, whereas kynurenine supports immunosuppression. Similarly, extracellular ATP derived from dying cells is converted by CD39 and CD73 to adenosine, which is supportive of the immune suppressive activity of Tregs and can inhibit effector T cell function 52. Overall, there are likely multiple metabolic networks mediating suppression of adaptive immunity in PDAC 53.

3. T cell antigens in pancreatic cancer

A priority in the field of PDAC immunotherapy is to develop strategies for unleashing the anti-tumor potential of endogenous effector T cells. These strategies rely on the presence of immunogenic tumor antigens. Tumor antigens capable of eliciting spontaneous, cancer-specific T cell responses ideally are selectively expressed in cancer cells, with relatively little expression in normal cells. Here, we focus on two major categories of antigens: shared antigens derived from differential cancer cell-specific expression and antigens resulting from mutations or rearrangements in gene-coding sequences (mutational neoantigens).

The study of tumor-specific T cells in PDAC using preclinical modeling has relied on multiple immunocompetent mouse models, of which the most broadly studied is the KPC GEMM of PDAC. In KPC mice, Cre-Lox technology is used to induce pancreatic neoplasia via conditional expression of mutant Kras and Trp53 (Pdx-1-Cre;LSL-KrasG12D/+;LSL-Trp53R172H/+) 54. KPC mice develop spontaneous, stepwise, pre-invasive to invasive PDAC that recapitulates both histologic and immunologic features of human PDAC 55. Cell lines derived from KPC mice have been used for development of orthotopic, subcutaneous, and metastatic models of PDAC 56. In most cases, the cell lines used in these transplantation models reproduce many of the elements of the stromal architecture and immune infiltration that is seen in spontaneously arising tumors. However, differences can exist as illustrated by the capacity of some forms of immunotherapy to induce T cell-dependent tumor responses in implantation models but not in the spontaneous KPC model 14. Thus, interpretation of preclinical findings investigating the immunobiology of PDAC needs to consider the model in which the findings were observed. However, both models have significant similarities to the immunobiology observed in humans, and no clear superior model with greater relevance to humans has emerged.

In the KPC model, the intratumoral T cell infiltrate has been shown to be significantly heterogeneous 5760. KPC cancer cells harbor a low mutational burden, with a nearly complete absence of predicted neoepitopes 21, 61. Neoantigens are therefore unlikely endogenous T cell targets in KPC mice, suggesting the presence of non-mutational tumor antigens 21. Consistent with this, spontaneous T cell responses to shared antigens in KPC mice have been reported 37. CD8+ T cells from KPC mice respond to tumors arising spontaneously in other KPC mice 37, demonstrating the presence of a spontaneous anti-tumor T cell response to shared antigens.

Multiple shared antigens have been studied in pancreatic cancer, including telomerase, Muc1, enolase, WT1, Kras, mesothelin, and others. Telomerase and Muc1 responses have both been demonstrated in pancreatic cancer, which may also be signs of early transformation across epithelial cell types 62, 63. DNA vaccination against enolase in mice can induce a polyfunctional adaptive immune response capable of preventing tumor formation 64. WT1 is also overexpressed in human PDAC, 65 and WT1-specific T cells have been shown to kill PDAC cell lines in vitro 66. In a phase-I clinical trial, WT1-peptide vaccination was shown to be safe and associated with disease stability in some patients 67. Similarly, endogenous mesothelin-specific CD4+ and CD8+ T cells have been detected in PDAC patients, 68, 69 and these cell populations can be subsequently boosted in the setting of therapeutic vaccination 7072. Immunotherapeutic targeting of mesothelin can also delay tumor outgrowth in KPC mice 73. Cancer germline antigen expression has also been detected in PDAC, with both antibody and T cell responses to cancer germline antigens detected in PDAC patients 74, 75. Overall, it is likely that multiple shared antigens may be targets for T cell immune responses in PDAC. However, whether a dominant shared antigen(s) directs the heterogeneous endogenous T cell infiltrate remains unclear. It is, however, recognized from PDAC orthotopic tumor models that T cell exclusion in tumors arising in the pancreas can be overcome by introducing a strongly immunogenic antigen (e.g., ovalbumin), which effectively restricts successful engraftment of KPC tumors in a T cell-dependent manner 61. However, the consequences of conditional expression of a highly immunogenic antigen on T cell immunosurveillance in an established tumor in PDAC remains unknown.

Although self-antigens are more universal T cell targets for human PDAC, neoantigens remain important. The prevailing belief is that neoantigen formation is a stochastic process 76, and therefore tumors with higher mutational loads generate more neoantigens. Extending this logic, early reports posited that neoantigens were unlikely to be T cell antigens in human PDAC, owing to a relatively low somatic mutational prevalence 76. However, these estimates have been confounded by stromal contamination, as more recent sequencing efforts utilizing techniques to maximize tumor DNA capture have identified a higher burden of somatic mutations 77, 78. Moreover, while some reports show no clear associations between putative neoantigen load, activated T cell infiltrates, and survival in human PDAC 79, 80, recent findings demonstrate substantial heterogeneity in intratumoral T cell infiltrates, with neoantigens possibly serving as T cell targets in select subgroups 22. To this end, PDACs harboring double-strand break repair and mismatch repair mutational signatures show increased expression of markers of adaptive immune activation and are correlated with a higher neoantigen frequency 81. Neoantigens have also been identified as T cell targets in rare long-term PDAC survivors 22. Tumors from long-term PDAC survivors show a 12-fold greater density of cytolytic CD8+ T cells. Further, a neoantigen quality fitness model, integrating clonal genealogy, epitope homology, and T cell receptor affinity, was prognostic of survival in two independent PDAC datasets 22. Hence, spontaneous T cell responses in mouse models and human PDAC show substantial heterogeneity, although the presence of neoantigens serving as T cell targets in PDAC patient subsets remains an important distinction between the autochthonous mouse model and humans.

4. Key insights from preclinical models

Pancreatic cancer is an immunologic outlier

Clinicians have long appreciated the unusual difficulties posed by pancreatic cancer, and many of these same challenges exist in the KPC GEMM 56. Unfortunately, many immunotherapy approaches that are promising in other cancer types have shown little effect in PDAC 82. These agents include: IL-2, oncolytic viruses, checkpoint blockade, TGFβ inhibitors, neoantigen vaccines, Treg depletion, and CD47 blockade 83. While it is possible that these agents can be incorporated into combination therapies, their failures as single agents reinforce the idea that multiple immunologic barriers regulate productive immunosurveillance in PDAC. Even agents with moderate efficacy, such as single chemokine receptor antagonists, stimulate the emergence of compensatory mechanisms 84, 85, reinforcing the notion that successful immunotherapy in PDAC will need to target multiple pathways.

Chronic inflammation drives tumor progression

Although epidemiologic studies in humans suggest a link between pancreatitis and PDAC, the mechanistic underpinnings by which chronic inflammation promotes tumor growth have been elucidated largely from mouse models. Indeed, pancreatitis is a critical determinant of PDAC development in mice genetically modified to conditionally express mutant Kras in the pancreas during adulthood 86, 87. From KPC mice, we have also learned that components of chronic inflammation, including IL-6, innate immune cells, and regulatory B cells, drive early tumorigenesis 32, 34, 88, 89. IL-13 converts inflammatory macrophages into tumor-promoting macrophages 90. CD4+ T cell production of IL-17 serves as a direct growth factor for IL-17R+ tumor cells 28. Further, CD4+ T cells contribute to early suppression of CD8+ T cell responses during tumor initiation, 91 and exclusion of T cells is maintained during later tumor stages by extratumoral F4/80+ macrophages 14.

Malignant cells play an active role in immune suppression

Kras activation in malignant cells drives malignant transformation and orchestrates immunosuppression in PDAC. In mice with inducible Kras targeted to the pancreas, myeloid-rich microenvironments form and neoplastic lesions develop in the setting of inflammation. However, silencing Kras expression induces regression of both the tumor and the microenvironment, implying that the microenvironment is dependent on sustained cues received from malignant cells 87, 92. To this end, pancreatic tumor cells can secrete cytokines and chemokines, including GM-CSF, CCL2, CXCL1, CXCL2, CXCL5, and others, which have established roles in the recruitment and differentiation of immunosuppressive myeloid cells. Of these, CXCL1 has recently been shown to be produced by tumor cells in preclinical models and to coordinate the recruitment of myeloid cells and the exclusion of T cells within tumors 21. In addition, the IRE-1/XBP-1 axis is important in regulating Major Histocompatibility Complex (MHC) expression and latency in micrometastatic lesions and can control T cell infiltration into metastatic lesions 93. Thus, malignant cells are, not surprisingly, master orchestrators of immune suppression.

T cell responses can be elicited by in situ vaccination

The goal of vaccination is to elicit priming of tumor-specific T cells, and perhaps the simplest and most effective vaccination strategies involve direct delivery of immune stimulatory agents into the tumor microenvironment to produce “in situ” vaccines 94, 95. Successful in situ vaccination requires both induction of tumor cell death and the presence of an adjuvant. Presumably, spontaneous induction of this vaccination process explains the tumor antigen-specific responses that are seen in a rare subset of PDAC patients with microsatellite instability high (MSI-high) tumors that are responsive to PD-1 blockade 96 and the generation of tumor-specific T cells detected in resected tumors from long-term PDAC survivors 22. However, the extent to which standard of care gemcitabine/nab-paclitaxel or FOLFIRINOX can elicit tumor antigen release and prime tumor-specific cell responses in PDAC remains ill-defined.

Local delivery of adjuvants, such as stimulator of interferon genes (STING) agonists and toll-like receptor (TLR) ligands, can potently activate dendritic cells, leading to both dendritic cell maturation and production of type I and type II IFNs 97. However, the choice of adjuvants may be critical, as some adjuvants that can stimulate T cell priming have also been associated with PDAC development in preclinical models 98, 99. As such, manipulating the immune reaction induced by an adjuvant may be critical to unleashing its tumor-suppressive potential. Pancreatic tumor cells often have innate inflammatory pathways activated at baseline due to expression of TLR7 and chromosomal damage that lead to STING pathway activation, and both of which promote tumor cell survival through increased NF-kB signaling 98, 100, 101. TGFβ blockade can enhance the efficacy of PD-1 therapy to invoke T cell-dependent anti-tumor responses in non-pancreatic tumor models 47, 102 and synergizes with radiation in other tumor types 103, 104. However, blockade of TGFβ signaling in pancreatic tumors can inhibit systemic immunity induced by anti-CD40 and radiation 105. Thus, manipulating elements of the tumor microenvironment may be a favorable approach for harnessing tumor-specific T cells in some settings, but this also may depend on the adjuvant, tumor type, or both.

