Immunotherapy has dramatically changed the cancer treatment landscape during the past decade, but very limited efficacy has been reported against pancreatic cancer. Several factors unique to pancreatic cancer may explain the resistance: the well-recognized suppressive elements in the tumor microenvironment, the functional and structural barrier imposed by the stroma components, T-cell exhaustion, the choice of perhaps the wrong immune targets, and microbial factors including gut dysbiosis and the unexpected presence of tumor microbes. Furthermore, we discuss various strategies to overcome these barriers.

Immunotherapy has revolutionized cancer medicine, providing long-lasting disease control in malignancies that were previously uniformly fatal (1). One illustrative example is seen in metastatic melanoma, in which the median survival has doubled from 8–12 months to greater than 24 months with the advent of immunotherapy (2). In some cases—former President Jimmy Carter being one well-known example—immunotherapy can result in metastatic cancer cures. While the promise of immunotherapy has profoundly altered the management of many cancer types, this paradigm shift has not extended to pancreatic ductal adenocarcinoma (PDAC). Indeed, PDAC, with an incidence of 12.9 per 100,000 is much less common than cancers such as breast, lung, and prostate, and yet in 2019 was the third leading cause of cancer-related death (3), and it is projected to become the second leading cause of cancer-related death by 2030 (4).

Treatment options for PDAC remain quite limited. Surgical resection is the only curative option but possible in only 15% to 20% of cases due to diagnosis at advanced stages in most cases. Even following major surgery and adjuvant chemotherapy with 5-fluorouracil (5-FU), irinotecan, and oxaliplatin (FOLFIRINOX), disease recurrence is common; a recent study showed distant and/or locoregional recurrence over 50% at a median follow-up of 33.6 months (5). For those patients who are more commonly diagnosed with locally advanced or metastatic disease, medical therapies are limited to combination chemotherapy. For the most medically fit patients with good performance status, combination therapy with FOLFIRINOX has shown to improve progression-free survival (PFS) from 3.3 months with single-agent gemcitabine to 6.8 months with the combination. Still, median overall survival (OS) with this regimen remains poor at 11.1 months (6). For those patients who do respond to treatment, toxicities including myelosuppresion, peripheral neuropathy, diarrhea, and profound fatigue can frequently decrease the quality of life. The second regimen used is combination of gemcitabine and nab-paclitaxel which prolongs survival to 8.5 months compared with single-agent gemcitabine (7). While generally better tolerated, this regimen nonetheless comes with its own unique toxicities which include neuropathy and myelosuppression.

It is clear that more effective therapies are desperately needed for PDAC. Despite herculean research efforts from the scientific community and pharmaceutical industry to combat this disease, even incremental improvements in terms of toxicity profile and OS remain elusive. However, there are several promising avenues of research which have shown hope in making inroads in the fight against PDAC, including the use of immunotherapy. As Robert Vonderheide, professor and immunologist expert in pancreatic cancer at University of Pennsylvania (Philadelphia, PA), once stated at a national meeting, “copy–paste of immunotherapy from other cancers to pancreatic cancer will not work”. To date, clinical trials in the realm of immunotherapy in PDAC have been disappointing. In a review article, Henriksen and colleagues pointed to over 20 published trials using checkpoint inhibitors in PDAC (8). A trial of CTLA4 monotherapy showed no responders. Two trials of PD-1 or PD-L1 inhibitor monotherapy showed no objective response for anti-PD-L1 monotherapy, and combined PD-L1 inhibition with CTLA4 inhibition showed a low disease control rate of only 9%. Trials of combination chemotherapy and immunotherapy have had equally lackluster results. For example, two clinical trials which evaluated combined CTLA4 inhibition and gemcitabine therapy showed similar efficacy rates to gemcitabine monotherapy. Another trial evaluated nivolumab with combination gemcitabine/nab-paclitaxel for patients with advanced PDAC, and showed a PFS of 5.5 months, identical to PFS seen in the original study from Von Hoff that led to approval of combination gemcitabine/nab-paclitaxel in 2013. The lack of ongoing trials of checkpoint inhibitor monotherapy point to absence of clinical response or OS demonstrated to date with use of these agents in PDAC.

Here we explore the potential explanations for PDAC tumor resistance mechanisms to immunotherapy and compile strategies to overcome resistance to immunotherapy in PDAC, an approach that still holds promise for the control of this disease.

The PDAC tumor microenvironment (TME) has come into focus as a crucial adaptive mechanism by which cancer cells evade immune surveillance, enabling progressive invasion and distant metastatic spread (9). The PDAC TME is mostly comprised of pancreatic stellate cells (PSC), extracellular matrix (ECM), and immunosuppressive cellular populations (10). The TME can be heavily influenced by host factors including obesity, a known risk factor for development of PDAC (11). Gomez-Chou and colleagues have recently demonstrated that higher levels of lipocalin-2 (LCN2), an adipokine elevated in the serum and in adipose tissue of obese individuals, can adversely modulate the TME through a variety of mechanisms including activation of PSCs with subsequent stromal remodeling, and upregulation of tumor-associated macrophages (TAM), which are implicated in ductal metaplasia leading to PDAC development (12).

The activities of TAMs include secretion of immunosuppressive cytokines and enzymes such as TGFβ and IL10, interference with the metabolism of effector T cells, recruitment of regulatory T cells (Treg), and suppression of cytotoxic CD8+ T-cell activation (13). TAMs, in turn, upregulate PSCs, further contributing to the immunosuppressive microenvironment of PDAC (14).

Myeloid-derived suppressor cells (MDSC) are immune cells that play a critical role in pancreatic cancer by suppressing the function of effector T cells through numerous pathways, including through the use of reactive oxygen species (ROS), production of adenosine, and by inducing de novo Treg development through secretion of IFNγ and IL10 (15). Zhang and colleagues have shown that in vivo genetic depletion of myeloid cells in murine PDAC transgenic models resulted in tumor arrest and CD8+ T-cell–dependent tumor regression (16).

Treg CD4+ lymphocyte–infiltrating tumors are linked to worse outcomes in patients with PDAC (17). However, recently Zhang and colleagues have depleted Tregs in transgenic murine models of PDAC and unexpectedly, tumor progression was accelerated by reprogramming of the fibroblast population, highlighting the complex relationships between Tregs and fibroblasts in PDAC (18).

Another protumorigenic cell type that infiltrates the pancreas during tumorigenesis, particularly in the context of chronic pancreatitis, is Th17. These cells are the main source of IL17, a cytokine that binds IL17RA, which is overexpressed in pancreatic epithelium upon Kras oncogenic activation, resulting in increased immunosuppression through various mechanisms including induction of genes related to embryonic stemness and secretion of chemokines that attract MDSCs and neutrophils to the TME (19). GM-CSF is a cytokine that is also secreted in the context of oncogenic Kras-dependent pancreatic tumorigenesis, and also has the capacity of attracting suppressive myeloid cells that interfere with CD8+ T-cell infiltration and tumor cell killing (20).

Neutrophils account for another immune-suppressive population present in the PDAC TME and their depletion in pancreatic cancer murine models have resulted in antitumorigenic effects (21). Mechanisms of immune suppression via neutrophils include the secretion of cytokines and chemokines, and suppression of cytotoxic CD8+ T lymphocytes (22, 23). Production of neutrophil extracellular traps (NET) has been proposed as another mechanism of immunotherapy resistance induced by neutrophils. There are several known inducers of NETs, and recently IL17 has been described as an indirect NET inducer, as it needs to first interact with pancreatic tumor cells (24).

An additional crucial resistance mechanism of the PDAC TME is angiogenesis by way of VEGF, which leads to disorganized tumor vasculature, hypoxia, high interstitial pressures, and low pH in the TME, which serves to hinder the trafficking and effector function of immune cells. VEGF facilitates an immunosuppressive environment through multiple pathways in addition to angiogenesis, including the direct inhibition of cytotoxic T-cell trafficking and function, inhibition of antigen presentation, and recruitment of the abovementioned immunosuppressive cells including Tregs, MDSCs, and M2-like TAMs to the TME (25). The use of VEGFR inhibitors in combination with immunotherapy has been successful in several tumor types including hepatocellular carcinoma (HCC) and renal cell carcinoma (RCC), through normalization of the tumor vasculature, and is currently being evaluated in pancreatic cancer in combination with immunotherapy (26).

Despite the complex interactions of these various cell signaling pathways in the PDAC TME, several trials are currently underway employing combination strategies to overcome the PDAC immunosuppressive TME. These completed and ongoing trials combine traditional chemotherapy or immunotherapy with drugs or antibodies that target the suppressive TME, rather than the cancer cells themselves (Table 1).

Table 1.

Summary of ongoing combinatorial clinical trials in PDAC.

