Pancreatic ductal adenocarcinoma (PDAC) remains one of the most challenging cancers to treat. For patients with advanced and metastatic disease, chemotherapy has yielded only modest incremental benefits, which are not durable. Immunotherapy has revolutionized the treatment of other solid tumors by leading to cures where none existed only a decade ago, yet it has made few inroads with PDAC. A host of trials with promising preclinical data have failed, except for in a small minority of patients with selected biomarkers. There is, however, a glimmer of hope, which we seek to cultivate. In this review, we discuss recent advances in the understanding of the uniquely immunosuppressive tumor microenvironment (TME) in PDAC, learnings from completed trials of checkpoint inhibitors, TME modifiers, cellular and vaccine therapies, oncolytic viruses, and other novel approaches. We go on to discuss our expectations for improved preclinical models of immunotherapy in PDAC, new approaches to modifying the TME including the myeloid compartment, and emerging biomarkers to better select patients who may benefit from immunotherapy. We also discuss improvements in clinical trial design specific to immunotherapy that will help us better measure success when we find it. Finally, we discuss the urgent imperative to better design and execute bold, but rational, combination trials of novel agents designed to cure patients with PDAC.

Pancreatic ductal adenocarcinoma (PDAC), lying at the other end of the spectrum from melanoma with regards to response to immunotherapy, has stolen from it the dubious distinction of being called “the cancer that gives cancer a bad name.” While the rise of agents targeting the immune system has led to a renaissance in cancer care overall, immunotherapy remains largely ineffective in PDAC. Due in part to our improved understanding of basic cancer biology, mechanisms of drug resistance, and advances in drug, vaccine, and cell therapy discovery, better therapeutic options are on the horizon. Further, an increasing number of rationally designed clinical trials featuring novel treatment combinations are becoming available for patients with PDAC, paving the way for a brighter future.

While the incidence of PDAC continues to rise, the 5-year overall survival (OS) rate has remained nearly stagnant at 11%; as such, PDAC is expected to become the second most common cause of cancer-related mortality by 2030 (1, 2). Multiagent chemotherapy regimens have led to only modest improvements in patient outcomes (3, 4). Various immunotherapy trials in PDAC have failed to show meaningful results despite strong preclinical backing. Nevertheless, many of these studies offer important insights into the biology of the disease, potential biomarkers, patient selection strategies, and how to best combine existing and novel immunotherapy agents with other treatment modalities to trigger an effective, and hopefully, curative response for more patients with this disease.

In this review, we discuss barriers to effective immunotherapy in PDAC, ranging from cancer cell–intrinsic factors to immunosuppressive features of the tumor microenvironment (TME). We also examine lessons learned from recent immunotherapy clinical trials. Finally, we discuss our hopes for a future where novel immunotherapies will become effective treatments for patients with pancreatic cancer.

The pancreatic TME is a complex ecosystem including cancer cells, immune cells, cancer-associated fibroblasts (CAF), and other stromal elements that create a milieu hidden from the immune system, permissive of tumor growth and metastasis (5). Intratumoral effector T cells in PDAC are relatively rare, particularly in the juxtatumoral stroma located most closely to cancer cells (6), making it one of the “coldest of the cold” tumors. This contrasts with many other “warm” and “hot” solid tumors where infiltration of effector T cells is prominent and immune checkpoint inhibitors (CPI) have made significant inroads by both reinvigorating exhausted effector T cells and recruiting new cells. Aside from the TME, cancer cell–intrinsic factors may also hinder the efficacy of immunotherapy. Namely, oncogenic RAS signaling, ubiquitous in PDAC, drives an inflammatory program that establishes immune privilege in the TME (7), with TME cells in turn secreting specific cytokines to reprogram the metabolic landscape of PDAC cells, upregulating glycolytic genes and glucose uptake (8). This contributes to metabolic derangement in the TME, creating a hostile environment for T cells, which similar to cancer cells are reliant on glycolytic pathways for survival (9). The hypoxic and acidic milieu of PDAC with increased lactic acid production may further limit proper function and proliferation of tumor-infiltrating lymphocytes (10). Tumor mutational burden (TMB), a modest predictor of response to CPI, correlates with neoantigen burden and is often low in PDAC (11, 12). PDAC generally expresses an average of <100 neoepitopes per tumor; and only a small portion of these can modulate immunogenicity (13). To compound this problem, antigen presentation may be impaired by mechanisms such as MHC-I downregulation on cancer cells and a paucity of dendritic cells (DC; refs. 14, 15). In addition, tumor infiltration by immunosuppressive myeloid cells and a dense desmoplastic stroma with its related chemokine axis deflects the immune response and supports tumor progression (6, 16–18). A summary of key mechanisms underlying resistance to immunotherapy in PDAC is illustrated in Fig. 1.

Figure 1.

Mechanisms of immune resistance in pancreatic cancer. PDAC has several mechanisms driving resistance to immunotherapy, including limited infiltration of effector T in the juxtatumoral stroma located close to cancer cells, and increased infiltration by immunosuppressive Tregs, MDSCs, and TAMs of the immunosuppressive phenotype (M2). In addition, cytokines produced by cancer cells, CAFs, and other stromal cells, such as IL10, IL1β, TGFβ, and CXCL12, contribute to the immunosuppressive architecture of the TME, suppressing the cytotoxic activities of immune cells. CCR2, C-C chemokine receptor type 2; CCR5, C-C chemokine receptor type 5; CCR8, C-C chemokine receptor type 8; CD40L, CD40 ligand; CXCR2, CXC chemokine receptor 2; CXCR4, CXC chemokine receptor 4; LAG3, lymphocyte activation gene 3 protein; TIGIT, T-cell immunoglobulin and ITIM domain; TCR, T-cell receptor.

Figure 1.

Mechanisms of immune resistance in pancreatic cancer. PDAC has several mechanisms driving resistance to immunotherapy, including limited infiltration of effector T in the juxtatumoral stroma located close to cancer cells, and increased infiltration by immunosuppressive Tregs, MDSCs, and TAMs of the immunosuppressive phenotype (M2). In addition, cytokines produced by cancer cells, CAFs, and other stromal cells, such as IL10, IL1β, TGFβ, and CXCL12, contribute to the immunosuppressive architecture of the TME, suppressing the cytotoxic activities of immune cells. CCR2, C-C chemokine receptor type 2; CCR5, C-C chemokine receptor type 5; CCR8, C-C chemokine receptor type 8; CD40L, CD40 ligand; CXCR2, CXC chemokine receptor 2; CXCR4, CXC chemokine receptor 4; LAG3, lymphocyte activation gene 3 protein; TIGIT, T-cell immunoglobulin and ITIM domain; TCR, T-cell receptor.