Radiation has emerged as an immunostimulatory strategy for cancer therapy and is being combined with immunotherapy in PDAC (Table 1). In mouse models of PDAC, although radiation depletes CD8+ T cells and recruits tumor-promoting myeloid cells 106 via increased CCL2, blocking CSF1 released by malignant cells responding to radiation therapy inhibits radiation-induced myeloid suppression and invokes T cell-mediated anti-tumor responses 16. Radiation has also been shown to induce an increase in MHC class I expression on tumor cells and to synergize with anti-CD40, anti-PD1, and anti-CTLA4 107, 108. Several studies have shown that radiation can also broaden the oligoclonality of the T cell response to cancer, presumably by inducing T cell responses against a wider array of tumor antigens 108, 109. Thus, strategies that combine radiation with immune-stimulating interventions hold promise.

Table 1.

Select immunotherapy trials in PDAC

Treatment category Immunotherapy targets ClinicalTrials.gov Identifier
Radiation plus immunotherapy PD1, CTLA-4, radiotherapy NCT02639026
Cyclophosphamide, PD1, GVAX, radiotherapy NCT02648282, NCT0316379
PD1, radiotherapy NCT03245541
Vaccines plus immune checkpoint inhibitors CRS-207, PD1, CTLA-4, GVAX NCT03190265
Cyclophosphamide, GVAX, PD1 NCT02451982
Cyclophosphamide, GVAX, PD1, CSF1R NCT03153410
Cyclophosphamide, GVAX, CRS-207, PD1, IDO NCT03006302
Chemotherapy plus immune agonists and antagonists Chemotherapy, CD40 agonist, anti- PD1 NCT03214250
Anti-stromal drugs plus immunotherapy FAK, PD1 NCT02758587
FAK, PD1, Gemcitabine NCT02546531
Matrix-targeted drugs plus immunotherapy PEGPH20, PD1 NCT03481920, NCT03193190, NCT03634332
Myeloid inhibitors plus immunotherapy PDL1, CFS1R NCT02777710
PD1, CSF1R NCT02526017
Cyclophosphamide, GVAX, PD1, CSF1R NCT03153410
Nab-paclitaxel, gemcitabine, BTK NCT02436668
Nab-paclitaxel, gemcitabine, CCR2/5, PD1 NCT03496662
PARP inhibitor plus immunotherapy PARP, PD1, CTLA-4 NCT03404960

BTK: Bruton’s Tyrosine Kinase; CRS-207: live attenuated listeria-encoding human mesothelin vaccine; CSF-1R: Colony Stimulating Factor 1 Receptor; CTLA-4: Cytotoxic T-lymphocyte-associated Antigen 4; CCR2/5: Chemokine (C-C motif) Receptor 2 and 5; FAK: Focal Adhesion Kinase; GVAX: GM-CSF transduced allogeneic whole tumor cell vaccine; IDO: Indoleamine 2,3-dioxygenase; PARP: poly ADP ribose polymerase; PD-1 : Programmed Cell Death Protein 1; PD-L1 : Programmed Death-Ligand 1; PEGPH20: pegvorhyaluronidase alfa.

Augmenting T cell priming is important

T cells express both co-inhibitory and co-stimulatory receptors, and augmentation of the latter represents an important avenue for pancreatic cancer, where boosting co-stimulation may compensate for deficits in antigen quality or quantity 110. Most co-stimulatory receptors are members of the TNF receptor superfamily and signal via NF-κB. Augmentation of canonical or non-canonical NF-κB signaling in immune cells can potently augment anti-tumor immunity 111, 112. Agonistic antibodies to co-stimulatory receptors have also been developed, and of these anti-CD40 has shown promise in pancreatic cancer 113. CD40 is a TNF receptor family member expressed on professional antigen presenting cells. When engaged by CD40L or by an agonistic antibody, CD40 signaling induces expression of co-stimulatory ligands, such as B7–1 and B7–2, production of IL-12 and other cytokines, enhanced antigen presentation, and, in the case of dendritic cells, upregulation of CCR7 and trafficking to draining lymph nodes. Agonistic antibodies to CD40 as monotherapy generate limited responses in both mice and humans with pancreatic tumors. In mice, combining CD40 agonists with chemotherapy can enhance T cell priming, producing T cell-dependent anti-tumor responses, and in other cases modulate the activation of myeloid cells to condition tumors for increased sensitivity to cytotoxic therapies 114117. Together, these data imply that restoring productive immunosurveillance in cancer may require immune stimulation and not just disrupting elements of immune suppression.

Generating T cells is not enough

Adoptive cell therapy obviates the need for endogenous T cell responses and allows for a setting to study the anti-tumor potential of tumor-reactive T cells. In acute lymphoblastic leukemia, CAR-T cells that target CD19 produce remarkable clinical responses. However for patients with PDAC, significant clinical activity with CAR-T cells has not yet been appreciated 118. Nonetheless, in mouse models, CAR-T cells directed against CEA, Her2/neu, and CD24 induce clearance of cognate antigen-positive tumor cells, with increased activity seen when Her2/neu and CD24 CAR-T cells are delivered in sequence 119, 120. Intratumoral delivery of a cytokine-armed oncolytic adenovirus can also enhance the efficacy of CAR T cells in implantation models of PDAC 121. Consistent with a role for the microenvironment in defining T cell fate, activated tumor-reactive T cells expressing a mesothelin-specific T cell receptor were found to effectively infiltrate PDAC tumors in the KPC GEMM. However, within tumors, T cells demonstrate poor persistence and rapidly upregulate several co-inhibitory receptors 73. As these therapeutic approaches are limited by the quality and uniform expression of antigenic targets, the potential for adoptive cell therapy in PDAC may ultimately be dependent on the capacity to generate productive polyfunctional endogenous T cell responses 122. Nonetheless, data from both mouse and human trials suggest that transferring tumor-specific T cells alone will be inadequate for inciting deep and durable remissions.

Myeloid and stromal immunosuppression is both targetable and interconnected with T cells

Embryonically derived, tissue-resident macrophages can expand in pancreatic cancer 7, complementing a population of short-lived myeloid cells that are sustained by continual recruitment from the circulation 123. CCR2 and CXCR2 are major chemokine receptors expressed by monocytes and neutrophils, respectively, and are responsible for their trafficking into tissues. Blocking each receptor has demonstrated efficacy in mouse models, and there is preliminary evidence of activity in human clinical trials 18, 124, 125. However, derailing the recruitment of CCR2+ myeloid cells produces compensatory infiltration of CXCR2+ neutrophils into tumors and vice versa, such that blockade of both chemokine receptors is more effective than either alone 84.

The phenotype and maintenance of tumor-infiltrating myeloid cells are determined by cues received within the tumor microenvironment. For instance, CSF1/CSF-1R is an important survival signal for resident macrophages. Blocking CSF-1R in vivo can produce a loss of intratumoral macrophages in PDAC leading to improved anti-tumor responses with immune checkpoint inhibitors (e.g., anti-CTLA-4 and anti-PD-1) 17. This finding suggests that effective T cell responses in PDAC may be generally curtailed by immunosuppressive myeloid cells, a finding that has now been seen in several preclinical settings 13, 41, 84, 110.

Cytokines are potent immune modulators

Cytokines generally act in an autocrine or paracrine fashion and demonstrate short half-lives necessitating local or sustained delivery 126. In PDAC models, exogenous application of IL-12 or IFNγ can reprogram myeloid cells, induce T cell responses, and lead to tumor regression 44, 127. Other Th1 family cytokines and chemokines can have similar effects, and strategies to deliver or induce these responses in vivo are under active investigation. On the other hand, cytokines can also be potently immunosuppressive. For example, IL-6, TGFβ, and IL-10 have all been implicated in PDAC progression 89. Of these, IL-6 may be the most problematic given its ability to activate fibroblasts and induce production of extracellular matrix 128. Indeed, blockade of IL-6 in KPC mice has shown therapeutic benefit when combined with gemcitabine, 129 and in BRCA-deficient KPC mice when combined PD-1 blockade 130.

Cancer associated fibroblasts (CAFs) regulate tumor-infiltrating T cells. CAFs secrete several factors that can negatively impact the immune microenvironment in tumors, including myeloid-recruiting chemokines and inflammatory cytokines, notably IL-6 131. CAF-derived extracellular matrix can be a physical barrier to drug delivery in PDAC 43, 132, but its role in PDAC progression has been debated. Stromal targeting agents, such as PEGylated hyaluronidase combined with gemcitabine, may reduce MDSC populations and increase CD8+ T cell infiltrates in KPC mice. Inhibition of FAK has also been found to reduce collagen and other extracellular matrix components in mouse models of PDAC. By suppressing tumor-derived chemokine production, FAK inhibitors can reduce myeloid cell accumulation, leading to T cell-dependent tumor regression in the setting of chemotherapy and checkpoint blockade 41. Together, these studies highlight the interconnected nature of stroma, myeloid cells, and T cells, and demonstrate that each component may be a critical determinant of therapeutic efficacy.

Effector activity by other immune cell types may be leveraged

Loss or downregulation of MHC class I molecules on malignant cells renders them invisible to CD8+ T cells and is a common explanation for acquired resistance to immunotherapy 133, 134. In contrast, NK cells recognize and destroy MHC-negative cells. However, NK cell abundance in pancreatic cancer is low and recruitment may be antagonized by malignant cell expression of the NK inhibitory ligand CD155 135. Nonetheless, NK cells have been implicated in the prevention of tumor recurrence after R0 resection in PDAC 136.

Other lymphocytes recruited to the PDAC TME are gamma delta T cells, which are activated by nonclassical MHC molecules. Gamma delta T cells infiltrate human PDAC, and their depletion in orthotopic mouse models of PDAC results in smaller tumor size and delayed tumor progression 29. Although these innate lymphocytes appear to be tumor-promoting, their baseline abundance and ability to secrete inflammatory cytokines raises the possibility that they might be repurposed for therapeutic benefit.