Trial NameIdentifierSponsorTreatment strategyResistance mechanism
Pilot study with cyclophosphamide, pembrolizumab, GVAX, and IMC-CS4 (LY3022855) in patients with borderline resectable adenocarcinoma of the pancreas NCT03153410 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of vaccine, chemotherapy, checkpoint blockade, and CSF1-R inhibition Suppressive TME/T-cell exclusion/exhaustion 
A study to evaluate the safety and tolerability of SX-682 in combination with nivolumab as maintenance therapy in patients with metastatic panceatic ductal adenocarcinoma NCT04477343 University of Rochester Combination of checkpoint blockade and CXCR1/2 inhibition Suppressive TME 
Trial of neoadjuvant and adjuvant nivolumab and BMS-813160 with or without GVAX for locally advanced pancreatic ductal adenocarcinomas NCT03767582 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of vaccine, checkpoint blockade, and CCR2/5 inhibition Suppressive TME 
Plerixafor and cemiplimab in metastatic pancreatic cancer NCT04177810 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of bone marrow stimulant and checkpoint blockade T-cell exclusion/exhaustion 
Chemo4METPANC combination chemokine inhibitor, immunotherapy, and chemotherapy in patients with pancreatic adenocarcinoma NCT04543071 Columbia University Combination of chemotherapy, checkpoint blockade, and CXCR4 inhibitor Suppressive TME/T-cell exclusion/exhaustion 
LOAd703 oncolytic virus therapy for pancreatic cancer NCT02705196 Lokon Pharmaceuticals Combination of immunostimulatory oncolytic adenovirus, chemotherapy, and checkpoint blockade T-cell exclusion/exhaustion 
Study of CRS-207, nivolumab, and ipilimumab with or without GVAX pancreas vaccine (with cyclophsphamide; in patients with pancreatic cancer NCT03190265 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination vaccine and checkpoint blockade T-cell exclusion/exhaustion 
Paricalcitol and hydroxychloroquine in combination with gemcitabine and NabP for the treatment of advanced or metastatic pancreatic cancer NCT04524702 Emory University Combination of Vit D, chemotherapy, and an autophagy inhibitor Fibrotic stroma 
Study of pembrolizumab with or without defactinib following chemotherapy as a neoadjuvant and adjuvant treatment for resectable pancreatic ductal adenocarcinoma NCT03727880 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of checkpoint blockade and FAK inhibition Fibrotic stroma 
Trial NameIdentifierSponsorTreatment strategyResistance mechanism
Pilot study with cyclophosphamide, pembrolizumab, GVAX, and IMC-CS4 (LY3022855) in patients with borderline resectable adenocarcinoma of the pancreas NCT03153410 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of vaccine, chemotherapy, checkpoint blockade, and CSF1-R inhibition Suppressive TME/T-cell exclusion/exhaustion 
A study to evaluate the safety and tolerability of SX-682 in combination with nivolumab as maintenance therapy in patients with metastatic panceatic ductal adenocarcinoma NCT04477343 University of Rochester Combination of checkpoint blockade and CXCR1/2 inhibition Suppressive TME 
Trial of neoadjuvant and adjuvant nivolumab and BMS-813160 with or without GVAX for locally advanced pancreatic ductal adenocarcinomas NCT03767582 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of vaccine, checkpoint blockade, and CCR2/5 inhibition Suppressive TME 
Plerixafor and cemiplimab in metastatic pancreatic cancer NCT04177810 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of bone marrow stimulant and checkpoint blockade T-cell exclusion/exhaustion 
Chemo4METPANC combination chemokine inhibitor, immunotherapy, and chemotherapy in patients with pancreatic adenocarcinoma NCT04543071 Columbia University Combination of chemotherapy, checkpoint blockade, and CXCR4 inhibitor Suppressive TME/T-cell exclusion/exhaustion 
LOAd703 oncolytic virus therapy for pancreatic cancer NCT02705196 Lokon Pharmaceuticals Combination of immunostimulatory oncolytic adenovirus, chemotherapy, and checkpoint blockade T-cell exclusion/exhaustion 
Study of CRS-207, nivolumab, and ipilimumab with or without GVAX pancreas vaccine (with cyclophsphamide; in patients with pancreatic cancer NCT03190265 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination vaccine and checkpoint blockade T-cell exclusion/exhaustion 
Paricalcitol and hydroxychloroquine in combination with gemcitabine and NabP for the treatment of advanced or metastatic pancreatic cancer NCT04524702 Emory University Combination of Vit D, chemotherapy, and an autophagy inhibitor Fibrotic stroma 
Study of pembrolizumab with or without defactinib following chemotherapy as a neoadjuvant and adjuvant treatment for resectable pancreatic ductal adenocarcinoma NCT03727880 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Combination of checkpoint blockade and FAK inhibition Fibrotic stroma 

Abbreviation: Vit D, Vitamin D.

One such combinatorial strategy involves inhibition of colony-stimulating factor-1 receptor (CSF1R; which recruits TAMs to the TME) combined with immunotherapy. CSF1R inhibition has been shown to upregulate T-cell checkpoint molecules including PD-L1 and CTLA4 (27). Nywening and colleagues have shown that the use of small-molecule inhibitors against chemokine receptor type 2 (CXCR2) receptor expressed in the membrane of tumor-associated neutrophils and MDSCs can induce antitumor immunity against PDAC when combined with chemotherapy (28). CXCR2 and its ligands are responsible for recruiting neutrophils to the TME, which contribute to tumor immune evasion. Combined CCR2 and CXCR2 blockade, along with FOLFIRINOX chemotherapy, increase OS in a KPC mouse model. Loss of p53 can induce overexpression of CCL2, which further recruits suppressive myeloid cells into the TME, and promotes epithelial-to-mesenchymal transition (EMT), which has been associated with resistance to immunotherapy (29). The CCL2/CCR2 and chemokine receptor type 4 (CXCR4) receptor pathways that are involved in the recruitment of immunosuppressive monocytes to the TME and are currently under evaluation as drug targets in conjunction with chemotherapy and immunotherapy (10). Recent preclinical work has also demonstrated a decrease in total immune infiltration of suppressive myeloid cells through the inhibition of CCR1 (18). Genetic or pharmacologic deletion of neutrophils or NETs can reverse PDAC resistance to checkpoint blockade (24).

Other current avenues for research include the use of radiotherapy and/or radiofrequency ablation in conjunction with immunotherapy to circumvent the naturally suppressive TME of PDAC (27). An “abscopal effect” has been posited as justification for use of radiotherapy in conjunction with a systemic immune-stimulatory agent in metastatic solid tumors. However, recent data indicate that in tumors with low immunogenicity such as PDAC, multiple levels of immune activation may be required to see a desired effect (30). One novel ablative strategy involves irreversible electroporation (IRE), a nonthermal ablative technique in which high-voltage electrical pulses are applied directly to the tumor. The phase II PANFIRE trial explored the use of IRE in patients with locally advanced or recurrent PDAC, in conjunction with cytotoxic chemotherapy. The target median OS exceeded, and a phase III trial in conjunction with immunotherapy is currently under way (31). Another combinatorial approach is the use of “super” IL15 in conjunction with immunotherapy, which acts as a long-acting cytokine to increase circulating natural killer (NK) cells, as well as activate CD8+ T cells in the TME. The use of “super” IL15 has been tested in patients with advanced solid tumor malignancies and found to be safe while inducing a strong immune response. Its efficacy in combination with checkpoint blockade is currently under investigation (32). Galunisertinib, a TGFβ receptor inhibitor that blocks this strongly immunosuppressive cytokine, is also currently under investigation in combination with both cytotoxic chemotherapy and immunotherapy (3). It should be noted that toxicity may be a limiting factor as researchers continue to explore combinations of chemotherapy and immunotherapy regimens in patients with PDAC (33, 34).

On the basis of the dense desmoplastic stroma and tumor infiltration with the immune-suppressive cells described above, recent models of the human PDAC tumor microenvironment point to immune exclusion as a key factor for resistance to therapies (35). Novel imaging techniques that allow for spatial analysis of multiple cellular types within PDAC tissue have found that high levels of total T-cell infiltration are associated with prolonged survival, desmoplastic elements do not impair cytotoxic T-cell infiltration, and close proximity of cytotoxic T cells to pancreatic cancer cells significantly correlates with prolonged patient survival (36). A second study on PDAC immune infiltrates spatial analysis also confirmed a significant association between high CD8+ cell infiltration in the tumor as a whole and improved patient survival. Interestingly, the authors describe a favorable prognostic association with higher CD8+ cell density in the tumor center but not in the tumor margin (37). These findings suggest that the combination of T-cell density data at the tumor center and the tumor margin could be applied as a prognostic tool in PDAC (37). Another recent study proposed that not only the degree of intratumoral T-cell infiltration, but the degree of “attack” as measured by the presence of intraepithelial lymphocytes indicate a more favorable prognosis. More specifically, CD8+ intraepithelial attack was an independent favorable prognostic indicator for OS, while CD8+-high intratumoral infiltration without CD8+ T-cell intraepithelial attack was a poor prognostic factor (38).