Close modal

Inhibitors of cytotoxic T lymphocyte—associated protein 4 (CTLA-4) and programmed death 1 (PD-1) T-cell checkpoints are the backbone of contemporary immunotherapy. While both pathways suppress T-cell responses, temporal and site-specific differences impart nonredundant functions. Notably, CTLA-4's B7 ligands are primarily restricted to professional antigen-presenting cells (APC) in secondary and tertiary lymphoid organs, while programmed death-ligand 1/2 (PD-L1/2) are inducible and broadly expressed by leukocytes, non-hematopoietic, and cancer cells (19). Further, owing to PD-1 expression on chronically stimulated T cells, PD-L1 antagonists enhance the activity of activated antigen-experienced effector T cells (19). In contrast, CTLA-4 expression is limited to naïve and regulatory T cells (Treg) such that CTLA-4 antagonists selectively enhance naïve T-cell activation (20). These nuances may explain why responses to conventional immunotherapy are limited to small subsets of patients in immunogenic ‘hot’ tumors (e.g., melanoma) but not in ‘cold’ tumors (e.g., PDAC). Notably, recent discoveries on the critical role of fragment crystallizable-Fc gamma receptor (Fc-FcγR) co-engagement for CTLA-4 antibodies have led to the emergence of next-generation Fc-enhanced anti–CTLA-4 antibodies that harness multiple mechanisms of action to overcome barriers associated with ‘cold’ tumors for more effective antitumor immunity (21, 22).

Clinical experience with first generation CTLA-4 and PD-1 inhibitors to date has been underwhelming in PDAC. The anti–CTLA-4 antibody ipilimumab was ineffective as monotherapy with only 1 patient of 27 with advanced PDAC experiencing an objective response (23). In addition, ipilimumab showed no additive benefit when combined with gemcitabine in previously treated patients with PDAC, with an objective response rate (ORR) of 14% and disease stabilization in 33% of patients (24). In a phase II trial of durvalumab, a monoclonal antibody (mAb) against PD-L1, with or without the anti–CTLA-4 antibody tremelimumab, ORR was only 3.1% for patients receiving combination therapy and 0% for those receiving monotherapy (25). Table 1 summarizes the results of a select group of completed immunotherapy trials in PDAC (23, 25–33).

Table 1.

Selected completed clinical trials of immunotherapy in patients with pancreatic cancera.

Trial identifier number and study namePhasePopulationNInvestigational treatmentComparator treatmentResultsReference
NCT02734160 mPDAC, ≤2 lines 32 Galunisertib (TGFβi) + Durvalumab – DCR 25%; mOS 5.72 months (95% CI, 4.0–8.4) 26 
NCT00112580 LA and mPDAC 27 Ipilimumab – ORR 0% per RECIST, 1 delayed PR 23 
NCT02558894 mPDAC, 2nd line 65 Arm A: Durvalumab + Tremelimumab Arm B: Durvalumab Arm A: ORR 3.1%; mOS 3.1 months (95% CI, 2.2–6.1) 25 
      Arm B: ORR 0%; mOS 3.6 months (95% CI, 2.7–6.1)  
NCT02879318 mPDAC, 1st line 180 Arm A: Gem/NP + Durvalumab + Tremelimumab Arm B: Gem/NP Arm A: mOS 9.8 months ClinicalTrials.gova 
Canadian CTG PA.7 trial      Arm B: mOS 8.8 months  
      HR = 0.94 (90% CI, 0.71–1.25; P = 0.72)  
NCT02077881 mPDAC, 1st line 135 Indoximod (IDO i) + Gem/NP – ORR 46.2%; mOS mOS 10.9 months 27 
NCT03250273 mPDAC, ≥2nd line 30 Entinostat (HDACi) + Nivolumab – ORR 16.7%; mOS 3.9 months (95% CI, 1.9–9.4) ClinicalTrials.gova 
NCT01417000 mPDAC, ≥1st line 90 Arm A: Cy/GVAX + CRS-207 Arm B: Cy/GVAX Arm A: mOS 6.1 months 28 
      Arm B: 3.9 months  
      HR = 0.59 (95%CI, 0.36–0.97; P = 0.02)  
NCT02826486 mPDAC, 2nd line 43 Motixafortide (CXCR4 i) + Pembrolizumab + NAPOLI-1 chemo – ORR 21.1%; DCR 63.2%; mOS 6.6 months (95% CI, 4.5–8.7 months) 33 
COMBAT trial        
NCT03214250 PRINCE mPDAC, 1st line 93 Arm A: Gem/NP + Nivolumab Historical 1-y OS of 35% for Gem/NP Arm A: 1-y OS 57%, P = 0.007 29 
      Arm B: 1-y OS 51%, P = 0.029  
      Arm C: 1-y OS 41%, P = 0.236  
        
    Arm B: Gem/NP + Sotigalimab (aCD40 agonist)    
    Arm C: Gem/NP + Sotigalimab + Nivo    
NCT01836432 PILLAR trial BR or LA PDAC, neoadjuvant 303 Arm A: Algenpantucel-L + SOC chemo + RT Arm B: SOC chemo + RT Arm A: mPFS 14.3 months 30 
      Arm B: mPFS 14.9 months  
      HR = 1.02 (95% CI, 0.66–1.58; P = 0.98)  
NCT02923921 SEQUOIA trial mPDAC, 2nd line 567 Arm A: FOLFOX + Pegilodecakin (peg-rIL10) Arm B: FOLFOX Arm A: mOS 5.8 months 31 
      Arm B: mOS 6.3 months  
      HR = 1.05 (95% CI, 0.86–1.27)  
        
NCT02436668 RESOLVE trial mPDAC, 1st line 424 Arm A: Gem/NP + Ibrutinib (BTK i) Arm B: Gem/NP Arm A: mOS 9.7 months 32 
      Arm B: mOS 10.8 months  
      HR = 1.1 (95% CI, 0.9–1.3)  
Trial identifier number and study namePhasePopulationNInvestigational treatmentComparator treatmentResultsReference
NCT02734160 mPDAC, ≤2 lines 32 Galunisertib (TGFβi) + Durvalumab – DCR 25%; mOS 5.72 months (95% CI, 4.0–8.4) 26 
NCT00112580 LA and mPDAC 27 Ipilimumab – ORR 0% per RECIST, 1 delayed PR 23 
NCT02558894 mPDAC, 2nd line 65 Arm A: Durvalumab + Tremelimumab Arm B: Durvalumab Arm A: ORR 3.1%; mOS 3.1 months (95% CI, 2.2–6.1) 25 
      Arm B: ORR 0%; mOS 3.6 months (95% CI, 2.7–6.1)  
NCT02879318 mPDAC, 1st line 180 Arm A: Gem/NP + Durvalumab + Tremelimumab Arm B: Gem/NP Arm A: mOS 9.8 months ClinicalTrials.gova 
Canadian CTG PA.7 trial      Arm B: mOS 8.8 months  
      HR = 0.94 (90% CI, 0.71–1.25; P = 0.72)  
NCT02077881 mPDAC, 1st line 135 Indoximod (IDO i) + Gem/NP – ORR 46.2%; mOS mOS 10.9 months 27 
NCT03250273 mPDAC, ≥2nd line 30 Entinostat (HDACi) + Nivolumab – ORR 16.7%; mOS 3.9 months (95% CI, 1.9–9.4) ClinicalTrials.gova 
NCT01417000 mPDAC, ≥1st line 90 Arm A: Cy/GVAX + CRS-207 Arm B: Cy/GVAX Arm A: mOS 6.1 months 28 
      Arm B: 3.9 months  
      HR = 0.59 (95%CI, 0.36–0.97; P = 0.02)  
NCT02826486 mPDAC, 2nd line 43 Motixafortide (CXCR4 i) + Pembrolizumab + NAPOLI-1 chemo – ORR 21.1%; DCR 63.2%; mOS 6.6 months (95% CI, 4.5–8.7 months) 33 
COMBAT trial        
NCT03214250 PRINCE mPDAC, 1st line 93 Arm A: Gem/NP + Nivolumab Historical 1-y OS of 35% for Gem/NP Arm A: 1-y OS 57%, P = 0.007 29 
      Arm B: 1-y OS 51%, P = 0.029  
      Arm C: 1-y OS 41%, P = 0.236  
        