Myeloid cells can be directly tumoricidal 114. Macrophages can also acquire anti-fibrotic properties and, as a result, condition tumors with enhanced sensitivity to chemotherapy 116. Neutrophils, eosinophils, and mast cells produce reactive oxygen species and proteases that are effective at destroying tissue, yet it is unclear how best to harness this destructive prowess for control of tumor growth. One potential avenue may involve Tuft cells, which are chemosensory DCLK1+ cells. In the intestine, DCLK1+ Tuft cells proliferate in response to parasites, secrete IL-25, and initiate Th2 responses via IL-13 production from ILC2 cells 137. In pancreatic cancer, IL-17R+ DCLK1+ cells have been found in early PanIN lesions and may have a role in early establishment of the malignant microenvironment 138140. Whether these Tuft cells could be repurposed for therapeutic benefit is unknown.

5. Current status of I/O in human PDAC

For most patients with PDAC, immunotherapy has not demonstrated significant clinical activity 60. Early clinical studies investigating PD-1/PDL1 antagonists showed no activity in patients with PDAC, despite remarkable efficacy seen across a wide range of malignancies 141. Similar findings have been reported with CTLA-4 antagonists 142 and most recently with combining PD1 blockade with a small molecule inhibitor of indoleamine 2,3-deoxygenase (IDO) 143. The apparent lack of efficacy observed with checkpoint blockade in PDAC has now led to combination studies with chemotherapy under the premise that chemotherapy can be immunogenic 144, 145. However, to date, efforts to combine immune checkpoint blockade (i.e., anti-PD1 and anti-CTLA-4) with chemotherapy have not produced remarkable clinical activity in human PDAC beyond what would be expected with chemotherapy alone 146148.

The lack of efficacy seen with checkpoint blockade as monotherapy in PDAC has suggested the need to incorporate vaccine strategies to prime and activate tumor-specific T cells. To this end, numerous vaccine strategies have been evaluated, but unfortunately no vaccine approach by itself has demonstrated significant clinical benefit 60. This includes vaccines incorporating irradiated autologous tumor cells (e.g., GVAX, algenpantucel), live attenuated bacteria expressing mesothelin (CRS-207), and peptide-based vaccine approaches 149151. Despite this, correlative studies have shown that vaccines can stimulate tumor-specific T cells and significantly alter immune biology in resected tumors of some but not all patients. Specifically, it has been shown that GVAX, a GM-CSF gene-transfected, irradiated, allogeneic whole tumor cell vaccine, stimulates the formation of tertiary lymphoid aggregates in tumor tissue which correlate with improved outcomes for patients with surgically resected disease 71. GVAX vaccination can also impact the immune microenvironment of PDAC by stimulating an increase in T cell infiltrates and myeloid cell activation 57. To potentiate the activity of GVAX, cyclophosphamide (Cy) has been incorporated. At low doses (e.g., 250 mg/m2), Cy inhibits Tregs, and in preclinical models, pretreatment with low-dose Cy enhances anti-tumor activity induced by vaccination 152. In patients with unresectable PDAC, inclusion of Cy with GVAX also appears to augment the generation of tumor-specific T cells detected in the peripheral blood 153. Expansion of a diverse repertoire of mesothelin-specific T cells in the peripheral blood has been detected in some patients responding to Cy/GVAX/CRS-207 72. These T cell immune responses correlate with improved outcomes. Together, findings from these translational studies form the impetus for current investigations (Table 1) evaluating vaccines in combination with immune checkpoint blockade, which seek to prime T cells while also disrupting immunoregulatory mechanisms that may limit T cell effector activity.

The success of cancer immunotherapy in PDAC will rely on restoring multiple key steps involved in immune activation, as described in the cancer immunity cycle 154. Recent advances in synthetic biology offer the opportunity to engineer autologous T cells with tumor reactivity and, in doing so, restore steps of the cancer immunity cycle related to T cell priming and activation 118. This approach has been investigated in PDAC using autologous T cells engineered with a chimeric antigen receptor (CAR) that recognizes mesothelin, a self-antigen overexpressed on malignant cells 155. Whereas mesothelin-specific CAR T cells demonstrate potent anti-tumor activity in vitro against autologous tumor cells 156, infusion of CAR T cells into patients has shown limited clinical benefit 155. This lack of clinical efficacy implies that additional immunoregulatory barriers beyond antigen release and T cell priming will need to be addressed to realize the potential of immunotherapy in PDAC 157. It is noteworthy, though, that mesothelin-specific CAR T cells were observed to produce some anti-tumor activity in one patient 155. Specifically, in this patient there was a loss of FDG-uptake detected in liver lesions but not the primary lesion. This mixed treatment response suggests that distinct mechanisms of immune evasion may be present depending on the location of disease. In addition, these findings imply there is significant biological heterogeneity between lesions even in the same patient. Consistent with this hypothesis, remarkable treatment response heterogeneity has also been reported based on FDG-PET/CT imaging in newly diagnosed metastatic PDAC patients treated with chemotherapy and a CD40 agonist 115. Together, these observations imply that immunotherapy may need to be tailored based on the uniformity of treatment responses to accommodate intralesional heterogeneity. In this regard, a treatment would not be considered a failure if some lesions respond. Rather, mixed responses would signal a need to incorporate additional treatment regimens to capture non-responsive lesions rather than abandoning the initial therapeutic approach for lack of efficacy.

The limited clinical activity seen with immunotherapy in PDAC has produced a generalized belief that PDAC is immunologically “cold”. However, correlates from vaccine strategies suggest that the poor immunogenicity of pancreatic cancer may be reversible 71. Moreover, the notion that PDAC is unlikely to respond to immunotherapy is beginning to evolve. A new perspective is emerging based on findings in PDAC patients with microsatellite instable tumors, where responsiveness to PD-1/PDL1 antagonists can be observed 158. This also suggests that the promise of immunotherapy in PDAC may rely on precise patient selection. However, even some chemotherapy-refractory PDAC patients with microsatellite stable disease have demonstrated responses to PD-1 blockade when combined with CSF1R antibodies designed to deplete a subset of myeloid cells with immunosuppressive properties 159, 160. Thus, targeting elements of the tumor microenvironment may shift PDAC from an immunologically-resistant to immunologically-sensitive phenotype. Overall, these findings underscore the genetic and biological variability that exists in PDAC and indicate that non-cytotoxic therapies designed to harness the immune system may provide clinical benefit to at least a subset of patients. However, thus far this patient subset appears to be rare, representing less than 2% of all PDAC patients 161.

Efforts to broaden the impact of immunotherapy in PDAC are focusing on two major approaches. First, multi-targeted strategies are being examined to stimulate anti-tumor T cell responses. For example, an ongoing clinical study is evaluating a four-drug regimen involving chemotherapy (i.e., gemcitabine plus nab-paclitaxel) with an immune agonist targeting CD40 and an immune checkpoint inhibitor (i.e., anti-PD1) (Table 1). Other strategies are addressing roles for PD-1/PD-L1 blockade when combined with drugs that inhibit immunosuppressive mechanisms mediated by fibroblasts (e.g., FAK), matrix proteins (e.g., hyaluronidase), and myeloid cells (e.g., CSF1R, BTK CCR2, CXCR2) (Table 1). A second approach is examining immune-based strategies to condition tumors for enhanced responsiveness to chemotherapy. For example, disrupting the immune cell recruitment to tumors (e.g., CCR2 inhibitor) that occurs in the setting of cytotoxic stress has demonstrated promising activity in patients with locally advanced PDAC 124. In general, these two strategic approaches to applying immunotherapy in PDAC are being pursued in combination with chemotherapy as a therapeutic backbone.

While most studies applying immunotherapy to PDAC are currently building on chemotherapy, there remains little understanding around the immunogenicity and mechanisms of anti-tumor activity induced with chemotherapy in PDAC. A recent phase-II study evaluating gemcitabine plus nab-paclitaxel in combination with indoximod revealed that responders to treatment demonstrate increased CD8+ T cell density in on-treatment tumor biopsies 162. Moreover, all patients (both responders and non-responders) showed increases in the CD8+ effector to Foxp3+ regulatory T cell ratio in on-treatment biopsies (compared to baseline). These findings imply that chemotherapy may alter the immune microenvironment of PDAC and that these alterations may be important for therapeutic efficacy.

It remains unclear at this point, though, whether concurrent chemotherapy and immunotherapy will be the best therapeutic approach despite elegant studies in preclinical models demonstrating the potential of chemotherapy to be immunogenic 14, 144, 145, 163, 164. In this regard, unlike in most preclinical studies, chemotherapy is administered repeatedly to patients over the course of multiple cycles of therapy, raising the possibility that chemotherapy may ultimately attenuate the maintenance of any anti-tumor immune response by ablating proliferating immune cells and inducing T cell senescence 165. Chemotherapy can also be myelosuppressive, and thus while chemotherapy has the potential to ablate suppressive myeloid cells, it may also impact the efficacy of immunotherapeutic approaches designed to redirect myeloid cells with anti-tumor activity 116, 166, 167. An alternative approach to concurrent chemo-immunotherapy involves treatment sequencing as recently reported in non-small cell lung cancer where immunotherapy is initiated after chemotherapy induction 168. Such an approach is also now being evaluated in PDAC, wherein patients without progression on first-line platinum-based chemotherapy are switched to maintenance therapy with a PARP inhibitor and checkpoint immunotherapy (anti-CTLA-4 or anti-PD1) (Table 1). This approach to sequencing therapies represents a shift in the current treatment paradigm, which has historically been grounded on continuing treatment until progression (Figure 2).

Figure 2. Therapeutic sequencing versus treat-to-progression for PDAC.

Figure 2.

(A) Conceptual model showing treatment-naïve PDAC responding initially to first-line (1L) chemotherapy but then acquiring resistance on continued (cont’d) chemotherapy and ultimately manifesting as tumor progression. (B) Conceptual model showing treatment-naïve PDAC responding initially to 1L chemotherapy; chemotherapy is then discontinued prior to tumor progression. A series of sequential treatments (i.e., A, B, and C) are then initiated to coax the tumor into remission. Each treatment is designed to anticipate mechanisms of acquired resistance that emerge from the prior treatment.