Given these findings of a microenvironment characterized by effector T-cell exclusion, current research has focused on enhancing both the quantity and the quality of the immune infiltrate in the pancreatic tumor milieu. One promising avenue of research has explored the inhibition of CXCR4, an alpha chemokine receptor that inhibits T-cell chemotaxis. Using time-lapse confocal microscopy on fresh PDAC tumors treated in organotypic slice culture, researchers demonstrated enhanced tumor-cell apoptosis through the combined use of PD-1 and CXCR4 blockade, as compared with PD-1 monotherapy or control (35). These results showed a striking shift in the distribution of CD8+ T cells from the fibroblastic stroma into the immediate juxtatumoral stroma using combined PD-1 and CXCR4 blockade, indicating that the sequestration of already clonally expanded tumor-reactive T cells, rather than lack of immunogenicity, is responsible for the lack of efficacy of immunotherapy in PDAC, and that this sequestration can be reversed through targeted therapy (35). However, the transient nature of these T-cell responses may be a limiting factor, and likely more robust T-cell priming is vital to not only expanding preexisting antitumor T-cell clones, but recruiting new T-cell clones to the TME (35). One proposed strategy involves activation of CD40 found on both dendritic cells and B cells, which has been shown in mouse models to reverse T-cell exhaustion through the upregulation of cytokines, antigen-presenting molecules, costimulatory molecules, and adhesion molecules (39). Chemotherapy with gemcitabine and nab-paclitaxel, followed by CD40 activation led to the establishment of effective, T-cell–dependent immunity and memory in mouse models compared with CD40 alone, presumably due to the “spill” of antigens induced by chemotherapy (40). The addition of PD-1 or CTLA4 inhibition has been shown to further extend the activity and durability of response to chemotherapy plus CD40 activation (41). Clinical trials with various CD40 antibodies in combination with chemotherapy and/or immunotherapy are now underway across multiple tumor types, including PDAC (Table 1; ref. 39).

Another approach to improve T-cell activation is the use of vaccines. The laboratory of Elizabeth Jaffee at Johns Hopkins University (Baltimore, MD) has pioneered the development of vaccines for pancreatic cancer for more than 20 years (42–44). The first vaccine developed was granulocyte-macrophage colony-stimulating factor gene transfected tumor cells vaccine (GVAX), which is capable of recruiting antigen-presenting cells (APC), mostly dendritic cells (DC), to the vaccination site which results in priming of CD8+ T cells by a mechanism of cross-priming (42, 43). The second vaccine, CRS-207, is made of deactivated Listeria-expressing mesothelin, a tumor-associated antigen (TAA) highly expressed in PDAC, with the goal of also priming T cells. Several murine studies have shown efficacy of combining vaccines with checkpoint inhibitors (45), and previous and current trials are testing combinatorial strategy (46). A trial with GVAX/cyclophosphamide followed by CRS-207 with or without nivolumab was completed, and though it did not meet the primary endpoint of improvement in OS, there were beneficial effects seen within the TME including an increase of CD8+ T cells and decrease in CD68+ myeloid cells (47).

In the future, the development of vaccines for PDAC will likely continue and will likely include combination with chemotherapy and other checkpoint blockade agents or TME-targeting agents.

The fibrotic and desmoplastic stroma in PDAC has been postulated as a structural and functional barrier for therapeutics delivery and efficacy (48). PSCs form the main component of tumor stroma and are inactive in healthy tissue, but secrete extracellular matrix proteins including collagen and fibronectin in the presence of tissue injury or tumorigenesis, decreasing access of immune cells to the tumor (49). In mice coinjected with pancreatic cancer cells plus PSCs, the suppressive immune-cell population of regulatory T cells, M2-type macrophages, and MDSCs was significantly increased, while the immune-cell populations of CD8+ T cells, CD4+ T cells, and M1 macrophages in the tumor tissues were significantly decreased compared with mice solely injected with pancreatic cancer cells (49). The PDAC stromal reaction is mostly driven by the conversion of quiescent to activated PSCs. One strategy to combat the fibrotic PDAC stroma is by targeting of the vitamin D receptor (VDR), which is expressed in the PDAC stroma. Therapy with calcipotriol, which binds VDR, decreases inflammation and fibrosis in a pancreatitis murine model (50). On the basis of this finding, combination of Vitamin D with chemotherapy has been proposed, and clinical trials are underway.

Hedgehog (Hh) signaling, which is absent in the normal adult pancreas, is known to be upregulated in pancreatic cancer stem cells (CSC), facilitating tumorigenesis through paracrine signaling to stromal fibroblasts, which in turn stimulates growth of the fibrotic and immune-suppressive tumor stroma (51). The targeting of Sonic hedgehog (Shh), a soluble ligand overexpressed by PDAC cells, presented a reasonable new avenue of therapeutic research after preclinical studies revealed that Shh inhibition resulted in major improvement in outcomes by facilitating entry of chemotherapy into pancreatic tumors (52). On the basis of these remarkable results, high expectations were built over the clinical trial using standard chemotherapy (gemcitabine/nab-paclitaxel) plus vismodegib (an Hh inhibitor) in patients with metastatic PDAC. Unfortunately, results from a phase II single-arm therapy reported no clinical benefit in this patient population (53, 54).

In light of the clinical findings, a PDAC mouse model was generated with genetic deletion of Shh, which resulted in Shh-deficient tumors with significantly reduced stroma. However, these genetically altered mice lacking Shh expression developed PDAC tumors faster and had a significantly decreased survival compared with control mice, indicating that Shh—and by extension the fibrotic PDAC tumor stroma itself—may in fact restrain tumor progression and aggressiveness (55). Interestingly, these stroma-depleted tumors were found to be more sensitive to VEGFR inhibition, very likely due to increased blood vessel density (55). A second study also used transgenic mouse models to selectively deplete aSMA myofibroblasts in a PDAC mouse model, with a resultant decrease in tumor stroma but an increase in poorly differentiated tumors and significantly diminished survival were found compared with control PDAC mice (56). Myofibroblast-depleted tumors had an increase in the percentage of Tregs, with an overall decrease in the ratio of effector T cells to Tregs. Myofibroblast depletion also resulted in increased CTLA4 expression and anti-CTLA4 antibodies significantly increased mice survival (56). Analysis of PDAC specimens revealed that low aSMA was associated with worse survival (56). Recently, the groups that led these clinical trials reported no differences in number of cancer stem cells or stromal density betwen tumors from visomodegib-treated patients compared with those untreated, questioning the activity of the drug in influencing these components (53, 54).

Focal adhesion kinase (FAK) and specifically FAK1 is a nonreceptor tyrosine kinase which is implicated in activating proinflammatory cytokines and upregulating pathologic fibrosis. A 2016 study published in Nature Medicine showed that FAK1 is upregulated in PDAC, and tumors with high FAK1 expression had higher levels of total stromal collagen and collagen I deposition. The same group demonstrated a synergistic effect in a KPC mouse model between a FAK inhibitor, gemcitabine chemotherapy, and anti-PD-1 immunotherapy as measured by reduced tumor burden and improved OS. Mice treated with this regimen had significantly increased number of CD8+ tumor-infiltrating lymphocytes (TIL) compared with mice treated with gemcitabine and anti-PD-1 immunotherapy alone (57). Several FAK inhibitors are currently under investigation in clinical trials.

Another stromal-related reason for the lack of penetration of drugs is attributed to the extremely high interstitial fluid pressures (IFP) seen in the PDAC microenvironment. A study using KPC mice with autochthonous PDAC tumors demonstrated IFPs in excess of 10 times that of mice with normal pancreata. The elevated IPF in KPC mice was directly associated to hyaluronic acid (HA) deposition in the pancreatic tumor ECM. Administration of pegylated recombinant human hyaluronidase (PEGPH20), an engineered form of the enzyme hyaluronidase with longer half-life, resulted in rapid appearance of patent blood vessels in the TME and normalization of IFP. The combination of PEGPH20 with gemcitabine in spontaneous murine models of PDAC resulted in prolonged OS compared with gemcitabine alone (58). Given this promising preclinical data, a phase Ib/II trial of PEGPH20 in combination with gemcitabine/nab-paclitaxel compared with nab-paclitaxel alone was performed, but it did not show improvements in PFS (5.7 months vs. 5.2 months, P = 0.11; ref. 59). However, a subgroup analysis based on HA content showed a significant improvement in overall response rate (52% vs. 24%, P = 0.038) in patients with tumors of high HA compared with low HA (59). In parallel, a Phase IB/II randomized study of FOLFIRINOX plus human hyaluronidase PEGPH20 compared with FOLFIRINOX alone was performed in patients with metastatic pancreatic cancer with good performance status [Eastern Cooperative Oncology Group (ECOG) 0–1]. Surprisingly, the trial was closed to accrual after interim analysis showed not only increased toxicity in the form of thrombotic events and GI bleeding in the hyaluronidase arm, but a dramatically worse median OS (7.7 months) in the combination group compared with the FOLFIRINOX arm (14.4 months). The authors postulate that the poor outcomes seen in the hyaluronidase arm could be due to dose delays and/or reductions from increased adverse events, drug–drug interactions with components of FOLFIRINOX, or more complex effects on the TME which have yet to be elucidated (60). On the basis of these results, a phase III trial based of combination of PEGPH20 and gemcitabine/nab-paclitaxel compared with nab-paclitaxel alone was planned for patients with high HA expression in metastatic PDAC. The results of this study, recently disclosed, showed that the treatment arm of PEGPH20 in combination with gemcitabine and nab-paclitaxel failed to demonstrate an improvement in median OS compared with gemcitabine and nab-paclitaxel alone (11.2 months compared with 11.5 months, HR = 1.00, P = 0.97; ref. 61). These results ended the development of PEGPH20 for metastatic PDAC.

These studies illustrate the complex nature of the fibrotic TME, and hint that the fibrotic stroma may play a role in suppressing tumorigenesis and disease progression while also dampening the host immune response. The combination of stromal-directed therapy with immunotherapy could represent a new avenue for research in PDAC.