    Arm B: Gem/NP + Sotigalimab (aCD40 agonist)    
    Arm C: Gem/NP + Sotigalimab + Nivo    
NCT01836432 PILLAR trial BR or LA PDAC, neoadjuvant 303 Arm A: Algenpantucel-L + SOC chemo + RT Arm B: SOC chemo + RT Arm A: mPFS 14.3 months 30 
      Arm B: mPFS 14.9 months  
      HR = 1.02 (95% CI, 0.66–1.58; P = 0.98)  
NCT02923921 SEQUOIA trial mPDAC, 2nd line 567 Arm A: FOLFOX + Pegilodecakin (peg-rIL10) Arm B: FOLFOX Arm A: mOS 5.8 months 31 
      Arm B: mOS 6.3 months  
      HR = 1.05 (95% CI, 0.86–1.27)  
        
NCT02436668 RESOLVE trial mPDAC, 1st line 424 Arm A: Gem/NP + Ibrutinib (BTK i) Arm B: Gem/NP Arm A: mOS 9.7 months 32 
      Arm B: mOS 10.8 months  
      HR = 1.1 (95% CI, 0.9–1.3)  

aClinicalTrials.gov was accessed on April 14, 2022, to obtain updated study results.

Abbreviations: aCD40, anti-CD40 antibody; BR, borderline resectable; BTK i, Bruton's tyrosine kinase inhibitor; CRS-207, live-attenuated mesothelin-expressing Listeria monocytogenes vaccine; CXCR4 i, C-X-C motif chemokine receptor 4 inhibitor; Cy, cyclophosphamide; FOLFOX, regimen of infusional fluorouracil, leucovorin, and oxaliplatin; IDO i, indoleamine 2,3-dioxygenase 1 inhibitor; Gem, gemcitabine; HDAC i, histone deacetylase inhibitor; LA, locally advanced; mPDAC, metastatic pancreatic ductal adenocarcinoma; NAPOLI1, regimen of liposomal irinotecan plus fluorouracil and leucovorin; N, number of patients; NP, nab-paclitaxel; peg-rIL10, pegylated recombinant interleukin-10; RT, radiotherapy; TGFβi, TGFβ inhibitor.

Targeting CAFs and associated stroma

Pancreatic cancer stroma is dominated by a robust desmoplastic and hypo-vascular matrix that creates a mechanical and biochemical boundary around cancer cells that prevents appropriate vascularization, diminishes exposure to chemotherapy, and limits immune cell infiltration into the tumor (34–36). CAFs contribute to this stroma and comprise a diverse population of cells that have been implicated in tumor progression, metastatic spread, and in immunotherapy failure (16, 37, 38). Many of the immunomodulatory effects of CAFs are thought to involve cellular cross-talk through the secretion of cytokines (39–41) which lead to T-cell exclusion from the tumor parenchyma and resistance to CPI (42–44). Conflicting evidence exists that certain stromal elements restrain, rather than promote tumor growth (45, 46), suggesting the existence of functionally and temporally divergent populations of immune cells and CAFs in the TME. Thus far, CAF-directed therapies have been primarily targeted to the highly expressed serine protease fibroblast activation protein-α (FAP), but without clear efficacy signals. For instance, in a phase II trial, the small molecule inhibitor of FAP, talabostat, was combined with gemcitabine (47). Of 21 evaluable patients with advanced PDAC, only 3 achieved objective responses and median progression-free survival (mPFS) was 3.5 months (47). Recent studies have noted the heterogeneity and plasticity of CAFs in PDAC, including identification of myofibroblast (myCAF), antigen-presenting (apCAF), and inflammatory fibroblast (iCAF) subtypes (48, 49). This phenotypic heterogeneity may underlie observations that unselected targeting of CAFs could paradoxically accelerate tumor progression (45, 46).

TGFβ is a cytokine released by pancreatic tumor cells, fibroblasts and other stromal cells contributing to the immunosuppressive architecture of the TME (50). Targeting of TGFβ with the small molecule inhibitor galunisertib combined with gemcitabine as first-line therapy in PDAC only slightly increased median overall survival (mOS) versus gemcitabine monotherapy and did not reach statistical significance [8.9 vs. 7.1 months; HR, 0.79; 95% confidence interval (CI), 0.59–1.09; ref. 51]. Galunisertib was subsequently tested in combination with durvalumab in 32 patients with advanced PDAC and showed limited efficacy, with only one partial response (PR; ref. 26). New strategies, for example using a bifunctional fusion of a mAb against TGFβ with other CPIs are under investigation (52).

All-trans retinoic acid (ATRA) is a metabolite of vitamin A that has pleiotropic effects on immune and nonimmune cell types. Under tumorigenic conditions, the tissue-resident fibroblasts, pancreatic stellate cells, lose their intracellular stores of retinoic acid and convert to critical mediators of desmoplasia (53). Restoring retinoic acid with ATRA limits desmoplasia and reduces cancer growth in a preclinical model (54). In addition, ATRA increases the ratio of CD8-to-Tregs in preclinical models (55). In a phase Ib trial, ATRA combined with gemcitabine and nab-paclitaxel showed a mOS of 11.7 months in the first-line setting for locally advanced and metastatic disease (56).

Targeting the CXCR4/CXCL12 axis

Stromal cell–derived factor-1 (CXCL12), a chemokine produced by CAFs, is often expressed at higher levels in the PDAC TME and creates a network of dense stroma restricting immune cell migration and recognition of cancer cell antigens (16). Preclinically, disrupting the binding of CXCL12 with its receptor, C-X-C chemokine receptor type 4 (CXCR4) improved the effect of CPI in PDAC models (6, 16). A phase II trial with the small molecule CXCR4 antagonist, motixafortide in combination with pembrolizumab, demonstrated enhanced CD8+ effector T-cell infiltration in the tumor, decreased numbers of intratumoral myeloid-derived suppressor cells (MDSC), and decreased circulating Tregs (57). Despite these effects, the combination of motixafortide/pembrolizumab/NAPOLI-1 regimen (liposomal irinotecan, 5-fluorouracil, and leucovorin) led to a modest ORR of 21.1%, disease control rate (DCR) of 63.2% and mOS of 6.6 months in patients with de novo metastatic PDAC who progressed after gemcitabine-based chemotherapy (33). Notably, patients without liver metastasis derived more benefit from this immunotherapy regimen, with an ORR of 37.5% and DCR of 87.5% (33).