With so many combinations of immunotherapies possible, there is a clear need for a framework for advancing immunotherapy in PDAC. It is likely that most attempts to stimulate productive anti-tumor immunity will be unsuccessful. Further, incorporation of immunotherapy into the treatment of PDAC has the potential to unveil an array of inflammatory-based side effects, 169 which can complicate management. For example, determining the etiology of colitis or hepatitis in the setting of immunotherapy is not straightforward and could reflect disease progression or infection in addition to immune activation induced by immunotherapy 170, 171. As inflammation is a key driver of PDAC, it also remains possible that immunotherapy may accelerate disease progression in some instances as has been recently suggested for a subset of patients with non-small cell lung cancer treated with PD-1 antagonists 172. Thus, it will be critical for studies which do not produce benefit or worsen outcomes to fail rapidly. However, it will be equally vital to learn from these efforts by including correlatives beyond safety assessments and pharmacokinetic assays 173 (Figure 3). Specifically, incorporating appropriate translational correlatives to inform both treatment responses and failures will be important. Well-designed clinical studies that are unsuccessful despite effective triggering of immune activity can provide key insights into compensatory mechanisms of resistance. For immunotherapy, these correlates will need to address at a minimum the capacity of the investigational agent to target the biological pathway of interest. For example, this might entail demonstration that a small molecule inhibitor blocks in vivo phosphorylation of the target protein of interest. However, additional pharmacodynamic assays probing immune activation and immune complexity within and outside of tumors will also be beneficial to understanding changes in immune and tumor biology that may inform appropriate treatment combinations, sequencing, or both. Fundamentally, there is strong value in learning from every patient and understanding their immune status to guide the design of subsequent clinical trials in a strategic manner.

Figure 3. Workflow for advancing therapeutic strategies in PDAC.

Figure 3.

Patients enrolled in a clinical study are monitored with repeat liquid and tissue biopsies beginning at baseline prior to treatment and at defined time points on-treatment. Responders (partial or complete response) and non-responders (stable or progressive disease) are defined early after treatment (within 2–4 cycles) based on radiologic imaging (CT, MRI, or PET/CT). Sample biopsies are interrogated in the laboratory to assess pre-defined biomarkers of biological response and their relationship to clinical response. Pharmacodynamic markers in blood and tissue that are defined a priori are used to understand mechanisms of response and resistance to therapy. Findings are then used to inform development of new preclinical data and scientific rationale for the conduct of subsequent clinical studies.

6. Conclusions

The incidence of PDAC is rising – this, coupled with a >90% mortality despite our best current treatments, accentuates the urgent need to find effective therapies. Long-term remissions with current standard treatments of chemotherapy, surgery, and targeted therapies remain rare, underscoring the need for novel approaches. Immunotherapy is a promising new therapeutic strategy for PDAC given the remarkable and durable outcomes observed in many other treatment-refractory cancers, including in patients that have failed multiple standard treatments. Burgeoning evidence has now confirmed in humans that despite lower T cell density in PDAC tumors compared to more immunogenic counterparts, T cell recognition of tumors is associated with improved survival. Moreover, checkpoint blockade has demonstrated efficacy in some PDAC patients with microsatellite instability. Together, these clinical observations are the best testaments to the potential of immune-based therapies in PDAC and argue against organ site-specific resistance to immunotherapy in the pancreas, while also providing proof-of-concept for PDAC treatment with immunotherapy. However, it is clear that PDAC is uniquely characterized by multiple redundant barriers to immunotherapy, and effective therapies likely will require combination approaches with current standard agents, many of which are currently in clinical testing. These clinical trials are expected to uncover fundamental concepts critical to the development of new therapies for PDAC, with preclinical models providing key insights into the mechanistic underpinnings underlying these observations, and thus are essential for propelling the much-needed progress in PDAC treatment. Through concerted and coordinated efforts between scientists, clinicians, and patients to identify the most efficacious strategies, we anticipate immunotherapy to become a key component of future PDAC therapies.

Acknowledgments

Financial support: This work was supported by National Institutes of Health grants R01 CA197916 (G.L.B.), U01 CA224193–01 (G.L.B.), K12 CA184746–01A1 (V.P.B), U01 CA224175–01 (V.P.B), Damon Runyon Clinical Investigator Award (V.P.B), the Richard and Susan Smith Family Foundation (S.K.D), the Hale Center for Pancreatic Cancer Research (S.K.D), the Harvard-MIT Bridge Project (S.K.D), and NIH U01 CA224146–01 (S.K.D).