Knowledge of the mechanisms and receptors which become unregulated during T-cell exhaustion in pancreatic cancer would be important for development of effective therapy (62). Because checkpoint inhibitors targeting PD-1/PD-L1 and CTLA4 are effective in a large portion of patients with melanoma, the same compounds represent the first checkpoint blockade agents tested against PDAC, with disappointing results as previously mentioned. In murine models of PDAC, it has been described that CTLA-4 inhibition was not sufficient to increase infiltration of CD8+ T cells in the pancreatic TME (63).

A recent study compared the density of immune cells expressing PD-1/PD-L1, and found them significantly higher in melanoma compared with PDAC (64). The same study reported over-expression of V-domain Ig suppressor of T-cell activation (VISTA), an inhibitory checkpoint molecule, in PDAC in comparison with melanoma, and postulated that VISTA represents a more dominant inhibitory pathway and may represent a more efficacious target for immunotherapy in PDAC (64).

LAG-3 represents another inhibitory molecule present on PDAC TILs which works by binding MHCII molecules on tumor cells. LAG-3 has been found upregulated on infiltrating lymphocytes in PDAC (65). Galectins, in particular Gal1, Gal3, and Gal9, have been found to be upregulated in the human PDAC TME (66). Galectin signaling has been linked to tumor-cell proliferation and T-lymphocyte apoptosis. Gal9 binds to the TIM-3, an inhibitory receptor expressed in T cells and NK cells (67). Inhibition of Gal9 renders PDAC sensitive to immunotherapy (68). Another inhibitory receptor is TIGIT, expressed on NK cells and T cells, which is capable of directly inhibiting T-cell proliferation and cytokine secretion (69, 70). Balli and colleagues found that cytolytic-high tumors had higher expression of several immune checkpoints, except for PD-L1 which was uniformly low. They also proposed categorizing PDAC based on coexpression of CTLA4, TIGIT, TIM-3, and VISTA for clinical targeting purposes (71). Combinatorial therapies targeting these various novel receptors, depending on an individual patient's expression, require further exploration, as they represent a potential promising approach for PDAC treatment.

The low surface presentation of neoantigens in PDAC has been postulated as one of the reasons that explain why PDAC is considered an “immunologically cold” tumor, unresponsive to most immunotherapies (72). Neoantigen abundance has been linked to mutational burden and lymphocyte infiltration, with melanomas and RCC having the highest number of mutations, and with pancreatic cancer having the lowest. Mutant-specific neoantigens are also associated with TILs in both tumor types as well (73). The extreme example of correlation between mutations, neoantigen abundance, and lymphocyte infiltration is represented by tumors with mismatch repair impairment (74). Unfortunately, this tumor subtype represents less than 1% of all cases of PDAC (75).

A study that analyzed publicly available data from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) revealed that while PDAC tumors lack the neoantigen diversity of melanomas, most tumors have mutations that result in candidate neoantigens, many of which would be expected to have efficient presentation by HLA class I (76). The same study also showed that despite the lower number of neoantigens, T-cell infiltration in PDAC was not very different from melanoma, although the cytotoxic capacity of those T cells was markedly lower in PDAC (76).

Consistent with these findings, Balli and colleagues found that rather than being associated with mutational burden or neoepitope abundance, immune activation in PDAC was inversely associated with expression of genes related to pancreatic differentiation status, suggesting that intrinsic oncogenic processes drive immune reactivity in PDAC (71).

A study which sequenced and analyzed TCRs from both PDAC short-term and long-term survivors revealed that the quality of neoantigens, rather than the quantity, was predictive of long-term survivorship (77). The study also suggested that neoantigens which mimic microbial epitopes have higher likelihood of TCR recognition. A recent study postulated that melanoma cancer cells can present peptides derived from intracellular bacteria on their HLA-I and HLA-II molecules to elicit immune reactivity. More functional work is required to further validate if these mechanisms may influence in vivo immune activation and responses to therapy (78).

The role of the gut microbiome on tumor responses to chemotherapy and immunotherapy has been extensively studied in several tumor types (79–82). Preclinical studies have shown that bacterial ablation by antibiotics or generation of germ-free mouse models blunt responses to immunotherapy and chemotherapy (83, 84). However, pancreatic cancer does not seem to follow the same pattern. As observed in several studies, PDAC-associated microbes have been associated with immunosuppression, and their ablation is linked to improved responses to therapies. In genetically engineered mouse models (GEMM) of PDAC, studies have shown differential evolution of the gut microbiome throughout the pancreatic tumorigenesis process compared with control mice (85–87). Rederivation of GEMM in germ-free conditions resulted in delayed tumorigenesis, which was rescued by oral reconstitution with fecal content from cancer mice. Furthermore, bacterial ablation in pancreatic cancer mouse models led to a reduction in immune-suppressive M2-like tumor-associated macrophages, an increased intratumoral CD8:CD4 T-cell ratio, as well as an increase in PD-1 expression (85). Combination therapy of anti-PD-1 antibodies and antibiotics resulted in synergistic antitumoral effect when compared with anti-PD-1 therapy alone (85). A second study showed the effect of antibiotics in slowing growth in an implantable subcutaneous model of pancreatic cancer, as well as a reduction in metastases (88).

In summary, data in PDAC suggests that antibiotics can potentially improve therapy responses. However, careful consideration must be given to this strategy, considering the toxicities associated with long-term use of broad-spectrum antibiotics including the acquisition of antibiotic multi-resistance.

Human studies have reported that patients with pancreatic cancer also present a different gut microbiome with lower alpha-diversity (85, 89). Geller and colleagues have described that pancreatic tumors harbor their own microbiome, with Gammaproteobacteria being the most abundant bacteria present in PDAC specimens (90). This bacteria class has been linked with resistance to the chemotherapy drug gemcitabine through the expression of the bacterial enzyme cytidine deaminase, which metabolizes gemcitabine to its inactive form (90). Riquelme and colleagues have analyzed the microbial content of tumors from pancreatic cancer short-term survivors (STS) and long-term survivors (LTS) from two geographically distant cohorts and found higher tumor microbial diversity LTS tumors. Furthermore, a tumor microbial signature was detected in PDAC LTS which directly associated with higher infiltration of cytotoxic T cells (91).

Humanized microbial mouse (HMM) models have been generated through transfer of human fecal microbial transplants (FMT) into mice transplanted with orthotropic pancreatic tumors. These models showed a protective effect in tumor growth when using FMT from LTS donors with no evidence of disease and healthy control donors (to a smaller extent), compared with tumors from mice who received FMT from patients with pancreatic cancer which had the largest tumors. These finding suggest that the gut microbial dysbiosis present in PDAC supports tumor growth, and its disruption is fundamental for antitumoral efficacy (91). The protective effect of FMT was lost when mice had no CD8+ T cells, providing preliminary evidence that modification of the gut/tumor microbiome is able to induce an antitumor response and activation of the immune system in tumor-bearing mice (92). A clinical trial that utilizes FMT in patients with resected PDAC is currently being launched at MD Anderson Cancer Center. Very recently, two publications have reported approximately 30% responses when FMT from responders to immunotherapy was combined with reinduction with checkpoint blockade for patients with melanoma who were considered refractory to immunotherapy (93, 94).

Treatment with T cells transduced with a chimeric antigen receptor (CAR-T) is a novel treatment approach (95) that has shown remarkable efficacy in some blood cancers but has had limited responses in most solid tumors including PDAC (95–98). Early trials using CAR designs that lacked a costimulatory domain and provided signaling only through CD3 were universally unsuccessful due to poor persistence of the CAR-T and early development of hypofunction. The introduction of the costimulatory domain, along with the practice of lymphodepletion preceding treatment led to the first signs of high efficacy in B-cell–derived cancers we see today (99, 100). Unfortunately, these innovations have not been sufficient to drive to clinical success in PDAC.

One challenge in solid tumors is finding a safe antigen to target. However, there are targets that are enriched on the surface of solid tumor cells compared with normal epithelial tissue, suggesting that there are CAR-T doses at which efficacy can be achieved without causing lethal destruction of epithelial tissue (101–103). Target engagement between CAR-T and PDAC cells occurs slowly, creating an additional challenge, as T cells, once activated and exposed to consistent antigen, begin a process of terminal differentiation towards an exhausted and hypo-functional state (29).

PDAC has relatively few endogenous cancer reactive T cells (102). CAR-T provides additional reactive T cells that can then be enhanced with other immunotherapies (104). CAR-T has the potential to be more potent than endogenous T cells as they can be easily engineered ex vivo. As an example, enforced expression of cDNA encoding the chemokine receptor CCR2, found in the myeloid lineage cells that infiltrate PDAC in great numbers, led to an increase in trafficking by CAR-T to tumors (105). Other examples include the use of switch receptors to change signaling effects of suppressive compounds (TGFBR and FAS switch receptors) from suppressive to stimulatory (106). To address CAR-T exhaustion, regions of the IL2 receptor were included into the CAR construct, resulting in IL2/JAK/STAT signaling upon target engagement, which delays exhaustion (107). Research points to the transcription factors AP1, TCF7, and c-Jun signaling as being key to maintaining a memory-like phenotype and the capacity for long-term function. Enforced expression of those transcription factors alongside CAR can prevent an exhaustion phenotype (108, 109).