Targeting immunosuppressive myeloid cells with CSF-1R inhibitors

PDAC tumors are infiltrated by tumor-associated macrophages (TAM) of the immunosuppressive phenotype (M2) defined by expression of CD206, colony-stimulating factor 1 receptor (CSF-1R), IL10 and decreased expression of MHC class II (58). The CSF-1 pathway is critical for the differentiation and survival of M2 macrophages. Blockade of the CSF-1 pathway has been shown to shift TAMs towards the M1 phenotype and induce distinct TME remodeling (59–61). Cabiralizumab, a humanized IgG4 mAb, binds to CSF-1R and blocks its signaling. In a phase I trial combining cabiralizumab and nivolumab, the ORR was 10% in a cohort of 31 heavily pretreated patients with PDAC (62). However, in a subsequent phase II trial cabiralizumab in combination with nivolumab, with or without standard of care (SOC) chemotherapy did not improve progression-free survival (PFS) compared with SOC chemotherapy alone in the second-line setting (NCT03336216; ref. 63).

IL10 agonism

Preclinical characterization of IL10 has revealed its direct regulation of MHC class II antigens, expansion of T cells, B cells, and mast cells, as well as promotion of T cell–mediated tumor rejection (64, 65). Pegylated IL10 (pegilodecakin) was tested in an early-phase study demonstrated clinical activity when combined with folinic acid, fluorouracil, and oxaliplatin (FOLFOX) in patients with gemcitabine-refractory metastatic PDAC, with an ORR of 15.8%, including two (10.5%) complete responses (CR), and a 1-year survival rate of 43% (66). Despite these promising early results, the addition of pegilodecakin to FOLFOX did not improve efficacy in the phase III SEQUOIA trial (31). Notably, exploratory analyses revealed that pegilodecakin caused immunostimulatory changes such as an increase in total IL18, IFNγ, and granzyme B, as well as a reduction of TGFβ (31). In addition, larger on-treatment increases in IL18 correlated with improved clinical outcomes in pegilodecakin-treated patients. Further investigation into whether baseline levels of IL18 may be predictive of benefit is warranted.

Engaging the myeloid compartment via CD40 agonism

DCs are essential to generate antitumor adaptive immunity through their priming of de novo naïve antigen-specific T cells, but often fail to accumulate in the tumor milieu where they acquire tumor antigen (67, 68). Further, DCs that do accumulate in the tumor may be immature and less effective at antigen presentation and T-cell stimulation (69). DC dysfunction is mediated in part by dysregulated cytokines such as IL10 and TGFβ secreted directly by the tumor or tumor-recruited immune cells (69, 70). Thus, therapeutic approaches to engage DCs may improve adaptive immune responses against the tumor. CD40 ligand, a costimulatory molecule expressed on T cells, binds to its cognate receptor (CD40) on DCs and other APCs leading to lymphocyte activation (71). As demonstrated in preclinical models, CD40 signaling upregulates the expression of B7 costimulatory molecules and promotes antigen presentation by APCs to activate antigen-specific T cells (72–74). Recently, the phase I PRINCE trial with the CD40 agonist mAb sotigalimab (APX005M)/gemcitabine/nab-paclitaxel/±nivolumab, yielded a median OS of 20.1 months in the first-line for patients with locally advanced and metastatic PDAC (29). In a phase II study, gemcitabine/nab-paclitaxel/nivolumab had superior outcomes with a 1-year OS rate of 57.3%, meeting the primary endpoint. Gemcitabine/nab-paclitaxel/sotigalimab had intermediate efficacy with 1-year OS rate of 51% and unexpectedly, chemotherapy/sotigalimab/nivolumab had lower efficacy with a 1-year OS of only 41% (75). Further, a non-fucosylated IgG1 CD40 agonist mAb (SEA-CD40) with gemcitabine/nab-paclitaxel/pembrolizumab in the first-line setting demonstrated an ORR of 44% with a mOS of 15 months (76). A potential pitfall for targeting CD40 in PDAC may be the prevalence of liver metastases, in which a scarcity of DCs may directly contribute to immunotherapy resistance (77).

Another approach to augment T cell–mediated immune surveillance is to employ vaccines targeting tumor-associated antigens that are overexpressed in cancer cells relative to normal tissues or tumor-specific neoantigens derived from mutational events. GVAX is a whole-cell vaccine derived from allogeneic or autologous PDAC cell lines engineered to release GM-CSF at the vaccination site (78). In a phase I trial, previously treated patients with advanced PDAC were randomized to either ipilimumab or ipilimumab plus GVAX. Three patients in the combination arm had prolonged stable disease with a decline in carbohydrate antigen 19–9 (CA19–9; ref. 79). Median OS was 3.6 months in the ipilimumab arm and 5.7 months in the GVAX/ipilimumab arm, with encouraging induction of mesothelin-specific T cells and enhancement of the T-cell repertoire with GVAX (79). However, a phase II trial of GVAX in combination with CRS-207, a live-attenuated listeria vaccine expressing mesothelin, showed no survival advantage over physician's choice of single-agent chemotherapy in the second-line for metastatic PDAC (80). These data underscore the challenges in eliciting cancer-specific T-cell responses in PDAC and suggest that a combination of vaccines with immune modulators beyond first generation anti–CTLA-4 and/or anti–PD-1 antibodies will be necessary to achieve clinical benefit.

Oncolytic viruses can similarly promote tumor-directed innate and adaptive immune responses. Pelareorep, an oncolytic reovirus, was tested in a phase Ib trial with pembrolizumab and chemotherapy (gemcitabine, 5-fluorouracil, or irinotecan) in previously treated patients with PDAC. Some clinical benefit was observed, with 3 of 11 patients experiencing a PR or stable disease (81). Biomarker analysis revealed changes in the peripheral T-cell repertoire, with early and durable clonal expansion correlating with longer OS (81). Additional investigation is warranted to determine if these dynamic changes in peripheral T-cell clonality may serve as a predictive biomarker of response to oncolytic viral therapy. VCN-01, another oncolytic virus, was designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase (82). A phase I trial of gemcitabine/nab-paclitaxel in combination with intratumoral injection of VCN-01 showed that the agent was well tolerated, patients had increasing levels of serum hyaluronidase and injected lesions achieved stabilization or regression (82).