Biography

graphic file with name nihms-1526872-b0001.gif

graphic file with name nihms-1526872-b0002.gif

graphic file with name nihms-1526872-b0003.gif

Footnotes

Conflict of interest statement: The authors have no competing financial interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.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] [PubMed] [Google Scholar]
  • 2.Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, Neoptolemos JP. Pancreatic cancer. Nat Rev Dis Primers 2016;2:16022. [DOI] [PubMed] [Google Scholar]
  • 3.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 2017;14:399–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest 2015;125:3365–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest 2015;125:3356–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhu Y, Herndon JM, Sojka DK, Kim KW, Knolhoff BL, Zuo C, Cullinan DR, Luo J, Bearden AR, Lavine KJ, Yokoyama WM, Hawkins WG, Fields RC, Randolph GJ, DeNardo DG. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017;47:323–338 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo DG. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res 2013;73:1128–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012;21:822–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rosati A, Basile A, D’Auria R, d’Avenia M, De Marco M, Falco A, Festa M, Guerriero L, Iorio V, Parente R, Pascale M, Marzullo L, Franco R, Arra C, Barbieri A, Rea D, Menichini G, Hahne M, Bijlsma M, Barcaroli D, Sala G, di Mola FF, di Sebastiano P, Todoric J, Antonucci L, Corvest V, Jawhari A, Firpo MA, Tuveson DA, Capunzo M, Karin M, De Laurenzi V, Turco MC. BAG3 promotes pancreatic ductal adenocarcinoma growth by activating stromal macrophages. Nat Commun 2015;6:8695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012;21:836–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Waghray M, Yalamanchili M, Dziubinski M, Zeinali M, Erkkinen M, Yang H, Schradle KA, Urs S, Pasca Di Magliano M, Welling TH, Palmbos PL, Abel EV, Sahai V, Nagrath S, Wang L, Simeone DM. GM-CSF Mediates Mesenchymal-Epithelial Cross-talk in Pancreatic Cancer. Cancer Discov 2016;6:886–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhang Y, Velez-Delgado A, Mathew E, Li D, Mendez FM, Flannagan K, Rhim AD, Simeone DM, Beatty GL, Pasca di Magliano M. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017;66:124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Beatty GL, Winograd R, Evans RA, Long KB, Luque SL, Lee JW, Clendenin C, Gladney WL, Knoblock DM, Guirnalda PD, Vonderheide RH. Exclusion of T Cells From Pancreatic Carcinomas in Mice Is Regulated by Ly6C(low) F4/80(+) Extratumoral Macrophages. Gastroenterology 2015;149:201–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Stromnes IM, Brockenbrough JS, Izeradjene K, Carlson MA, Cuevas C, Simmons RM, Greenberg PD, Hingorani SR. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut 2014;63:1769–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Seifert L, Werba G, Tiwari S, Giao Ly NN, Nguy S, Alothman S, Alqunaibit D, Avanzi A, Daley D, Barilla R, Tippens D, Torres-Hernandez A, Hundeyin M, Mani VR, Hajdu C, Pellicciotta I, Oh P, Du K, Miller G. Radiation Therapy Induces Macrophages to Suppress T-Cell Responses Against Pancreatic Tumors in Mice. Gastroenterology 2016;150:1659–1672 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, Wang-Gillam A, Goedegebuure SP, Linehan DC, DeNardo DG. 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]
  • 18.Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, Mitchem JB, Plambeck-Suess SM, Worley LA, Goetz BD, Wang-Gillam A, Eberlein TJ, Denardo DG, Goedegebuure SP, Linehan DC. Inflammatory Monocyte Mobilization Decreases Patient Survival in Pancreatic Cancer: A Role for Targeting the CCL2/CCR2 Axis. Clin Cancer Res 2013;19:3404–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ino Y, Yamazaki-Itoh R, Shimada K, Iwasaki M, Kosuge T, Kanai Y, Hiraoka N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br J Cancer 2013;108:914–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Takeuchi S, Baghdadi M, Tsuchikawa T, Wada H, Nakamura T, Abe H, Nakanishi S, Usui Y, Higuchi K, Takahashi M, Inoko K, Sato S, Takano H, Shichinohe T, Seino K, Hirano S. Chemotherapy-Derived Inflammatory Responses Accelerate the Formation of Immunosuppressive Myeloid Cells in the Tissue Microenvironment of Human Pancreatic Cancer. Cancer Res 2015;75:2629–40. [DOI] [PubMed] [Google Scholar]
  • 21.Li J, Byrne KT, Yan F, Yamazoe T, Chen Z, Baslan T, Richman LP, Lin JH, Sun YH, Rech AJ, Balli D, Hay CA, Sela Y, Merrell AJ, Liudahl SM, Gordon N, Norgard RJ, Yuan S, Yu S, Chao T, Ye S, Eisinger-Mathason TSK, Faryabi RB, Tobias JW, Lowe SW, Coussens LM, Wherry EJ, Vonderheide RH, Stanger BZ. Tumor Cell-Intrinsic Factors Underlie Heterogeneity of Immune Cell Infiltration and Response to Immunotherapy. Immunity 2018;49:178–193 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Balachandran VP, Luksza M, Zhao JN, Makarov V, Moral JA, Remark R, Herbst B, Askan G, Bhanot U, Senbabaoglu Y, Wells DK, Cary CIO, Grbovic-Huezo O, Attiyeh M, Medina B, Zhang J, Loo J, Saglimbeni J, Abu-Akeel M, Zappasodi R, Riaz N, Smoragiewicz M, Kelley ZL, Basturk O, Gonen M, Levine AJ, Allen PJ, Fearon DT, Merad M, Gnjatic S, Iacobuzio-Donahue CA, Wolchok JD, DeMatteo RP, Chan TA, Greenbaum BD, Merghoub T, Leach SD. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017;551:512–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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]
  • 24.Jang JE, Hajdu CH, Liot C, Miller G, Dustin ML, Bar-Sagi D. Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer. Cell Rep 2017;20:558–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bengsch F, Knoblock DM, Liu A, McAllister F, Beatty GL. CTLA-4/CD80 pathway regulates T cell infiltration into pancreatic cancer. Cancer Immunol Immunother 2017;66:1609–1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Liyanage UK, Moore TT, Joo HG, Tanaka Y, Herrmann V, Doherty G, Drebin JA, Strasberg SM, Eberlein TJ, Goedegebuure PS, Linehan DC. 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]
  • 27.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]
  • 28.McAllister F, Bailey JM, Alsina J, Nirschl CJ, Sharma R, Fan H, Rattigan Y, Roeser JC, Lankapalli RH, Zhang H, Jaffee EM, Drake CG, Housseau F, Maitra A, Kolls JK, Sears CL, Pardoll DM, Leach SD. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 2014;25:621–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Daley D, Zambirinis CP, Seifert L, Akkad N, Mohan N, Werba G, Barilla R, Torres-Hernandez A, Hundeyin M, Mani VRK, Avanzi A, Tippens D, Narayanan R, Jang JE, Newman E, Pillarisetty VG, Dustin ML, Bar-Sagi D, Hajdu C, Miller G. gammadelta T Cells Support Pancreatic Oncogenesis by Restraining alphabeta T Cell Activation. Cell 2016;166:1485–1499 e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gnerlich JL, Mitchem JB, Weir JS, Sankpal NV, Kashiwagi H, Belt BA, Porembka MR, Herndon JM, Eberlein TJ, Goedegebuure P, Linehan DC. Induction of Th17 cells in the tumor microenvironment improves survival in a murine model of pancreatic cancer. J Immunol 2010;185:4063–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wu HH, Hwang-Verslues WW, Lee WH, Huang CK, Wei PC, Chen CL, Shew JY, Lee EY, Jeng YM, Tien YW, Ma C, Lee WH. Targeting IL-17B-IL-17RB signaling with an anti-IL-17RB antibody blocks pancreatic cancer metastasis by silencing multiple chemokines. J Exp Med 2015;212:333–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lee KE, Spata M, Bayne LJ, Buza EL, Durham AC, Allman D, Vonderheide RH, Simon MC. Hif1a Deletion Reveals Pro-Neoplastic Function of B Cells in Pancreatic Neoplasia. Cancer Discov 2016;6:256–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gunderson AJ, Kaneda MM, Tsujikawa T, Nguyen AV, Affara NI, Ruffell B, Gorjestani S, Liudahl SM, Truitt M, Olson P, Kim G, Hanahan D, Tempero MA, Sheppard B, Irving B, Chang BY, Varner JA, Coussens LM. Bruton Tyrosine Kinase-Dependent Immune Cell Cross-talk Drives Pancreas Cancer. Cancer Discov 2016;6:270–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pylayeva-Gupta Y, Das S, Handler JS, Hajdu CH, Coffre M, Koralov SB, Bar-Sagi D. IL35-Producing B Cells Promote the Development of Pancreatic Neoplasia. Cancer Discov 2016;6:247–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mace TA, Ameen Z, Collins A, Wojcik S, Mair M, Young GS, Fuchs JR, Eubank TD, Frankel WL, Bekaii-Saab T, Bloomston M, Lesinski GB. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res 2013;73:3007–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010;330:827–30. [DOI] [PubMed] [Google Scholar]
  • 37.Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Connell CM, Roberts EW, Zhao Q, Caballero OL, Teichmann SA, Janowitz T, Jodrell DI, Tuveson DA, Fearon DT. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2013;110:20212–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, Marshall JF, Chin-Aleong J, Chelala C, Gribben JG, Ramsay AG, Kocher HM. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013;145:1121–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, Westphalen CB, Kitajewski J, Fernandez-Barrena MG, Fernandez-Zapico ME, Iacobuzio-Donahue C, Olive KP, Stanger BZ. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014;25:735–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014;25:719–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, Nywening TM, Hawkins WG, Shapiro IM, Weaver DT, Pachter JA, Wang-Gillam A, DeNardo DG. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med 2016;22:851–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Seifert L, Werba G, Tiwari S, Giao Ly NN, Alothman S, Alqunaibit D, Avanzi A, Barilla R, Daley D, Greco SH, Torres-Hernandez A, Pergamo M, Ochi A, Zambirinis CP, Pansari M, Rendon M, Tippens D, Hundeyin M, Mani VR, Hajdu C, Engle D, Miller G. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature 2016;532:245–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dougan M, Ingram JR, Jeong HJ, Mosaheb MM, Bruck PT, Ali L, Pishesha N, Blomberg O, Tyler PM, Servos MM, Rashidian M, Nguyen QD, von Andrian UH, Ploegh HL, Dougan SK. Targeting Cytokine Therapy to the Pancreatic Tumor Microenvironment Using PD-L1-Specific VHHs. Cancer Immunol Res 2018;6:389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Thiriot A, Perdomo C, Cheng G, Novitzky-Basso I, McArdle S, Kishimoto JK, Barreiro O, Mazo I, Triboulet R, Ley K, Rot A, von Andrian UH. Differential DARC/ACKR1 expression distinguishes venular from non-venular endothelial cells in murine tissues. BMC Biol 2017;15:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Motz GT, Santoro SP, Wang LP, Garrabrant T, Lastra RR, Hagemann IS, Lal P, Feldman MD, Benencia F, Coukos G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014;20:607–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, Kadel EE III, Koeppen H, Astarita JL, Cubas R, Jhunjhunwala S, Banchereau R, Yang Y, Guan Y, Chalouni C, Ziai J, Senbabaoglu Y, Santoro S, Sheinson D, Hung J, Giltnane JM, Pierce AA, Mesh K, Lianoglou S, Riegler J, Carano RAD, Eriksson P, Hoglund M, Somarriba L, Halligan DL, van der Heijden MS, Loriot Y, Rosenberg JE, Fong L, Mellman I, Chen DS, Green M, Derleth C, Fine GD, Hegde PS, Bourgon R, Powles T. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liu H, Naxerova K, Pinter M, Incio J, Lee H, Shigeta K, Ho WW, Crain JA, Jacobson A, Michelakos T, Dias-Santos D, Zanconato A, Hong TS, Clark JW, Murphy JE, Ryan DP, Deshpande V, Lillemoe KD, Fernandez-Del Castillo C, Downes M, Evans RM, Michaelson J, Ferrone CR, Boucher Y, Jain RK. Use of Angiotensin System Inhibitors Is Associated with Immune Activation and Longer Survival in Nonmetastatic Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2017;23:5959–5969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kumar V, Boucher Y, Liu H, Ferreira D, Hooker J, Catana C, Hoover AJ, Ritter T, Jain RK, Guimaraes AR. Noninvasive Assessment of Losartan-Induced Increase in Functional Microvasculature and Drug Delivery in Pancreatic Ductal Adenocarcinoma. Transl Oncol 2016;9:431–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, Stylianopoulos T, Mousa AS, Han X, Adstamongkonkul P, Popovic Z, Huang P, Bawendi MG, Boucher Y, Jain RK. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun 2013;4:2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Beatty GL, O’Dwyer PJ, Clark J, Shi JG, Bowman KJ, Scherle PA, Newton RC, Schaub R, Maleski J, Leopold L, Gajewski TF. First-in-Human Phase I Study of the Oral Inhibitor of Indoleamine 2,3-Dioxygenase-1 Epacadostat (INCB024360) in Patients with Advanced Solid Malignancies. Clin Cancer Res 2017;23:3269–3276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Maj T, Wang W, Crespo J, Zhang H, Wang W, Wei S, Zhao L, Vatan L, Shao I, Szeliga W, Lyssiotis C, Liu JR, Kryczek I, Zou W. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol 2017;18:1332–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lyssiotis CA, Kimmelman AC. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol 2017;27:863–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA. 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]
  • 55.Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, Kawaguchi Y, Johann D, Liotta LA, Crawford HC, Putt ME, Jacks T, Wright CV, Hruban RH, Lowy AM, Tuveson DA. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003;4:437–50. [DOI] [PubMed] [Google Scholar]
  • 56.Lee JW, Komar CA, Bengsch F, Graham K, Beatty GL. Genetically Engineered Mouse Models of Pancreatic Cancer: The KPC Model (LSL-Kras(G12D/+);LSL-Trp53(R172H/+);Pdx-1-Cre), Its Variants, and Their Application in Immuno-oncology Drug Discovery. Curr Protoc Pharmacol 2016;73:14 39 1–14 39 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tsujikawa T, Kumar S, Borkar RN, Azimi V, Thibault G, Chang YH, Balter A, Kawashima R, Choe G, Sauer D, El Rassi E, Clayburgh DR, Kulesz-Martin MF, Lutz ER, Zheng L, Jaffee EM, Leyshock P, Margolin AA, Mori M, Gray JW, Flint PW, Coussens LM. Quantitative Multiplex Immunohistochemistry Reveals Myeloid-Inflamed Tumor-Immune Complexity Associated with Poor Prognosis. Cell Rep 2017;19:203–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Carstens JL, Correa de Sampaio P, Yang D, Barua S, Wang H, Rao A, Allison JP, LeBleu VS, Kalluri R. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun 2017;8:15095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Stromnes IM, Hulbert A, Pierce RH, Greenberg PD, Hingorani SR. T-cell Localization, Activation, and Clonal Expansion in Human Pancreatic Ductal Adenocarcinoma. Cancer Immunol Res 2017;5:978–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Beatty GL, Eghbali S, Kim R. Deploying Immunotherapy in Pancreatic Cancer: Defining Mechanisms of Response and Resistance. Am Soc Clin Oncol Educ Book 2017;37:267–278. [DOI] [PubMed] [Google Scholar]
  • 61.Evans RA, Diamond MS, Rech AJ, Chao T, Richardson MW, Lin JH, Bajor DL, Byrne KT, Stanger BZ, Riley JL, Markosyan N, Winograd R, Vonderheide RH. Lack of immunoediting in murine pancreatic cancer reversed with neoantigen. JCI Insight 2016;1:e88328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bernhardt SL, Gjertsen MK, Trachsel S, Moller M, Eriksen JA, Meo M, Buanes T, Gaudernack G. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: A dose escalating phase I/II study. Br J Cancer 2006;95:1474–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lepisto AJ, Moser AJ, Zeh H, Lee K, Bartlett D, McKolanis JR, Geller BA, Schmotzer A, Potter DP, Whiteside T, Finn OJ, Ramanathan RK. A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther 2008;6:955–964. [PMC free article] [PubMed] [Google Scholar]
  • 64.Capello M, Cappello P, Linty FC, Chiarle R, Sperduti I, Novarino A, Salacone P, Mandili G, Naccarati A, Sacerdote C, Beghelli S, Bersani S, Barbi S, Bassi C, Scarpa A, Nistico P, Giovarelli M, Vineis P, Milella M, Novelli F. Autoantibodies to Ezrin are an early sign of pancreatic cancer in humans and in genetically engineered mouse models. J Hematol Oncol 2013;6:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, Abeno S, Ikeba A, Takashima S, Tsujie M, Yamamoto H, Sakon M, Nezu R, Kawano K, Nishida S, Ikegame K, Kawakami M, Tsuboi A, Oka Y, Yoshikawa K, Aozasa K, Monden M, Sugiyama H. Overexpression of the Wilms’ tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci 2004;95:583–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Takahara A, Koido S, Ito M, Nagasaki E, Sagawa Y, Iwamoto T, Komita H, Ochi T, Fujiwara H, Yasukawa M, Mineno J, Shiku H, Nishida S, Sugiyama H, Tajiri H, Homma S. Gemcitabine enhances Wilms’ tumor gene WT1 expression and sensitizes human pancreatic cancer cells with WT1-specific T-cell-mediated antitumor immune response. Cancer Immunol Immunother 2011;60:1289–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Koido S, Homma S, Okamoto M, Takakura K, Mori M, Yoshizaki S, Tsukinaga S, Odahara S, Koyama S, Imazu H, Uchiyama K, Kajihara M, Arakawa H, Misawa T, Toyama Y, Yanagisawa S, Ikegami M, Kan S, Hayashi K, Komita H, Kamata Y, Ito M, Ishidao T, Yusa S, Shimodaira S, Gong J, Sugiyama H, Ohkusa T, Tajiri H. Treatment with chemotherapy and dendritic cells pulsed with multiple Wilms’ tumor 1 (WT1)-specific MHC class I/II-restricted epitopes for pancreatic cancer. Clin Cancer Res 2014;20:4228–39. [DOI] [PubMed] [Google Scholar]
  • 68.Johnston FM, Tan MC, Tan BR Jr., Porembka MR, Brunt EM, Linehan DC, Simon PO Jr., Plambeck-Suess S, Eberlein TJ, Hellstrom KE, Hellstrom I, Hawkins WG, Goedegebuure P. Circulating mesothelin protein and cellular antimesothelin immunity in patients with pancreatic cancer. Clin Cancer Res 2009;15:6511–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen Y, Ayaru L, Mathew S, Morris E, Pereira SP, Behboudi S. Expansion of anti-mesothelin specific CD4+ and CD8+ T cell responses in patients with pancreatic carcinoma. PLoS One 2014;9:e88133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Thomas AM, Santarsiero LM, Lutz ER, Armstrong TD, Chen YC, Huang LQ, Laheru DA, Goggins M, Hruban RH, Jaffee EM. Mesothelin-specific CD8(+) T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients. J Exp Med 2004;200:297–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, Solt S, Dorman A, Wamwea A, Yager A, Laheru D, Wolfgang CL, Wang J, Hruban RH, Anders RA, Jaffee EM, Zheng L. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res 2014;2:616–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, Morse M, Zeh H, Cohen D, Fine RL, Onners B, Uram JN, Laheru DA, Lutz ER, Solt S, Murphy AL, Skoble J, Lemmens E, Grous J, Dubensky T Jr., Brockstedt DG, Jaffee EM. Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015;33:1325–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Stromnes IM, Schmitt TM, Hulbert A, Brockenbrough JS, Nguyen HN, Cuevas C, Dotson AM, Tan X, Hotes JL, Greenberg PD, Hingorani SR. T Cells Engineered against a Native Antigen Can Surmount Immunologic and Physical Barriers to Treat Pancreatic Ductal Adenocarcinoma. Cancer Cell 2015;28:638–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wadle A, Kubuschok B, Imig J, Wuellner B, Wittig C, Zwick C, Mischo A, Waetzig K, Romeike BF, Lindemann W, Schilling M, Pfreundschuh M, Renner C. Serological immune response to cancer testis antigens in patients with pancreatic cancer. Int J Cancer 2006;119:117–25. [DOI] [PubMed] [Google Scholar]
  • 75.Abate-Daga D, Speiser DE, Chinnasamy N, Zheng Z, Xu H, Feldman SA, Rosenberg SA, Morgan RA. Development of a T cell receptor targeting an HLA-A*0201 restricted epitope from the cancer-testis antigen SSX2 for adoptive immunotherapy of cancer. PLoS One 2014;9:e93321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. Volume 348, 2015:69–74. [DOI] [PubMed] [Google Scholar]
  • 77.Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Kakkar N, Zhao F, Wu YQ, Wang M, Muzny DM, Fisher WE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012;491:399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, Choti MA, Yeo CJ, McCue P, White MA, Knudsen ES. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun 2015;6:6744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Balli D, Rech AJ, Stanger BZ, Vonderheide RH. Immune Cytolytic Activity Stratifies Molecular Subsets of Human Pancreatic Cancer. Clin Cancer Res 2017;23:3129–3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bailey P, Chang DK, Forget MA, Lucas FA, Alvarez HA, Haymaker C, Chattopadhyay C, Kim SH, Ekmekcioglu S, Grimm EA, Biankin AV, Hwu P, Maitra A, Roszik J. Exploiting the neoantigen landscape for immunotherapy of pancreatic ductal adenocarcinoma. Sci Rep 2016;6:35848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Connor AA, Denroche RE, Jang GH, Timms L, Kalimuthu SN, Selander I, McPherson T, Wilson GW, Chan-Seng-Yue MA, Borozan I, Ferretti V, Grant RC, Lungu IM, Costello E, Greenhalf W, Palmer D, Ghaneh P, Neoptolemos JP, Buchler M, Petersen G, Thayer S, Hollingsworth MA, Sherker A, Durocher D, Dhani N, Hedley D, Serra S, Pollett A, Roehrl MHA, Bavi P, Bartlett JMS, Cleary S, Wilson JM, Alexandrov LB, Moore M, Wouters BG, McPherson JD, Notta F, Stein LD, Gallinger S. Association of Distinct Mutational Signatures With Correlates of Increased Immune Activity in Pancreatic Ductal Adenocarcinoma. JAMA Oncol 2017;3:774–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res 2015;21:687–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer 2012;12:237–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Nywening TM, Belt BA, Cullinan DR, Panni RZ, Han BJ, Sanford DE, Jacobs RC, Ye J, Patel AA, Gillanders WE, Fields RC, DeNardo DG, Hawkins WG, Goedegebuure P, Linehan DC. Targeting both tumour-associated CXCR2(+) neutrophils and CCR2(+) macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 2018;67:1112–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, Hashimoto A, Vonteddu P, Behera R, Goins MA, Mulligan C, Nam B, Hockstein N, Denstman F, Shakamuri S, Speicher DW, Weeraratna AT, Chao T, Vonderheide RH, Languino LR, Ordentlich P, Liu Q, Xu X, Lo A, Pure E, Zhang C, Loboda A, Sepulveda MA, Snyder LA, Gabrilovich DI. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017;32:654–668 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 2007;11:291–302. [DOI] [PubMed] [Google Scholar]