CAR-T can also be engineered to secrete cytokines such as IL12 or IL18, or other immune stimulants such as flagellin, and that such secretion enhances CAR-T potency and endogenous T-cell recruitment (110, 111). CAR-T can be engineered for PDAC therapy to secrete cytokines that bias them and other endogenous T cells toward a memory phenotype, such as IL7 or chemokines to promote recruitment of endogenous immune cells, such as CCL19 (112, 113). Perhaps most exciting are examples demonstrating use of CAR-T to secrete scFv that block PD-1 itself, which could enable intratumoral immune activation without the toxicities of systemic delivery (114). A recent study describes synergy of joint administration of CAR-T following intratumoral injection with an oncolytic virus engineered to secrete the proinflammatory TNFα and the T-cell growth and survival factor IL2 (115). Another rational combination includes CAR-T with FAKi and checkpoint inhibition to clear stroma and enable better CAR-T trafficking (57, 116). Combining CAR-T with chemotherapeutics or radiation that enhance immunogenic cell death also seems promising (77, 117). Finally, treatment with targeted inhibitors that slow PDAC growth while sparing T cells could give CAR-T a “fighting chance” against large, established, aggressively proliferating PDAC tumors.

DNA damage repair deficiency not as promising in PDAC

Cells are capable of inducing five types of DNA damage pathways with the goal of maintaining the integrity of their genome. DNA damage repair (DDR) pathways include mismatch repair (MMR), homologous recombination, nonhomologous end joining (NHEJ), base excision repair (BER), and nucleotide excision repair (NER; ref. 118). A deficiency in a DDR pathway conduces to genomic abnormalities that can result in tumorigenesis promotion (119) but also in therapeutic opportunities.

DDR defects are also relevant in inducing sensitivity and efficacy to immunotherapy. Microsatellite instability/mismatch repair deficiency (MSH/dMMR) is an FDA-approved biomarker for immune checkpoint blockade, independent of tumor types (120). MMR deficient tumors are presumably more susceptible to immune therapy due to high expression of PD-L1 and high numbers of infiltrating lymphocytes due to a more immunogenic phenotype compared to microsatellite stable tumors. Despite this, a recent study of pembrolizumab in patients with noncolorectal solid tumors that were MSI-high (MSI-H) showed a lower response in patients with PDAC. In the KEYNOTE-158 study, the overall response rate (ORR) was 34.3% in a population of patients with a variety of MSI-H solid tumors including endometrial, gastric, cholangiocarcinoma, and small intestine; however, ORR was 18.2% in the subset of patients with PDAC (121). This is likely due to the uniquely immune-suppressive TME of PDAC, as discussed below.

There is a paucity of data regarding the use of alternative biomarkers in PDAC such as tumor mutational burden (TMB). One study found a low rate of 0.5% of high TMB (defined as ≥ 20 mutations/megabase) in a targeted genomic analysis of over 3,500 PDAC samples (122). The KEYNOTE-028 trial examined use of TMB as a predictive biomarker in patients receiving single-agent pembrolizumab across 20 different PD-L1–positive solid tumors. However, with an ORR of 0% (out of 24 patients included in the trial), patients with PDAC were excluded from biomarker analysis (123).

An example of another therapeutic opportunity based on a DDR pathway defect is represented by platinum agents and poly(ADP ribose) polymerase (PARP) inhibitors as therapy for HR aberrant tumors caused by mutations such as BRCA1/2 (124, 125). Recently, the POLO study showed that the PARP inhibitor olaparib is an efficient maintenance therapy for patients with metastatic PDAC and BRCA1/2 germline mutations, after platinum therapy (126). Zhang and colleagues showed that DNA repair inhibition by pharmacologic blockade or siRNA silencing of ataxia telangiectasia mutated (ATM) results in induction of type I IFN-mediated innate immune response in pancreatic cancer that is increased by radiation, and results in increased sensitivity to PD-L1 blockade (127). Ongoing trials are testing combination of therapies specific for DDR-deficient tumors (ex: PARP inhibitors) with immune checkpoint blockade (Table 1; ref. 128).

In the past decade, we have seen how immunotherapies have proven successful in many cancers. Unfortunately, this success has not been replicated in pancreatic cancer, which remains resistant to this breakthrough in oncologic care. The challenges cited in this review that are particularly potent in PDAC include poor T-cell trafficking to tumors, stromal barriers, abundance of immune suppressive cells and secreted factors, and T-cell exclusion and exhaustion. These various factors are summarized in Fig. 1.

Figure 1.

Approaches to overcoming resistance mechanisms to immunotherapy in PDAC. Illustration by Jordan Pietz (Medical Illustrator at the Creative Communications Department, MD Anderson Cancer Center). © 2021 The Board of Regents of the University of Texas System. Copyright used with the permission of The Board of Regents of the University of Texas System through The University of Texas MD Anderson Cancer Center.

Figure 1.

Approaches to overcoming resistance mechanisms to immunotherapy in PDAC. Illustration by Jordan Pietz (Medical Illustrator at the Creative Communications Department, MD Anderson Cancer Center). © 2021 The Board of Regents of the University of Texas System. Copyright used with the permission of The Board of Regents of the University of Texas System through The University of Texas MD Anderson Cancer Center.

Close modal

We expect that combinatorial therapy based on checkpoint inhibitors or CAR-T cells given in addition to compounds directed against the TME immunosuppressive components, as well as microbial modulation, may represent a potential avenue to efficacy in PDAC. Possibly, heterogeneity in individual patient TME characteristics, microbial composition, and immune checkpoint expression may require “personalized” immunotherapeutic combinatorial approaches. We look forward to results from these ongoing combinatorial trials.

P.K. Mazur is a scientific cofounder, consultant, and stockholder of Amplified Medicines, Inc. and Ikena Oncology, Inc. No disclosures were reported by the other authors.

This work was supported in part by grants to F. McAllister from the NCI (1R37CA237384–01A1), CPRIT (RP200173), V Foundation Translational Award, Andrew Sabin Family Fellowship, Philantropic Moonshot Programs (MDACC), and grants to P.K. Mazur from the NIH (R01 CA236118, CA236949), DOD PRCRP (CA181486), Neuroendocrine Tumor Research Foundation-AACR Grant (18–60–33-MAZU), the Andrew Sabin Family Foundation, and CPRIT Scholar in Cancer Research (RR160078).