Development of preclinical models to inform clinical trials

The use of appropriate models is critical to determine the dominant drivers of cancer immunity in pancreatic cancer and better inform clinical trials. Studies employing the genetically engineered mouse KPC model (LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre) have enabled the elucidation of several mechanisms of stromal-driven immunosuppression in PDAC (83). Unfortunately, immunotherapy approaches including CXCR4 and hedgehog inhibitors, among others, which demonstrated efficacy in the KPC model have failed in the clinic (33, 84–86). Potential explanations for these discrepancies may include dissimilarities in the immune cell composition of human pancreatic tumors and KPC, such as a higher abundance of effector memory CD8+ and Tregs in some human tumors (6, 87), whereas murine tumors have a more myeloid-predominant microenvironment (87). Hence, employing KPC as the only model for preclinical testing of immunotherapy may be inadequate to predict clinical responses. Other factors including the size and location of the tumor in the model (e.g., subcutaneous, orthotopic, or liver) as well as the magnitude of the response in the model may be important, that is, a modest increase in PFS may be less predictive in the clinic than a curative response in a majority of treated mice.

Patient-derived xenograft (PDX) avatar models have been developed to expand translational research and personalize the treatment of pancreatic cancer (88). A fundamental shortcoming of PDX models for immunotherapy studies is that the tumors are propagated in immunocompromised mice (88, 89). PDAC avatar tumors do not retain donor immune cells, including T cells and macrophages. To enable translational immunotherapy research, avatar tumors can be grown and propagated in mice with a “humanized” immune system (90, 91), such as NOD/SCID gamma mice engrafted with human CD34+ hematopoietic stem cells (91). While these humanized models have been used to evaluate responses to CPI in lung cancer and melanoma with varying success (91, 92), immune cells of humanized avatar PDAC models are not derived from the same patient as the tumor and this may hamper their ability to predict responses in the clinic.

Ex vivo tissue cultures maintain tumor histology, proliferation, and viability for up to 7 days (6, 93). In these cultures, the integrity of the PDAC TME is evidenced by viable stromal myofibroblasts, CD8+ T cells, and CD68+ macrophages (6, 93), and may serve as a close surrogate of the patient's tumor. A recent study using an ex vivo culture evaluated the immune effects of the CXCR4 inhibitor plerixafor and an anti–PD1 antibody, showing enhanced effector function of CD8+ T cells as well as enhanced migration through the stromal microenvironment (6), recapitulating a previous study in KPC models (16). Unfortunately, the clinical impact of this therapeutic strategy was more limited (33), again underscoring the challenges in translating preclinical findings to the clinic. Implementing methods to maintain viability beyond 7 days may be needed because many relevant immune responses take longer than a week to develop. In addition, this model does not account for immune cell migration from the periphery to the tumor.

Combining more than one of these methods may potentially help avoid late-stage clinical trial failures. Further, taking scientific insights derived from early-phase trials and applying them backwards to preclinical models as a means of reverse translation may help better characterize and develop more relevant preclinical models for immunotherapy of PDAC.

Multiple promising therapies impinging on a range of mechanisms are under preclinical and clinical development. These include novel approaches to promote T-cell responses, modify myeloid and stromal compartments, and recruit new immune cells to the PDAC TME. In addition to the CD40 agonistic antibodies discussed earlier, agonism of CD11b has emerged as an approach to reprogram myeloid cells (94). GB1275 is a first-in-class oral allosteric agonist of CD11b shown to blunt both monocyte and granulocyte infiltration in the TME, while simultaneously repolarizing tumor-resident TAMs to an M1 phenotype overcoming resistance to CPI in murine models of PDAC (18). These preclinical data have led to a clinical trial of GB1275 in combination with anti–PD1 or chemotherapy in PDAC and other tumors (NCT04060342; ref. 95). Beyond targeting the myeloid compartment directly, other approaches add to the immune repertoire via allogeneic natural killer (NK) and invariant natural killer T (iNKT) cells to bypass antigen presentation and reprogram the TME. These cellular therapies may overcome the immunosuppressive TME in PDAC with their innate ability to recognize tumor cells by bypassing downregulated antigen-MHC complex presentation on PDAC cells and modulating TAMs (96–98). A phase II multimodal study of an off-the-shelf allogeneic PD-L1–targeted high-affinity NK cell infusion (t-haNK) with an IL15 agonist mAb, radiation, and chemotherapy (NCT04390399), and a phase I trial of iNKT cells in combination with CPI in solid tumors including PDAC are underway (NCT05108623).

Aside from targeting other elements of the immune system, there is renewed interest in recruiting and activating effector T cells to cold tumors such as PDAC with a new generation of CPIs in combination with other modalities. Botensilimab (AGEN1181), an Fc-enhanced CTLA-4 inhibitor designed to strengthen APC/T cell co-engagement thereby promoting optimal T-cell priming, activation and memory formation has yielded curative responses combined with gemcitabine and nab-paclitaxel in the preclinical KPC model (99). In a phase I trial, objective responses with botensilimab have been reported in pancreaticobiliary, microsatellite stable (MSS) colorectal cancer and several other immunotherapy-resistant tumors (100) prompting development of a phase II combination trial with gemcitabine and nab-paclitaxel in PDAC. T-cell immunoglobulin and ITIM domain (TIGIT) has also been implicated in immune escape in PDAC (101), and blockade of the CD155/TIGIT axis in combination with other CPIs, such as CD40, may prove beneficial (101). Targeting stroma along with CPI is also a common theme, including a focus on the TGFβ and adenosine pathways (102, 103). Given prior failures with nonspecific targeting of CAFs, a novel approach entering phase I clinical trials is using antibody–drug conjugates to target FAP-expressing fibroblasts with the FAP protein as a dock for the local delivery of a cytotoxic payload to kill bystander tumor cells (92). Finally, microbial host factors have been shown to affect tumor responses in the KPC preclinical model following fecal microbiota transplantation from PDAC long-term human survivors, a concept currently being tested in clinical trials (NCT04975217; ref. 104). Table 2 provides a select group of ongoing clinical trials testing novel immunotherapy approaches that may hold promise in PDAC.

Table 2.

Ongoing clinical trials with novel immunotherapy approaches for pancreatic cancera.