  • 87.Collins MA, Bednar F, Zhang Y, Brisset JC, Galban S, Galban CJ, Rakshit S, Flannagan KS, Adsay NV, Pasca di Magliano M. Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. J Clin Invest 2012;122:639–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Liou GY, Doppler H, Necela B, Krishna M, Crawford HC, Raimondo M, Storz P. Macrophage-secreted cytokines drive pancreatic acinar-to-ductal metaplasia through NF-kappaB and MMPs. J Cell Biol 2013;202:563–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhu Z, Aref AR, Cohoon TJ, Barbie TU, Imamura Y, Yang S, Moody SE, Shen RR, Schinzel AC, Thai TC, Reibel JB, Tamayo P, Godfrey JT, Qian ZR, Page AN, Maciag K, Chan EM, Silkworth W, Labowsky MT, Rozhansky L, Mesirov JP, Gillanders WE, Ogino S, Hacohen N, Gaudet S, Eck MJ, Engelman JA, Corcoran RB, Wong KK, Hahn WC, Barbie DA. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov 2014;4:452–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liou GY, Bastea L, Fleming A, Doppler H, Edenfield BH, Dawson DW, Zhang L, Bardeesy N, Storz P. The Presence of Interleukin-13 at Pancreatic ADM/PanIN Lesions Alters Macrophage Populations and Mediates Pancreatic Tumorigenesis. Cell Rep 2017;19:1322–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhang Y, Yan W, Mathew E, Bednar F, Wan S, Collins MA, Evans RA, Welling TH, Vonderheide RH, di Magliano MP. CD4+ T lymphocyte ablation prevents pancreatic carcinogenesis in mice. Cancer Immunol Res 2014;2:423–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, Lee JJ, Kalluri R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017;546:498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pommier A, Anaparthy N, Memos N, Kelley ZL, Gouronnec A, Yan R, Auffray C, Albrengues J, Egeblad M, Iacobuzio-Donahue CA, Lyons SK, Fearon DT. Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases. Science 2018;360: eaao4908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Sagiv-Barfi I, Czerwinski DK, Levy S, Alam IS, Mayer AT, Gambhir SS, Levy R. Eradication of spontaneous malignancy by local immunotherapy. Sci Transl Med 2018;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fonseca C, Dranoff G. Capitalizing on the immunogenicity of dying tumor cells. Clin Cancer Res 2008;14:1603–8. [DOI] [PubMed] [Google Scholar]
  • 96.Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA Jr. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol 2016;17:1142–9. [DOI] [PubMed] [Google Scholar]
  • 98.Ochi A, Graffeo CS, Zambirinis CP, Rehman A, Hackman M, Fallon N, Barilla RM, Henning JR, Jamal M, Rao R, Greco S, Deutsch M, Medina-Zea MV, Bin Saeed U, Ego-Osuala MO, Hajdu C, Miller G. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J Clin Invest 2012;122:4118–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Zambirinis CP, Levie E, Nguy S, Avanzi A, Barilla R, Xu Y, Seifert L, Daley D, Greco SH, Deutsch M, Jonnadula S, Torres-Hernandez A, Tippens D, Pushalkar S, Eisenthal A, Saxena D, Ahn J, Hajdu C, Engle DD, Tuveson D, Miller G. TLR9 ligation in pancreatic stellate cells promotes tumorigenesis. J Exp Med 2015;212:2077–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Notta F, Chan-Seng-Yue M, Lemire M, Li Y, Wilson GW, Connor AA, Denroche RE, Liang SB, Brown AM, Kim JC, Wang T, Simpson JT, Beck T, Borgida A, Buchner N, Chadwick D, Hafezi-Bakhtiari S, Dick JE, Heisler L, Hollingsworth MA, Ibrahimov E, Jang GH, Johns J, Jorgensen LG, Law C, Ludkovski O, Lungu I, Ng K, Pasternack D, Petersen GM, Shlush LI, Timms L, Tsao MS, Wilson JM, Yung CK, Zogopoulos G, Bartlett JM, Alexandrov LB, Real FX, Cleary SP, Roehrl MH, McPherson JD, Stein LD, Hudson TJ, Campbell PJ, Gallinger S. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 2016;538:378–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.de Oliveira Mann CC, Kranzusch PJ. cGAS Conducts Micronuclei DNA Surveillance. Trends Cell Biol 2017;27:697–698. [DOI] [PubMed] [Google Scholar]
  • 102.Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Canellas A, Hernando-Momblona X, Byrom D, Matarin JA, Calon A, Rivas EI, Nebreda AR, Riera A, Attolini CS, Batlle E. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018;554:538–543. [DOI] [PubMed] [Google Scholar]
  • 103.Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, Barcellos-Hoff MH, Demaria S. TGFbeta Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res 2015;75:2232–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Young KH, Newell P, Cottam B, Friedman D, Savage T, Baird JR, Akporiaye E, Gough MJ, Crittenden M. TGFbeta inhibition prior to hypofractionated radiation enhances efficacy in preclinical models. Cancer Immunol Res 2014;2:1011–22. [DOI] [PubMed] [Google Scholar]
  • 105.Yasmin-Karim S, Bruck PT, Moreau M, Kunjachan S, Chen GZ, Kumar R, Grabow S, Dougan SK, Ngwa W. Radiation and Local Anti-CD40 Generate an Effective in situ Vaccine in Preclinical Models of Pancreatic Cancer. Front Immunol 2018;9:2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kalbasi A, Komar C, Tooker GM, Liu M, Lee JW, Gladney WL, Ben-Josef E, Beatty GL. Tumor-Derived CCL2 Mediates Resistance to Radiotherapy in Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2017;23:137–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Rech AJ, Dada H, Kotzin JJ, Henao-Mejia J, Minn AJ, Twyman-Saint Victor C, Vonderheide RH. Radiotherapy and CD40 Activation Separately Augment Immunity to Checkpoint Blockade in Cancer. Cancer Res 2018;78:4282–4291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, Herati RS, Mansfield KD, Patsch D, Amaravadi RK, Schuchter LM, Ishwaran H, Mick R, Pryma DA, Xu X, Feldman MD, Gangadhar TC, Hahn SM, Wherry EJ, Vonderheide RH, Minn AJ. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015;520:373–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Rudqvist NP, Pilones KA, Lhuillier C, Wennerberg E, Sidhom JW, Emerson RO, Robins HS, Schneck J, Formenti SC, Demaria S. Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells. Cancer Immunol Res 2018;6:139–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dougan SK, Dougan M. Regulation of innate and adaptive antitumor immunity by IAP antagonists. Immunotherapy 2018;10:787–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Clancy-Thompson E, Ali L, Bruck PT, Exley MA, Blumberg RS, Dranoff G, Dougan M, Dougan SK. IAP Antagonists Enhance Cytokine Production from Mouse and Human iNKT Cells. Cancer Immunol Res 2018;6:25–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dougan M, Dougan S, Slisz J, Firestone B, Vanneman M, Draganov D, Goyal G, Li W, Neuberg D, Blumberg R, Hacohen N, Porter D, Zawel L, Dranoff G. IAP inhibitors enhance co-stimulation to promote tumor immunity. J Exp Med 2010;207:2195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Beatty GL, Li Y, Long KB. Cancer immunotherapy: activating innate and adaptive immunity through CD40 agonists. Expert Rev Anticancer Ther 2017;17:175–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL, Torigian DA, O’Dwyer PJ, Vonderheide RH. 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]
  • 115.Beatty GL, Torigian DA, Chiorean EG, Saboury B, Brothers A, Alavi A, Troxel AB, Sun W, Teitelbaum UR, Vonderheide RH, O’Dwyer P. 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]
  • 116.Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL. IFNgamma and CCL2 Cooperate to Redirect Tumor-Infiltrating Monocytes to Degrade Fibrosis and Enhance Chemotherapy Efficacy in Pancreatic Carcinoma. Cancer Discov 2016;6:400–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.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]
  • 118.Beatty GL, O’Hara M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: Defining the challenges and next steps. Pharmacol Ther 2016;166:30–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chmielewski M, Hahn O, Rappl G, Nowak M, Schmidt-Wolf IH, Hombach AA, Abken H. T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. Gastroenterology 2012;143:1095–107 e2. [DOI] [PubMed] [Google Scholar]
  • 120.Maliar A, Servais C, Waks T, Chmielewski M, Lavy R, Altevogt P, Abken H, Eshhar Z. Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice. Gastroenterology 2012;143:1375–1384 e5. [DOI] [PubMed] [Google Scholar]
  • 121.Watanabe K, Luo Y, Da T, Guedan S, Ruella M, Scholler J, Keith B, Young RM, Engels B, Sorsa S, Siurala M, Havunen R, Tahtinen S, Hemminki A, June CH. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight 2018;3:e99573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Heckler M, Dougan SK. Unmasking Pancreatic Cancer: Epitope Spreading After Single Antigen Chimeric Antigen Receptor T-Cell Therapy in a Human Phase I Trial. Gastroenterology 2018;155:11–14. [DOI] [PubMed] [Google Scholar]
  • 123.Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, Pamer EG, Li MO. The cellular and molecular origin of tumor-associated macrophages. Science 2014;344:921–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, Toriola AT, Nieman RK, Worley LA, Yano M, Fowler KJ, Lockhart AC, Suresh R, Tan BR, Lim KH, Fields RC, Strasberg SM, Hawkins WG, DeNardo DG, Goedegebuure SP, Linehan DC. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol 2016;17:651–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Steele CW, Karim SA, Leach JD, Bailey P, Upstill-Goddard R, Rishi L, Foth M, Bryson S, McDaid K, Wilson Z, Eberlein C, Candido JB, Clarke M, Nixon C, Connelly J, Jamieson N, Carter CR, Balkwill F, Chang DK, Evans TR, Strathdee D, Biankin AV, Nibbs RJ, Barry ST, Sansom OJ, Morton JP. CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma. Cancer Cell 2016;29:832–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Dranoff G Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 2004;4:11–22. [DOI] [PubMed] [Google Scholar]
  • 127.Wang P, Li X, Wang J, Gao D, Li Y, Li H, Chu Y, Zhang Z, Liu H, Jiang G, Cheng Z, Wang S, Dong J, Feng B, Chard LS, Lemoine NR, Wang Y. Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat Commun 2017;8:1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, Chio II, Hwang CI, Tiriac H, Baker LA, Engle DD, Feig C, Kultti A, Egeblad M, Fearon DT, Crawford JM, Clevers H, Park Y, Tuveson DA. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med 2017;214:579–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Long KB, Tooker G, Tooker E, Luque SL, Lee JW, Pan X, Beatty GL. IL-6 receptor blockade enhances chemotherapy efficacy in pancreatic ductal adenocarcinoma. Mol Cancer Ther 2017;16:1898–1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mace TA, Shakya R, Pitarresi JR, Swanson B, McQuinn CW, Loftus S, Nordquist E, Cruz-Monserrate Z, Yu L, Young G, Zhong X, Zimmers TA, Ostrowski MC, Ludwig T, Bloomston M, Bekaii-Saab T, Lesinski GB. IL-6 and PD-L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer. Gut 2016;67:320–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jiang H, Hegde S, DeNardo DG. Tumor-associated fibrosis as a regulator of tumor immunity and response to immunotherapy. Cancer Immunol Immunother 2017;66:1037–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, Lolkema MP, Jiang P, Kultti A, Thompson CB, Maneval DC, Jodrell DI, Frost GI, Shepard HM, Skepper JN, Tuveson DA. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013;62:112–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017;168:707–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 2015;160:48–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Brooks J, Fleischmann-Mundt B, Woller N, Niemann J, Ribback S, Peters K, Demir IE, Armbrecht N, Ceyhan GO, Manns MP, Wirth TC, Kubicka S, Bernhardt G, Smyth MJ, Calvisi DF, Gurlevik E, Kuhnel F. Perioperative, Spatiotemporally Coordinated Activation of T and NK Cells Prevents Recurrence of Pancreatic Cancer. Cancer Res 2018;78:475–488. [DOI] [PubMed] [Google Scholar]