1.
Waldman
AD
,
Fritz
JM
,
Lenardo
MJ
. 
A guide to cancer immunotherapy: from T cell basic science to clinical practice
.
Nat Rev Immunol
2020
;
20
:
651
68
.
2.
Ugurel
S
,
Röhmel
J
,
Ascierto
PA
,
Flaherty
KT
,
Grob
JJ
,
Hauschild
A
, et al
Survival of patients with advanced metastatic melanoma: the impact of novel therapies – 2017 update
.
Eur J Cancer
2017
;
83
:
247
57
.
3.
National Cancer Institute: Surveillance, Epidemiology, and End Results Program
. 
Cancer Stat Facts: pancreatic cancer
. 
2019
.
Available from
: https://seer.cancer.gov/statfacts/html/pancreas.html.
4.
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
.
5.
Conroy
T
,
Hammel
P
,
Hebbar
M
,
Ben Abdelghani
M
,
Wei
AC
,
Raoul
JL
, et al
FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer
.
N Engl J Med
2018
;
379
:
2395
406
.
6.
Conroy
T
,
Desseigne
F
,
Ychou
M
,
Bouché
O
,
Guimbaud
R
,
Bécouarn
Y
, et al
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med
2011
;
364
:
1817
25
.
7.
Von Hoff
DD
,
Ervin
T
,
Arena
FP
,
Chiorean
EG
,
Infante
J
,
Moore
M
, et al
Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine
.
N Engl J Med
2013
;
369
:
1691
703
.
8.
Henriksen
A
,
Dyhl-Polk
A
,
Chen
I
,
Nielsen
D
. 
Checkpoint inhibitors in pancreatic cancer
.
Cancer Treat Rev
2019
;
78
:
17
30
.
9.
Banerjee
K
,
Kumar
S
,
Ross
KA
,
Gautam
S
,
Poelaert
B
,
Nasser
MW
, et al
Emerging trends in the immunotherapy of pancreatic cancer
.
Cancer Lett
2018
;
417
:
35
46
.
10.
Rosenberg
A
,
Mahalingam
D
. 
Immunotherapy in pancreatic adenocarcinoma – overcoming barriers to response
.
J Gastrointest Oncol
2018
;
9
:
143
59
.
11.
Eibl
G
,
Cruz-Monserrate
Z
,
Korc
M
,
Petrov
MS
,
Goodarzi
MO
,
Fisher
WE
, et al
Diabetes mellitus and obesity as risk factors for pancreatic cancer
.
J Acad Nutr Diet
2018
;
118
:
555
67
.
12.
Gomez-Chou
SB
,
Swidnicka-Siergiejko
AK
,
Badi
N
,
Chavez-Tomar
M
,
Lesinski
GB
,
Bekaii-Saab
T
, et al
Lipocalin-2 promotes pancreatic ductal adenocarcinoma by regulating inflammation in the tumor microenvironment
.
Cancer Res
2017
;
77
:
2647
60
.
13.
Noy
R
,
Pollard
JW
. 
Tumor-associated macrophages: from mechanisms to therapy
.
Immunity
2014
;
41
:
49
61
.
14.
Ren
B
,
Cui
M
,
Yang
G
,
Wang
H
,
Feng
M
,
You
L
, et al
Tumor microenvironment participates in metastasis of pancreatic cancer
.
Mol Cancer
2018
;
17
:
108
.
15.
Groth
C
,
Hu
X
,
Weber
R
,
Fleming
V
,
Altevogt
P
,
Utikal
J
, et al
Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression
.
Br J Cancer
2019
;
120
:
16
25
.
16.
Zhang
Y
,
Velez-Delgado
A
,
Mathew
E
,
Li
D
,
Mendez
FM
,
Flannagan
K
, et al
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
36
.
17.
Tang
Y
,
Xu
X
,
Guo
S
,
Zhang
C
,
Tang
Y
,
Tian
Y
, et al
An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma
.
PLoS One
2014
;
9
:
e91551
.
18.
Zhang
Y
,
Lazarus
J
,
Steele
NG
,
Yan
W
,
Lee
HJ
,
Nwosu
ZC
, et al
Regulatory T cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis
.
Cancer Discov
2020
;
10
:
422
39
.
19.
McAllister
F
,
Bailey
JM
,
Alsina
J
,
Nirschl
CJ
,
Sharma
R
,
Fan
H
, et al
Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia
.
Cancer Cell
2014
;
25
:
621
37
.
20.
Bayne
LJ
,
Beatty
GL
,
Jhala
N
,
Clark
CE
,
Rhim
AD
,
Stanger
BZ
, et al
Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer
.
Cancer Cell
2012
;
21
:
822
35
.
21.
Steele
CW
,
Karim
SA
,
Leach
JDG
,
Bailey
P
,
Upstill-Goddard
R
,
Rishi
L
, et al
CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma
.
Cancer Cell
2016
;
29
:
832
45
.
22.
Michaeli
J
,
Shaul
ME
,
Mishalian
I
,
Hovav
AH
,
Levy
L
,
Zolotriov
L
, et al
Tumor-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFalpha and NO-dependent mechanism, promoting a tumor-supportive environment
.
Oncoimmunology
2017
;
6
:
e1356965
.
23.
Masucci
MT
,
Minopoli
M
,
Carriero
MV
. 
Tumor associated neutrophils. their role in tumorigenesis, metastasis, prognosis and therapy
.
Front Oncol
2019
;
9
:
1146
.
24.
Zhang
Y
,
Chandra
V
,
Riquelme Sanchez
E
,
Dutta
P
,
Quesada
PR
,
Rakoski
A
, et al
Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer
.
J Exp Med
2020
;
217
:
e20190354
.
25.
Fukumura
D
,
Kloepper
J
,
Amoozgar
Z
,
Duda
DG
,
Jain
RK
. 
Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges
.
Nat Rev Clin Oncol
2018
;
15
:
325
40
.
26.
Lee
WS
,
Yang
H
,
Chon
HJ
,
Kim
C
. 
Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity
.
Exp Mol Med
2020
;
52
:
1475
85
.
27.
Young
KHD
,
Cunningham
D
,
Starling
N
. 
Immunotherapy and pancreatic cancer: unique challenges and potential opportunities
.
Ther Adv Med Oncol
2018
;
10
:
1
20
.
28.
Nywening
TM
,
Belt
BA
,
Cullinan
DR
,
Panni
RZ
,
Han
BJ
,
Sanford
DE
, et al
Targeting both tumour-associated CXCR2(+) neutrophils and CCR2(+) macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma
.
Gut
2018
;
67
:
1112
23
.
29.
Anderson
KG
,
Stromnes
IM
,
Greenberg
PD
. 
Obstacles posed by the tumor microenvironment to T cell activity: A case for synergistic therapies
.
Cancer Cell
2017
;
31
:
311
25
.
30.
Gajiwala
S
,
Torgeson
A
,
Garrido-Laguna
I
,
Kinsey
C
,
Lloyd
S
. 
Combination immunotherapy and radiation therapy strategies for pancreatic cancer-targeting multiple steps in the cancer immunity cycle
.
J Gastrointest Oncol
2018
;
9
:
1014
26
.
31.
Ruarus
AH
,
Vroomen
L
,
Geboers
B
,
van Veldhuisen
E
,
Puijk
RS
,
Nieuwenhuizen
S
, et al
Percutaneous irreversible electroporation in locally advanced and recurrent pancreatic cancer (PANFIRE-2): a multicenter, prospective, single-arm, phase II study
.
Radiology.
2020
;
294
:
212
20
.
32.
Margolin
K
,
Morishima
C
,
Velcheti
V
,
Miller
JS
,
Lee
SM
,
Silk
AW
, et al
Phase I trial of ALT-803, a novel recombinant IL15 complex, in patients with advanced solid tumors
.
Clin Cancer Res
2018
;
24
:
5552
61
.
33.
Greenhalf
W
,
Thomas
A
. 
Combination therapy for the treatment of pancreatic cancer
.
Anticancer Agents Med Chem
2011
;
11
:
418
26
.
34.
Melisi
D
,
Calvetti
L
,
Frizziero
M
,
Tortora
G
. 
Pancreatic cancer: systemic combination therapies for a heterogeneous disease
.
Curr Pharm Des
2014
;
20
:
6660
9
.
35.
Seo
YD
,
Jiang
X
,
Sullivan
KM
,
Jalikis
FG
,
Smythe
KS
,
Abbasi
A
, et al
Mobilization of CD8+ T cells via CXCR4 blockade facilitates PD-1 checkpoint therapy in human pancreatic cancer
.
Clin Cancer Res
2019
;
25
:
3934
45
.
36.
Carstens
JL
,
de Sampaio
PC
,
Yang
D
,
Barua
S
,
Wang
H
,
Rao
A
, et al
Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer
.
Nat Commun
2017
;
8
:
1
12
.
37.
Masugi
Y
,
Abe
T
,
Ueno
A
,
Fujii-Nishimura
Y
,
Ojima
H
,
Endo
Y
, et al
Characterization of spatial distribution of tumor-infiltrating CD8+ T cells refines their prognostic utility for pancreatic cancer survival
.
Mod Pathol
2019
;
32
:
1495
507
.
38.
Zhang
J
,
Wang
YF
,
Wu
B
,
Zhong
ZX
,
Wang
KX
,
Yang
LQ
, et al
Intraepithelial attack rather than intratumoral infiltration of CD8+ T lymphocytes is a favorable prognostic indicator in pancreatic ductal adenocarcinoma
.
Curr Mol Med
2017
;
17
:
689
98
.
39.
Vonderheide
R
. 
The immune revolution: a case for priming, not checkpoint
.
Cancer Cell
2018
;
33
:
563
9
.
40.
Byrne
KT
,
Vonderheide
RH
. 
CD40 stimulation obviates innate sensors and drives T cell immunity in cancer
.
Cell Rep
2016
;
15
:
2719
32
.
41.
Winograd
R
,
Byrne
KT
,
Evans
RA
,
Odorizzi
PM
,
Meyer
AR
,
Bajor
DL
, et al
Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma
.
Cancer Immunol Res
2015
;
3
:
399
411
.
42.
Laheru
D
,
Lutz
E
,
Burke
J
,
Biedrzycki
B
,
Solt
S
,
Onners
B
, et al
Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation
.
Clin Cancer Res
2008
;
14
:
1455
63
.
43.
Le
DT
,
Wang-Gillam
A
,
Picozzi
V
,
Greten
TF
,
Crocenzi
T
,