Trial identifier number and study namePhaseTreatment typeInvestigational treatmentComparator treatmentPopulationSettingStatus
NCT04390763 daNIS-1 trial TME modulation TGFβ antagonist ± Spartalizumab + Gem/NP Gem/NP mPDAC 1st line Recruiting 
NCT03821935 TME modulation GARP-TGFβ1 inhibitor + aPD-1  LA or mPDAC Refractory Recruiting 
NCT04581343 TME modulation aIL1β antagonist + Spartalizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
PanCAN-SR1 trial        
NCT04543071 TME modulation CXCR4i + Cemiplimab + Gem/NP  mPDAC 1st line Recruiting 
Chemo4METPANC trial        
NCT04477343 TME modulation CXCR1/2i + Nivolumab  mPDAC Maintenance after 4 months of first-line chemo Recruiting 
NCT03257761 1b TME modulation Durvalumab + Guadecitabine (epigenetic modifier)  LA or mPDAC ≥2nd line Active, not recruiting 
NCT04060342 1/2 TME modulation CD11b modulator ± Pembrolizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
NCT03727880 TME modulation Pembrolizumab ± FAKi  Resected PDAC Neoadjuvant & Adjuvant Recruiting 
NCT03611556 1/2 TME modulation aCD73 ± Durvalumab ± Chemo Gem/NP or mFOLFOX mPDAC 1st line Active, not recruiting 
NCT03496662 1/2 TME modulation CCR2/CCR5i + Nivolumab + Gem/NP Gem/NP BR or LA-PDAC Neoadjuvant Recruiting 
NCT04827953 1/2 TME modulation Hedgehog inhibitor + aCTLA-4 + Gem/NP  mPDAC 1st line Recruiting 
NUMANTIA trial        
        
NCT04807972 Immune agonists aCD40 agonist + mFOLFIRINOX ± aPD-1  mPDAC 1st line Recruiting 
NCT03329950 1/2 Immune agonists aCD40 agonist ± FLT3 L ± Pembrolizumab ± Gem/NP  mPDAC 1st line Recruiting 
NCT02376699 Immune agonists aCD40 agonist + Pembrolizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
NCT04387071 1/2 Immune agonists aOX40 agonist + TLR9 agonist (both intratumoral)  LA or mPDAC 2nd line Recruiting 
NCT03983954 1/2 Immune agonists Naptumomab (tumor targeting superantigen) + Obinutuzumab (aCD20) + Durvalumab  mPDAC Refractory Recruiting 
NCT03225989 1/2 Immune agonists TMZ-CD40 L and 41BBL oncolytic adenovirus + Gem/NP  LA or mPDAC ≥1 line Recruiting 
NCT03193190 1/2 Multi-agents Atezolizumab, Cobimetinib, Bevacizumab, Tocilizumab (aIL6 antagonist), PEGPH20, BL-8040 (CXCR4i), Selicrelumab (aCD40 agonist), RO6874281 (bispecific IL2v-FAP), AB928 (A2AR antagonist), Tiragolumab (aTIGIT) Gem/NP or mFOLFOX mPDAC Cohort 1: 1st line Active, not recruiting 
Morpheus-Pancreatic Cancer trial      Cohort 2: ≥2nd line  
        
NCT04612530 Multimodality Nivolumab + Irreversible Electroporation ± Intratumoral TLR9  Oligometastatic mPDAC Maintenance after FOLFIRINOX Recruiting 
PANFIRE-3 trial        
NCT04050085 Multimodality Nivolumab + Intratumoral TLR9 agonist + SBRT  mPDAC ≥ 1 line Active, not recruiting 
NCT04390399 Multimodality Arm 1: SBRT + Cy + Gem + NP + Aldoxorubicin + N-803 Gem + NP + SBRT (1st line) or mPDAC Metastatic Recruiting 
      Maintenance (group A)  
      2nd line (group B)  
      ≤3rd line (group C)  
   Arm 2: SBRT + Cy + Gem + NP + Aldoxorubicin + N-803 + PD-L1 t-haNK NAPOLI1 (2nd line)    
NCT03767582 1/2 Multimodality CCR2/CCR5i (BMS-813160) + Nivolumab + SBRT ± GVAX  LA-PDAC Neoadjuvant and adjuvant Recruiting 
NCT04999969 Multimodality Wee1 inhibitor (AZD0171)+ Durvalumab + Gem/NP  mPDAC 1st line Recruiting 
NCT03006302 Multimodality CRS-207 + IDOi (Epacadostat) + Pembrolizumab ± GVAX  mPDAC 2nd line Recruiting 
        
NCT04853017 Vaccine Amphiphile mKRAS peptide vaccine  Resected PDAC Adjuvant for MRD (mKRAS) Recruiting 
AMPLIFY-201 trial        
NCT03948763 Vaccine mRNA KRAS vaccine ± Pembrolizumab  LA or mPDAC Refractory (mKRAS) Recruiting 
V941–001 trial        
NCT02600949 Vaccine Personalized peptide vaccine + Pembrolizumab + Imiquimod  mPDAC ≥ 1 line Recruiting 
NCT03806309 Vaccine TAA vaccine + FOLFIRI FOLFIRI LA or mPDAC Maintenance after FOLFIRINOX Recruiting 
TEDOPAM trial        
NCT03161379 Vaccine GVAX/Cy + Nivolumab + SBRT  BR-PDAC Neoadjuvant Active, not recruiting 
NCT03190265 Vaccine Nivolumab + Ipilimumab + CRS-207 ± GVAX/Cy  mPDAC ≤2nd line Recruiting 
NCT02451982 Vaccine GVAX/Cy ± Nivolumab ± aCD137 agonist; Nivolumab + aIL8 mAb  Resected PDAC Neoadjuvant and adjuvant Recruiting 
NCT05108623 Cell therapy Allogeneic iNKT cell therapy ± Nivolumab or Pembrolizumab  Refractory solid tumors, including PDAC  Not yet recruiting 
Trial identifier number and study namePhaseTreatment typeInvestigational treatmentComparator treatmentPopulationSettingStatus
NCT04390763 daNIS-1 trial TME modulation TGFβ antagonist ± Spartalizumab + Gem/NP Gem/NP mPDAC 1st line Recruiting 
NCT03821935 TME modulation GARP-TGFβ1 inhibitor + aPD-1  LA or mPDAC Refractory Recruiting 
NCT04581343 TME modulation aIL1β antagonist + Spartalizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
PanCAN-SR1 trial        
NCT04543071 TME modulation CXCR4i + Cemiplimab + Gem/NP  mPDAC 1st line Recruiting 
Chemo4METPANC trial        
NCT04477343 TME modulation CXCR1/2i + Nivolumab  mPDAC Maintenance after 4 months of first-line chemo Recruiting 
NCT03257761 1b TME modulation Durvalumab + Guadecitabine (epigenetic modifier)  LA or mPDAC ≥2nd line Active, not recruiting 
NCT04060342 1/2 TME modulation CD11b modulator ± Pembrolizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
NCT03727880 TME modulation Pembrolizumab ± FAKi  Resected PDAC Neoadjuvant & Adjuvant Recruiting 
NCT03611556 1/2 TME modulation aCD73 ± Durvalumab ± Chemo Gem/NP or mFOLFOX mPDAC 1st line Active, not recruiting 
NCT03496662 1/2 TME modulation CCR2/CCR5i + Nivolumab + Gem/NP Gem/NP BR or LA-PDAC Neoadjuvant Recruiting 
NCT04827953 1/2 TME modulation Hedgehog inhibitor + aCTLA-4 + Gem/NP  mPDAC 1st line Recruiting 
NUMANTIA trial        
        