  • 136.Gurlevik E, Fleischmann-Mundt B, Brooks J, Demir IE, Steiger K, Ribback S, Yevsa T, Woller N, Kloos A, Ostroumov D, Armbrecht N, Manns MP, Dombrowski F, Saborowski M, Kleine M, Wirth TC, Oettle H, Ceyhan GO, Esposito I, Calvisi DF, Kubicka S, Kuhnel F. Administration of Gemcitabine After Pancreatic Tumor Resection in Mice Induces an Antitumor Immune Response Mediated by Natural Killer Cells. Gastroenterology 2016;151:338–350 e7. [DOI] [PubMed] [Google Scholar]
  • 137.Howitt MR, Lavoie S, Michaud M, Blum AM, Tran SV, Weinstock JV, Gallini CA, Redding K, Margolskee RF, Osborne LC, Artis D, Garrett WS. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 2016;351:1329–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Westphalen CB, Takemoto Y, Tanaka T, Macchini M, Jiang Z, Renz BW, Chen X, Ormanns S, Nagar K, Tailor Y, May R, Cho Y, Asfaha S, Worthley DL, Hayakawa Y, Urbanska AM, Quante M, Reichert M, Broyde J, Subramaniam PS, Remotti H, Su GH, Rustgi AK, Friedman RA, Honig B, Califano A, Houchen CW, Olive KP, Wang TC. Dclk1 Defines Quiescent Pancreatic Progenitors that Promote Injury-Induced Regeneration and Tumorigenesis. Cell Stem Cell 2016;18:441–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Bailey JM, Alsina J, Rasheed ZA, McAllister FM, Fu YY, Plentz R, Zhang H, Pasricha PJ, Bardeesy N, Matsui W, Maitra A, Leach SD. DCLK1 marks a morphologically distinct subpopulation of cells with stem cell properties in preinvasive pancreatic cancer. Gastroenterology 2014;146:245–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhang Y, Zoltan M, Riquelme E, Xu H, Sahin I, Castro-Pando S, Montiel MF, Chang K, Jiang Z, Ling J, Gupta S, Horne W, Pruski M, Wang H, Sun SC, Lozano G, Chiao P, Maitra A, Leach SD, Kolls JK, Vilar E, Wang TC, Bailey JM, McAllister F. Immune Cell Production of Interleukin 17 Induces Stem Cell Features of Pancreatic Intraepithelial Neoplasia Cells. Gastroenterology 2018;155:210–223 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. 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]
  • 142.Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, Sherry RM, Topalian SL, Yang JC, Lowy I, Rosenberg SA. 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]
  • 143.Naing A, Powderly JD, Falchook G, Creelan B, Nemunaitis J, Lutzky J, Diab A, Wang JS, Laing N, Niewood M, Gong X, Zhou G, Patel M. Abstract CT177: Epacadostat plus durvalumab in patients with advanced solid tumors: preliminary results of the ongoing, open-label, phase I/II ECHO-203 study. Cancer Research 2018;78:CT177. [Google Scholar]
  • 144.Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol 2008;8:59–73. [DOI] [PubMed] [Google Scholar]
  • 145.Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F, Yamazaki T, Poirier-Colame V, Newton A, Redouane Y, Lin YJ, Wojtkiewicz G, Iwamoto Y, Mino-Kenudson M, Huynh TG, Hynes RO, Freeman GJ, Kroemer G, Zitvogel L, Weissleder R, Pittet MJ. Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. Immunity 2016;44:343–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Aglietta M, Barone C, Sawyer MB, Moore MJ, Miller WH Jr., Bagala C, Colombi F, Cagnazzo C, Gioeni L, Wang E, Huang B, Fly KD, Leone F. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol 2014;25:1750–5. [DOI] [PubMed] [Google Scholar]
  • 147.Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, Korn RL, Desai N, Trieu V, Iglesias JL, Zhang H, Soon-Shiong P, Shi T, Rajeshkumar NV, Maitra A, Hidalgo M. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 2011;29:4548–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, Bennouna J, Bachet JB, Khemissa-Akouz F, Pere-Verge D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817–25. [DOI] [PubMed] [Google Scholar]
  • 149.Coveler AL, Rossi GR, Vahanian NN, Link C, Chiorean EG. Algenpantucel-L immunotherapy in pancreatic adenocarcinoma. Immunotherapy 2016;8:117–25. [DOI] [PubMed] [Google Scholar]
  • 150.Le DT, Ko AH, Wainberg ZA, Picozzi VJ, Kindler HL, Wang-Gillam A, Oberstein PE, Morse M, Zeh H, Weekes CD, Reid TR, Murphy A, McDougall K, Whiting CC, Nair N, Enstrom A, Ferber S, Dubensky TW, Brockstedt DG, Jaffee EM. Results from a phase 2b, randomized, multicenter study of GVAX pancreas and CRS-207 compared to chemotherapy in adults with previously-treated metastatic pancreatic adenocarcinoma (ECLIPSE Study). J Clin Oncol 2017;35: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, Propper D, Coxon F, Ross P, Madhusudan S, Roques T, Cunningham D, Falk S, Wadd N, Harrison M, Corrie P, Iveson T, Robinson A, McAdam K, Eatock M, Evans J, Archer C, Hickish T, Garcia-Alonso A, Nicolson M, Steward W, Anthoney A, Greenhalf W, Shaw V, Costello E, Naisbitt D, Rawcliffe C, Nanson G, Neoptolemos J. Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncol 2014;15:829–40. [DOI] [PubMed] [Google Scholar]
  • 152.Ercolini AM, Ladle BH, Manning EA, Pfannenstiel LW, Armstrong TD, Machiels JP, Bieler JG, Emens LA, Reilly RT, Jaffee EM. Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response. J Exp Med 2005;201:1591–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E, Hege K, Jaffee E. Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res 2008;14:1455–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39:1–10. [DOI] [PubMed] [Google Scholar]
  • 155.Beatty GL, O’Hara MH, Lacey SF, Torigian DA, Nazimuddin F, Chen F, Kulikovskaya IM, Soulen MC, McGarvey M, Nelson AM, Gladney WL, Levine BL, Melenhorst JJ, Plesa G, June CH. Activity of Mesothelin-specific Chimeric Antigen Receptor T Cells Against Pancreatic Carcinoma Metastases in a Phase 1 Trial. Gastroenterology 2018;155:29–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL, Albelda SM, Kalos M, June CH. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce antitumor activity in solid malignancies. Cancer Immunol Res 2014;2:112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Beatty GL, Moon EK. Chimeric antigen receptor T cells are vulnerable to immunosuppressive mechanisms present within the tumor microenvironment. Oncoimmunology 2014;3:e970027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Carleton M, Powers J, Beg MS, Lee JJ, Rahma OE, Piha-Paul SA, Lee E, Hestir K, Xu K, Pandya D, Inamdar SP, Perna SK, Huang SA, Wainberg ZA. Pharmacodynamics (PD) and genomic profiling of pts treated with cabiralizumab (cabira) + nivolumab (NIVO) provide evidence of on-target tumor immune modulations and support future clinical applications. J Clin Oncol 2018;36: [Google Scholar]
  • 160.Wainberg Z, Piha-Paul S, Luke J, Kim E, Thompson J, Pfanzelter N, Gordon M, Rasco D, Weise A, Hodi FS, Inamdar S, Perna S, Ma C, Powers J, Carleton M, Xiang H, Zhou L, Colling H, Lee Y, Lee J, Johnson J, Britten C, Ghoddusi M. First-in-human phase 1 dose escalation and expansion of a novel combination, anti–CSF-1 receptor (cabiralizumab) plus anti–PD-1 (nivolumab), in patients with advanced solid tumors. J Immunother Cancer 2017;5: [Google Scholar]
  • 161.Lupinacci RM, Goloudina A, Buhard O, Bachet JB, Marechal R, Demetter P, Cros J, Bardier-Dupas A, Collura A, Cervera P, Scriva A, Dumont S, Hammel P, Sauvanet A, Louvet C, Delpero JR, Paye F, Vaillant JC, Andre T, Closset J, Emile JF, Van Laethem JL, Jonchere V, Abd Alsamad I, Antoine M, Rodenas A, Flejou JF, Dusetti N, Iovanna J, Duval A, Svrcek M. Prevalence of Microsatellite Instability in Intraductal Papillary Mucinous Neoplasms of the Pancreas. Gastroenterology 2017;154:1061–1065. [DOI] [PubMed] [Google Scholar]
  • 162.Bahary N, Wang-Gillam A, Haraldsdottir S, Somer BG, Lee JS, O’Rourke MA, Nayak-Kapoor A, Beatty GL, Liu M, Delman D, Rossi GR, Kennedy EP, Vahanian NN, Link CJ, Garrido-Laguna I. Phase 2 trial of the IDO pathway inhibitor indoximod plus gemcitabine / nab-paclitaxel for the treatment of patients with metastatic pancreas cancer. J Clin Oncol 2018;36:suppl; [Google Scholar]
  • 163.Nowak AK, Robinson BW, Lake RA. Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res 2003;63:4490–6. [PubMed] [Google Scholar]
  • 164.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, Metivier D, Larochette N, van Endert P, Ciccosanti F, Piacentini M, Zitvogel L, Kroemer G. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007;13:54–61. [DOI] [PubMed] [Google Scholar]
  • 165.Brenchley JM, Karandikar NJ, Betts MR, Ambrozak DR, Hill BJ, Crotty LE, Casazza JP, Kuruppu J, Migueles SA, Connors M, Roederer M, Douek DC, Koup RA. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood 2003;101:2711–20. [DOI] [PubMed] [Google Scholar]
  • 166.Cioffi M, Trabulo S, Hidalgo M, Costello E, Greenhalf W, Erkan M, Kleeff J, Sainz B Jr., Heeschen C. Inhibition of CD47 Effectively Targets Pancreatic Cancer Stem Cells via Dual Mechanisms. Clin Cancer Res 2015;21:2325–37. [DOI] [PubMed] [Google Scholar]
  • 167.Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, Lovelace P, Scheeren FA, Chao MP, Weiskopf K, Tang C, Volkmer AK, Naik TJ, Storm TA, Mosley AR, Edris B, Schmid SM, Sun CK, Chua MS, Murillo O, Rajendran P, Cha AC, Chin RK, Kim D, Adorno M, Raveh T, Tseng D, Jaiswal S, Enger PO, Steinberg GK, Li G, So SK, Majeti R, Harsh GR, van de Rijn M, Teng NN, Sunwoo JB, Alizadeh AA, Clarke MF, Weissman IL. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A 2012;109:6662–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, Cho BC, Bourhaba M, Quantin X, Tokito T, Mekhail T, Planchard D, Kim YC, Karapetis CS, Hiret S, Ostoros G, Kubota K, Gray JE, Paz-Ares L, de Castro Carpeno J, Wadsworth C, Melillo G, Jiang H, Huang Y, Dennis PA, Ozguroglu M. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:1919–1929. [DOI] [PubMed] [Google Scholar]
  • 169.Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, Chau I, Ernstoff MS, Gardner JM, Ginex P, Hallmeyer S, Holter Chakrabarty J, Leighl NB, Mammen JS, McDermott DF, Naing A, Nastoupil LJ, Phillips T, Porter LD, Puzanov I, Reichner CA, Santomasso BD, Seigel C, Spira A, Suarez-Almazor ME, Wang Y, Weber JS, Wolchok JD, Thompson JA. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018;36:1714–1768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Reynolds K, Thomas M, Dougan M. Diagnosis and Management of Hepatitis in Patients on Checkpoint Blockade. Oncologist 2018;23:991–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Dougan M Checkpoint Blockade Toxicity and Immune Homeostasis in the Gastrointestinal Tract. Front Immunol 2017;8:1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ferrara R, Mezquita L, Texier M, Lahmar J, Audigier-Valette C, Tessonnier L, Mazieres J, Zalcman G, Brosseau S, Le Moulec S, Leroy L, Duchemann B, Lefebvre C, Veillon R, Westeel V, Koscielny S, Champiat S, Ferte C, Planchard D, Remon J, Boucher ME, Gazzah A, Adam J, Bria E, Tortora G, Soria JC, Besse B, Caramella C. Hyperprogressive Disease in Patients With Advanced Non-Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol 2018;4:1543–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Rahib L, Fleshman JM, Matrisian LM, Berlin JD. Evaluation of Pancreatic Cancer Clinical Trials and Benchmarks for Clinically Meaningful Future Trials: A Systematic Review. JAMA Oncol 2016;2:1209–16. [DOI] [PubMed] [Google Scholar]

RESOURCES