Springett
G
, et al
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
.
44.
Thomas
AM
,
Santarsiero
LM
,
Lutz
ER
,
Armstrong
TD
,
Chen
YC
,
Huang
LQ
, et al
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
.
45.
Soares
KC
,
Rucki
AA
,
Wu
AA
,
Olino
K
,
Xiao
Q
,
Chai
Y
, et al
PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors
.
J Immunother
2015
;
38
:
1
11
.
46.
Le
DT
,
Lutz
E
,
Uram
JN
,
Sugar
EA
,
Onners
B
,
Solt
S
, et al
Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer
.
J Immunother
2013
;
36
:
382
9
.
47.
Tsujikawa
T
,
Crocenzi
T
,
Durham
JN
,
Sugar
EA
,
Wu
AA
,
Onners
B
, et al
Evaluation of cyclophosphamide/GVAX pancreas followed by listeria-mesothelin (CRS-207) with or without nivolumab in patients with pancreatic cancer
.
Clin Cancer Res
2020
;
26
:
3578
88
.
48.
Liang
C
,
Shi
S
,
Meng
Q
,
Liang
D
,
Ji
S
,
Zhang
B
, et al
Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going
.
Exp Mol Med
2017
;
49
:
e406
.
49.
Li
C
,
Cui
L
,
Yang
L
,
Wang
B
,
Zhuo
Y
,
Zhang
L
, et al
Pancreatic stellate cells promote tumor progression by promoting an immunosuppressive microenvironment in murine models of pancreatic cancer
.
Pancreas
2020
;
49
:
120
7
.
50.
Sherman
MH
,
Yu
RT
,
Engle
DD
,
Ding
N
,
Atkins
AR
,
Tiriac
H
, et al
Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy
.
Cell
2014
;
159
:
80
93
.
51.
Pitarresi
JR
,
Liu
X
,
Avendano
A
,
Thies
KA
,
Sizemore
GM
,
Hammer
AM
, et al
Disruption of stromal hedgehog signaling initiates RNF5-mediated proteasomal degradation of PTEN and accelerates pancreatic tumor growth
.
Life Science Alliance
2018
;
1
:
1
12
.
52.
Olive
KP
,
Jacobetz
MA
,
Davidson
CJ
,
Gopinathan
A
,
McIntyre
D
,
Honess
D
, et al
Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer
.
Science
2009
;
324
:
1457
61
.
53.
De Jesus-Acosta
A
,
Sugar
EA
,
O'Dwyer
PJ
,
Ramanathan
RK
,
Von Hoff
DD
,
Rasheed
Z
, et al
Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma
.
Br J Cancer
2020
;
122
:
498
505
.
54.
Kim
EJ
,
Sahai
V
,
Abel
EV
,
Griffith
KA
,
Greenson
JK
,
Takebe
N
, et al
Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma
.
Clin Cancer Res
2014
;
20
:
5937
45
.
55.
Rhim
AD
,
Oberstein
PE
,
Thomas
DH
,
Mirek
ET
,
Palermo
CF
,
Sastra
SA
, et al
Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma
.
Cancer Cell
2014
;
25
:
735
47
.
56.
Ozdemir
BC
,
Pentcheva-Hoang
T
,
Carstens
JL
,
Zheng
X
,
Wu
CC
,
Simpson
TR
, et al
Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppresion and accelerates pancreas cancer with reduced survival
.
Cancer Cell
2014
;
25
:
719
34
.
57.
Jiang
H
,
Hegde
S
,
Knolhoff
BL
,
Zhu
Y
,
Herndon
JM
,
Meyer
MA
, et al
Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy
.
Nat Med
2016
;
22
:
851
60
.
58.
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
.
59.
Wong
KM
,
Horton
KJ
,
Coveler
AL
,
Hingorani
SR
,
Harris
WP
. 
Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20)
.
Curr Oncolo Rep
2017
;
19
:
1
9
.
60.
Ramanathan
RK
,
McDonough
SL
,
Philip
PA
,
Hingorani
SR
,
Lacy
J
,
Kortmansky
JS
, et al
Phase Ib/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313
.
J Clin Onc
2019
;
37
:
1062
9
.
61.
Van Cutsem
E
,
Tempero
MA
,
Sigal
D
,
Oh
DY
,
Fazio
N
,
Macarulla
T
, et al
Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma
.
J Clin Oncol
2020
;
38
:
3185
94
.
62.
Saka
D
,
Gokalp
M
,
Piyade
B
,
Cevik
NC
,
Arik Sever
E
,
Unutmaz
D
, et al
Mechanisms of T-cell exhaustion in pancreatic cancer
.
Cancers
2020
;
12
:
2274
.
63.
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
17
.
64.
Blando
J
,
Sharma
A
,
Higa
MG
,
Zhao
H
,
Vence
L
,
Yadav
SS
, et al
Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer
.
Proc Natl Acd Sci U S A
2019
;
116
:
1692
7
.
65.
Meng
Q
,
Liu
Z
,
Rangelova
E
,
Poiret
T
,
Ambati
A
,
Rane
L
, et al
Expansion of tumor-reactive T cells from patients with pancreatic cancer
.
J Immunother
2016
;
39
:
81
9
.
66.
Manero-Ruperez
N
,
Martinez-Bosch
N
,
Barranco
LE
,
Visa
L
,
Navarro
P
. 
The galectin family as molecular targets: hopes for defeating pancreatic cancer
.
Cells
2020
;
9
:
689
.
67.
Nebbia
G
,
Peppa
D
,
Schurich
A
,
Khanna
P
,
Singh
HD
,
Cheng
Y
, et al
Upregulation of the Tim-3/galectin-9 pathway of T cell exhaustion in chronic hepatitis B virus infection
.
PLoS One
2012
;
7
:
e47648
.
68.
Yazdanifar
M
,
Zhou
R
,
Grover
P
,
Williams
C
,
Bose
M
,
Moore
LJ
, et al
Overcoming immunological resistance enhances the efficacy of a novel anti-tMUC1-CAR T cell treatment against pancreatic ductal adenocarcinoma
.
Cells
2019
;
8
:
1070
.
69.
Stanietsky
N
,
Simic
H
,
Arapovic
J
,
Toporik
A
,
Levy
O
,
Novik
A
, et al
The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity
.
Proc Natl Acad Sci U S A
2009
;
106
:
17858
63
.
70.
Yu
X
,
Harden
K
,
Gonzalez
LC
,
Francesco
M
,
Chiang
E
,
Irving
B
, et al
The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells
.
Nat Immunol
2009
;
10
:
48
57
.
71.
Balli
D
,
Rech
AJ
,
Stanger
BZ
,
Vonderheide
RH
. 
Immune cytolytic activity stratifies molecular subsets of human pancreatic cancer
.
Clin Cancer Res
2017
;
23
:
3129
38
.
72.
Bailey
P
,
Chang
DK
,
Forget
MA
,
Lucas
FA
,
Alvarez
HA
,
Haymaker
C
, et al
Exploiting the neoantigen landscape for immunotherapy of pancreatic ductal adenocarcinoma
.
Sci Rep
2016
;
6
:
35848
.
73.
Cancer Genome Atlas Network
. 
Genomic classification of cutaneous melanoma
.
Cell
2015
;
161
:
1681
96
.
74.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
75.
Hu
ZI
,
Shia
J
,
Stadler
ZK
,
Varghese
AM
,
Capanu
M
,
Salo-Mullen
E
, et al
Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: challenges and recommendations
.
Clin Cancer Res
2018
;
24
:
1326
36
.
76.
Bailey
P
,
Chang
DK
,
Nones
K
,
Johns
AL
,
Patch
AM
,
Gingras
MC
, et al
Genomic analyses identify molecular subtypes of pancreatic cancer
.
Nature
2016
;
531
:
47
52
.
77.
Balachandran
VP
,
Luksza
M
,
Zhao
JN
,
Makarov
V
,
Moral
JA
,
Remark
R
, et al
Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer
.
Nature
2017
;
551
:
512
6
.
78.
Kalaora
S
,
Nagler
A
,
Nejman
D
,
Alon
M
,
Barbolin
C
,
Barnea
E
, et al
Identification of bacteria-derived HLA-bound peptides in melanoma
.
Nature
2021
;
592
:
138
43
.
79.
Viaud
S
,
Saccheri
F
,
Mignot
G
,
Yamazaki
T
,
Daillere
R
,
Hannani
D
, et al
The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide
.
Science
2013
;
342
:
971
6
.
80.
Vetizou
M
,
Pitt
JM
,
Daillere
R
,
Lepage
P
,
Waldschmitt
N
,
Flament
C
, et al
Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota
.
Science
2015
;
350
:
1079
84
.
81.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
,
Reuben
A
,
Andrews
MC
,
Karpinets
TV
, et al
Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients
.
Science
2018
;
359
:
97
103
.
82.
Routy
B
,
Le Chatelier
E
,
Derosa
L
,
Duong
CPM
,
Alou
MT
,
Daillere
R
, et al
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
.
Science
2018
;
359
:
91
7
.
83.
Matson
V
,
Fessler
J
,
Bao
R
,
Chongsuwat
T
,
Zha
Y
,
Alegre
ML
, et al
The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients
.
Science
2018
;
359
:
104
8
.
84.
Iida
N
,
Dzutsev
A
,
Stewart
CA
,
Smith
L
,
Bouladoux
N
,
Weingarten
RA
, et al
Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment
.
Science
2013
;
342
:
967
70
.
85.
Pushalkar
S
,
Hundeyin
M
,
Daley
D
,
Zambirinis
CP
,
Kurz
E
,
Mishra
A
, et al
The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression
.
Cancer Discov
2018
;
8
:
403
16
.
86.
Mendez
R
,
Kesh
K
,
Arora
N
,
Di Martino
L
,
McAllister
F
,
Merchant
N
, et al
Microbial dysbiosis and polyamine metabolism as predictive markers for early detection of pancreatic cancer
.
Carcinogenesis
2020
;
41
:
561
70
.
87.
Thomas
RM