NCT04807972 Immune agonists aCD40 agonist + mFOLFIRINOX ± aPD-1  mPDAC 1st line Recruiting 
NCT03329950 1/2 Immune agonists aCD40 agonist ± FLT3 L ± Pembrolizumab ± Gem/NP  mPDAC 1st line Recruiting 
NCT02376699 Immune agonists aCD40 agonist + Pembrolizumab + Gem/NP  mPDAC 1st line Active, not recruiting 
NCT04387071 1/2 Immune agonists aOX40 agonist + TLR9 agonist (both intratumoral)  LA or mPDAC 2nd line Recruiting 
NCT03983954 1/2 Immune agonists Naptumomab (tumor targeting superantigen) + Obinutuzumab (aCD20) + Durvalumab  mPDAC Refractory Recruiting 
NCT03225989 1/2 Immune agonists TMZ-CD40 L and 41BBL oncolytic adenovirus + Gem/NP  LA or mPDAC ≥1 line Recruiting 
NCT03193190 1/2 Multi-agents Atezolizumab, Cobimetinib, Bevacizumab, Tocilizumab (aIL6 antagonist), PEGPH20, BL-8040 (CXCR4i), Selicrelumab (aCD40 agonist), RO6874281 (bispecific IL2v-FAP), AB928 (A2AR antagonist), Tiragolumab (aTIGIT) Gem/NP or mFOLFOX mPDAC Cohort 1: 1st line Active, not recruiting 
Morpheus-Pancreatic Cancer trial      Cohort 2: ≥2nd line  
        
NCT04612530 Multimodality Nivolumab + Irreversible Electroporation ± Intratumoral TLR9  Oligometastatic mPDAC Maintenance after FOLFIRINOX Recruiting 
PANFIRE-3 trial        
NCT04050085 Multimodality Nivolumab + Intratumoral TLR9 agonist + SBRT  mPDAC ≥ 1 line Active, not recruiting 
NCT04390399 Multimodality Arm 1: SBRT + Cy + Gem + NP + Aldoxorubicin + N-803 Gem + NP + SBRT (1st line) or mPDAC Metastatic Recruiting 
      Maintenance (group A)  
      2nd line (group B)  
      ≤3rd line (group C)  
   Arm 2: SBRT + Cy + Gem + NP + Aldoxorubicin + N-803 + PD-L1 t-haNK NAPOLI1 (2nd line)    
NCT03767582 1/2 Multimodality CCR2/CCR5i (BMS-813160) + Nivolumab + SBRT ± GVAX  LA-PDAC Neoadjuvant and adjuvant Recruiting 
NCT04999969 Multimodality Wee1 inhibitor (AZD0171)+ Durvalumab + Gem/NP  mPDAC 1st line Recruiting 
NCT03006302 Multimodality CRS-207 + IDOi (Epacadostat) + Pembrolizumab ± GVAX  mPDAC 2nd line Recruiting 
        
NCT04853017 Vaccine Amphiphile mKRAS peptide vaccine  Resected PDAC Adjuvant for MRD (mKRAS) Recruiting 
AMPLIFY-201 trial        
NCT03948763 Vaccine mRNA KRAS vaccine ± Pembrolizumab  LA or mPDAC Refractory (mKRAS) Recruiting 
V941–001 trial        
NCT02600949 Vaccine Personalized peptide vaccine + Pembrolizumab + Imiquimod  mPDAC ≥ 1 line Recruiting 
NCT03806309 Vaccine TAA vaccine + FOLFIRI FOLFIRI LA or mPDAC Maintenance after FOLFIRINOX Recruiting 
TEDOPAM trial        
NCT03161379 Vaccine GVAX/Cy + Nivolumab + SBRT  BR-PDAC Neoadjuvant Active, not recruiting 
NCT03190265 Vaccine Nivolumab + Ipilimumab + CRS-207 ± GVAX/Cy  mPDAC ≤2nd line Recruiting 
NCT02451982 Vaccine GVAX/Cy ± Nivolumab ± aCD137 agonist; Nivolumab + aIL8 mAb  Resected PDAC Neoadjuvant and adjuvant Recruiting 
NCT05108623 Cell therapy Allogeneic iNKT cell therapy ± Nivolumab or Pembrolizumab  Refractory solid tumors, including PDAC  Not yet recruiting 

aSelected clinical trials evaluating new immunotherapy strategies for pancreatic cancer. ClinicalTrials.gov accessed on March 25, 2022.

Abbreviations: aPD-1, anti–PD-1 antibody; A2AR, adenosine A2A receptor; aCTLA-4, anti–CTLA-4 antibody; BR, borderline resectable pancreatic cancer; Cy, cyclophosphamide; CCR5, C-C chemokine receptor type 5; CCR8, C-C chemokine receptor type 8; Chemo, chemotherapy; CXCR2, CXC chemokine receptor 2; CRS-207 is a live-attenuated, mesothelin-expressing Listeria monocytogenes vaccine; CXCR4, CXC chemokine receptor 4; FAKi, focal adhesion kinase inhibitor; FAP, fibroblast activation protein-α; FLT3LG, Fms related receptor tyrosine kinase 3 ligand; FOLFIRI, regimen of leucovorin, irinotecan, and fluorouracil; Gem, gemcitabine; IDO1i, indoleamine 2,3-dioxygenase 1; LA, locally advanced; mPDAC, metastatic pancreatic cancer; mFOLFIRINOX, modified regimen of oxaliplatin, leucovorin, irinotecan, and fluorouracil; NAPOLI1, regimen of leucovorin, liposomal irinotecan, and fluorouracil; NP, nab-paclitaxel; PEGPH20, pegvorhyaluronidase alfa; TIGIT, T-cell immunoglobulin and ITIM domain; TLR9, Toll-like receptor 9.

The few biomarkers in clinical practice that may predict response to immunotherapy include PD-L1 expression, TMB, and microsatellite status. Of these, in PDAC, PD-L1 has no established role, and TMB and microsatellite status are of limited utility as only a small proportion of PDAC tumors are microsatellite instability-high (MSI-H), DNA mismatch repair deficiency, or high TMB (105). Response rates to CPI in these patients are modest (106, 107) and inferior to other gastrointestinal MSI-H tumors (106, 108). In PDAC, homologous repair deficiency (HRD) may predict response to platinum-based therapy (109) as well as to PARP inhibitors (110), and HRD is also an emerging biomarker of response to combined checkpoint inhibition including CTLA-4 inhibitors in PDAC (111). While HRD represents a larger segment of the PDAC population than MSI-H, it still represents only a minority of patients with PDAC even if the definition is extended beyond germline mutations. While not yet clinically relevant, efforts are underway to classify PDAC based on genomics and transcriptomics and link these molecular subtypes, including an “immunogenic” subtype to clinical activity (112, 113).