,
Jobin
C
. 
Microbiota in pancreatic health and disease: the next frontier in microbiome research
.
Nat Rev Gastroenterol Hepatol
2020
;
17
:
53
64
.
88.
Sethi
V
,
Kurtom
S
,
Tarique
M
,
Lavania
S
,
Malchiodi
Z
,
Hellmund
L
, et al
Gut microbiota promotes tumor growth in mice by modulating immune response
.
Gastroenterology
2018
;
155
:
33
7
.
89.
Ren
Z
,
Jiang
J
,
Xie
H
,
Li
A
,
Lu
H
,
Xu
S
, et al
Gut microbial profile analysis by MiSeq sequencing of pancreatic carcinoma patients in China
.
Oncotarget
2017
;
8
:
95176
91
.
90.
Geller
LT
,
Barzily-Rokni
M
,
Danino
T
,
Jonas
OH
,
Shental
N
,
Nejman
D
, et al
Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine
.
Science
2017
;
357
:
1156
60
.
91.
Riquelme
E
,
Maitra
A
,
McAllister
F
. 
Immunotherapy for pancreatic cancer: more than just a gut feeling
.
Cancer Discov
2018
;
8
:
386
8
.
92.
Riquelme
E
,
Zhang
Y
,
Zhang
L
,
Jenq
R
,
Wargo
J
,
McAllister
F
, et al
Tumor microbiome diversity and composition influence pancreatic cancer outcomes
.
Cell
2019
;
178
:
795
806
.
93.
Baruch
EN
,
Youngster
I
,
Ben-Betzalel
G
,
Ortenberg
R
,
Lahat
A
,
Katz
L
, et al
Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients
.
Science
2021
;
371
:
602
9
.
94.
Davar
D
,
Dzutsev
AK
,
McCulloch
JA
,
Rodrigues
RR
,
Chauvin
JM
,
Morrison
RM
, et al
Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients
.
Science
2021
;
371
:
595
602
.
95.
June
CH
,
O'Connor
RS
,
Kawalekar
OU
,
Ghassemi
S
,
Milone
MC
. 
CAR T cell immunotherapy for human cancer
.
Science
2018
;
359
:
1361
5
.
96.
Maude
SL
,
Frey
N
,
Shaw
PA
,
Aplenc
R
,
Barrett
DM
,
Bunin
NJ
, et al
Chimeric antigen receptor T cells for sustained remissions in leukemia
.
N Engl J Med
2014
;
371
:
1507
17
.
97.
Morello
A
,
Sadelain
M
,
Adusumilli
PS
. 
Mesothelin-targeted CARs: driving T cells to solid tumors
.
Cancer Discov
2016
;
6
:
133
46
.
98.
Beatty
GL
,
O'Hara
MH
,
Lacey
SF
,
Torigian
DA
,
Nazimuddin
F
,
Chen
F
, et al
Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial
.
Gastroenterology
2018
;
155
:
29
32
.
99.
Eshhar
Z
,
Waks
T
,
Gross
G
,
Schindler
DG
. 
Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors
.
Proc Natl Acad Sci U S A
1993
;
90
:
720
4
.
100.
Frigault
MJ
,
Lee
J
,
Basil
MC
,
Carpenito
C
,
Motohashi
S
,
Scholler
J
, et al
Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells
.
Cancer Immunol Res
2015
;
3
:
356
67
.
101.
Beatty
GL
,
Haas
AR
,
Maus
MV
,
Torigian
DA
,
Soulen
MC
,
Plesa
G
, et al
Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies
.
Cancer Immunol Res
2014
;
2
:
112
20
.
102.
Stromnes
IM
,
Schmitt
TM
,
Hulbert
A
,
Brockenbrough
JS
,
Nguyen
H
,
Cuevas
C
, et al
T cells engineered against a native antigen can surmount immunologic and physical barriers to treat pancreatic ductal adenocarcinoma
.
Cancer Cell
2015
;
28
:
638
52
.
103.
Du
H
,
Hirabayashi
K
,
Ahn
S
,
Kren
NP
,
Montgomery
SA
,
Wang
X
, et al
Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells
.
Cancer Cell
2019
;
35
:
221
37
.
104.
Mohammed
S
,
Sukumaran
S
,
Bajgain
P
,
Watanabe
N
,
Heslop
HE
,
Rooney
CM
, et al
Improving chimeric antigen receptor-modified T cell function by reversing the immunosuppressive tumor microenvironment of pancreatic cancer
.
Mol Ther
2017
;
25
:
249
58
.
105.
Moon
EK
,
Carpenito
C
,
Sun
J
,
Wang
LC
,
Kapoor
V
,
Predina
J
, et al
Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor
.
Clin Cancer Res
2011
;
17
:
4719
30
.
106.
Leen
AM
,
Sukumaran
S
,
Watanabe
N
,
Mohammed
S
,
Keirnan
J
,
Yanagisawa
R
, et al
Reversal of tumor immune inhibition using a chimeric cytokine receptor
.
Mol Ther
2014
;
22
:
1211
20
.
107.
Kagoya
Y
,
Tanaka
S
,
Guo
T
,
Anczurowski
M
,
Wang
CH
,
Saso
K
, et al
A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects
.
Nat Med
2018
;
24
:
352
9
.
108.
Sade-Feldman
M
,
Yizhak
K
,
Bjorgaard
SL
,
Ray
JP
,
de Boer
CG
,
Jenkins
RW
, et al
Defining T cell states associated with response to checkpoint immunotherapy in melanoma
.
Cell
2018
;
175
:
998
1013
.
109.
Lynn
RC
,
Weber
EW
,
Sotillo
E
,
Gennert
D
,
Xu
P
,
Good
Z
, et al
c-Jun overexpression in CAR T cells induces exhaustion resistance
.
Nature
2019
;
576
:
293
300
.
110.
Pegram
HJ
,
Lee
JC
,
Hayman
EG
,
Imperato
GH
,
Tedder
TF
,
Sadelain
M
, et al
Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning
.
Blood
2012
;
119
:
4133
41
.
111.
Chmielewski
M
,
Abken
H
. 
CAR T cells releasing IL-18 convert to T-Bet(high) FoxO1(low) effectors that exhibit augmented activity against advanced solid tumors
.
Cell Rep
2017
;
21
:
3205
19
.
112.
Shum
T
,
Omer
B
,
Tashiro
H
,
Kruse
RL
,
Wagner
DL
,
Parikh
K
, et al
Constitutive signaling from an engineered IL7 receptor promotes durable tumor elimination by tumor-redirected T cells
.
Cancer Discov
2017
;
7
:
1238
47
.
113.
Adachi
K
,
Kano
Y
,
Nagai
T
,
Okuyama
N
,
Sakoda
Y
,
Tamada
K
. 
IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor
.
Nat Biotechnol
2018
;
36
:
346
51
.
114.
Rafiq
S
,
Yeku
OO
,
Jackson
HJ
,
Purdon
TJ
,
van Leeuwen
DG
,
Drakes
DJ
, et al
Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo
.
Nat Biotechnol
2018
;
36
:
847
56
.
115.
Tahtinen
S
,
Blattner
C
,
Vaha-Koskela
M
,
Saha
D
,
Siurala
M
,
Parviainen
S
, et al
T-cell therapy enabling adenoviruses coding for IL2 and TNFalpha induce systemic immunomodulation in mice with spontaneous melanoma
.
J Immunother
2016
;
39
:
343
54
.
116.
Grosser
R
,
Cherkassky
L
,
Chintala
N
,
Adusumilli
PS
. 
Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors
.
Cancer Cell
2019
;
36
:
471
82
.
117.
Alcantara
M
,
Du Rusquec
P
,
Romano
E
. 
Current clinical evidence and potential solutions to increase benefit of CAR T-cell therapy for patients with solid tumors
.
Oncoimmunology
2020
;
9
:
1777064
.
118.
Lord
CJ
,
Ashworth
A
. 
The DNA damage response and cancer therapy
.
Nature
2012
;
481
:
287
94
.
119.
Burrell
RA
,
McGranahan
N
,
Bartek
J
,
Swanton
C
. 
The causes and consequences of genetic heterogeneity in cancer evolution
.
Nature
2013
;
501
:
338
45
.
120.
Lemery
S
,
Keegan
P
,
Pazdur
R
. 
First FDA approval agnostic of cancer site - when a biomarker defines the indication
.
N Engl J Med
2017
;
377
:
1409
12
.
121.
Marabelle
A
,
Le
DT
,
Ascierto
PA
,
Di Giacomo
AM
,
De Jesus-Acosta
A
,
Delord
JP
, et al
Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the Phase II KEYNOTE-158 Study
.
J Clin Oncol
2020
;
38
:
1
10
.
122.
Singhi
AD
,
George
B
,
Greenbowe
JR
,
Chung
J
,
Suh
J
,
Maitra
A
, et al
Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers
.
Gastroenterology
2019
;
156
:
2242
53
.
123.
Ott
PA
,
Bang
YJ
,
Piha-Paul
SA
,
Razak
ARA
,
Bennouna
J
,
Soria
JC
, et al
T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028
.
J Clin Oncol
2019
;
37
:
318
27
.
124.
Rouleau
M
,
Patel
A
,
Hendzel
MJ
,
Kaufmann
SH
,
Poirier
GG
. 
PARP inhibition: PARP1 and beyond
.
Nat Rev Cancer
2010
;
10
:
293
301
.
125.
Farmer
H
,
McCabe
N
,
Lord
CJ
,
Tutt
AN
,
Johnson
DA
,
Richardson
TB
, et al
Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy
.
Nature
2005
;
434
:
917
21
.
126.
Golan
T
,
Hammel
P
,
Reni
M
,
Van Cutsem
E
,
Macarulla
T
,
Hall
MJ
, et al
Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer
.
N Engl J Med
2019
;
381
:
317
27
.
127.
Zhang
Q
,
Green
MD
,
Lang
X
,
Lazarus
J
,
Parsels
JD
,
Wei
S
, et al
Inhibition of ATM increases interferon signaling and sensitizes pancreatic cancer to immune checkpoint blockade therapy
.
Cancer Res
2019
;
79
:
3940
51
.
128.
Stewart
RA
,
Pilie
PG
,
Yap
TA
. 
Development of PARP and immune-checkpoint inhibitor combinations
.
Cancer Res
2018
;
78
:
6717
25
.
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs International 4.0 License.