In addition to information gleaned from tumor and blood samples, patient intrinsic factors such as the neutrophil to lymphocyte ratio as represented in the Gustave Roussy Immune Score (GRIm) score (114), along with sites of metastatic disease are also emerging as important biomarkers of response to immunotherapy across solid tumors. Liver metastases are known to be particularly recalcitrant to immunotherapy (115) and because they predominate in PDAC, they likely contribute to primary resistance to CPI as they do in MSS colorectal cancer (116). Aside from selecting patients with PDAC without liver metastases for immunotherapy, another potential approach would be to locally treat liver metastases in conjunction with effective systemic immunotherapy. A study of ipilimumab, nivolumab, and stereotactic body radiation therapy (SBRT) in PDAC may support this approach with a DCR of 29% (5 of 17) and 1 patient experiencing a CR (117). In addition, in the trial of the CXCR4 inhibitor motixafortide with anti–PD-1 and chemotherapy discussed above, patients with PDAC without liver metastases had greater benefit compared with those with liver metastases (ORR 37.5% vs. 21.1%, respectively; ref. 33). Whether sites of metastatic disease are simply prognostic or predictive of benefit for immunotherapy in PDAC can be answered in future trials with more effective immunotherapy combinations. These trials should be thoughtfully coupled with translational research and biomarker development to maximize our ability to identify patients who will benefit from treatment.

“Cure” is a laden word in the context of metastatic cancer, with no universally accepted definition. As oncologists, we are loathe to use the word with patients out of fear that a late relapse will occur, and we will be guilty of having instilled false hope. Ultimately, what matters to most patients is a quality life that is not physically or temporally constrained by the presence of cancer. This goal is reflected in the statistical concept of the “tail” of the survival curve, which represents prolonged survival. James Allison, in his acceptance speech for the Nobel Prize, which he received for his role in the discovery of inhibition of negative immune regulation (CTLA-4), famously described the goal of immunotherapy combinations as “raising the tail” of the curve (118). The question then becomes, what practical surrogate endpoints will most accurately translate into greater long-term survival for patients with PDAC? While mOS has been the gold standard historically, using it as the primary endpoint in early-phase trials would unacceptably delay progress.

Endpoints

For late-phase immuno-oncology clinical trials, novel designs using milestone-based endpoints have been proposed to account for the uncertainty of survival kinetics with immunotherapy (119). These designs are informed by ipilimumab/nivolumab versus nivolumab (120) in melanoma where an improvement in ORR (with durability) gave an early signal of efficacy, leading to improved landmark PFS at 12 to 18 months, and finally an improvement in OS that became more pronounced over time. When the clinical effect of a therapy may be delayed and the cure rate is relatively small, traditional endpoints such as mPFS and mOS may be initially misleading. This is apparent in patients with MSI-H PDAC treated with pembrolizumab, where there was only an 18% ORR and median PFS of 2.1 months, but the durability of responses was substantial (median duration of response 13.4 months, range 8.1 to 16.0+; ref. 106).

The challenge in phase one immuno-oncology trials in a resistant disease like PDAC is even greater: how to appropriately select aggressive endpoints with near term readouts that will de-risk and set an appropriately high bar for combinations worth pursuing in subsequent trials. Drawing on experience in melanoma, we propose that these trials should aim for a substantial improvement in ORR with durability (e.g., 90% ORR in first-line PDAC), and if this is not achieved, then landmark PFS at 12 months should be at least doubled compared with SOC (greater than 30% to 40%; ref. 3, 4). If the bar is met, the phase II trial should incorporate an active control arm. In addition to imaging-based endpoints, we hope that circulating tumor DNA and other blood-based biomarkers will add dynamic range to early read outs, thereby improving trials in the metastatic setting, and allowing for more practical trials of immunotherapy in the neoadjuvant and minimal residual disease setting (e.g., NCT04853017).

Combinations

Pancreatic cancer exhibits multiple immunotherapy resistance mechanisms, and therapies targeting only one mechanism at a time have failed. While new immunotherapy agents with monotherapy activity in this disease would be welcome, we believe it is possible to maximize the benefit of existing agents through rational combinations. Indeed, a framework has been suggested for successful next-generation immunotherapy-based treatment of PDAC featuring combinations that boost immune priming and activation, block immune checkpoints, and optimize the TME, in concert with standard cytotoxic therapy (121). MSS colorectal cancer serves as an example: it is a disease where PD-1 inhibition alone has not shown activity (122) and regorafenib has an ORR of only 1% (123). Despite this, the REGONIVO trial combining regorafenib and nivolumab demonstrated encouraging results (124), igniting interest in PD-1 combinations that led to ongoing registration trials where the individual agents have no single agent activity (NCT04776148, NCT05064059).

Prior to the advent of modern immunotherapy, there were notable successes curing complex metastatic solid tumors and hematologic malignancies with novel combinations intended to overcome resistance to single agents (125–127). Testicular cancer and lymphoma exemplify the rapid progress made with this approach (126, 127). More recently, investigators have focused on “one step at a time” development but given substantial obstacles of time and cost and a sense of desperation for patients with cancer without viable options, members of the oncology research community are calling for a return to bold combination designs (128, 129). While there remain several unanswered questions with respect to optimal immunotherapy combinations for PDAC (e.g., optimal sequencing with chemotherapy to move into earlier lines) experience curing testicular cancer and lymphoma tells us it is not necessary that all these questions be answered to design successful combination trials if basic principles are followed. Combinations should have a strong biological rationale and supporting preclinical evidence, preference should be given to agents with individual activity, safety must be acceptable, the patient population should allow for adequate assessment of efficacy based on the expected time to response with immunotherapy, and endpoints should be selected appropriately as discussed above. To encourage such trials, there must be a path to approval for a highly effective combination of multiple novel agents. Progress necessitates that efficacy in an area of unmet need must take precedence over requirements for dose optimization and assessment of contribution of components. To succeed, there must be cooperation between regulatory authorities, academic investigators, biotech engines of drug discovery, and patient advocacy groups, allowing clinical research the flexibility necessary to achieve rapid and meaningful treatment advances for patients.

B. Bockorny reports grants from NanoView Biosciences, nonfinancial support from Erytech Pharma, and personal fees from Blueprint Medicines outside the submitted work. J.E. Grossman reports personal fees and other support from Agenus outside the submitted work. M. Hidalgo reports personal fees from BMS, InxMEd, Champions Oncology, MinKy, VelaVigo, Oncomatrix, and Kahr and other support from Nelum outside the submitted work; in addition, M. Hidalgo is a member of BMS Board of Directors.

1.
Siegel
RL
,
Miller
KD
,
Fuchs
HE
,
Jemal
A
.
Cancer statistics, 2021
.
CA Cancer J Clin
2021
;
71
:
7
33
.
2.
Rahib
L
,
Smith
BD
,
Aizenberg
R
,
Rosenzweig
AB
,
Fleshman
JM
,
Matrisian
LM
.
Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States
.
Cancer Res
2014
;
74
:
2913
21
.
3.
Conroy
T
,
Desseigne
F
,
Ychou
M
,
Bouche
O
,
Guimbaud
R
,
Becouarn
Y
, et al
.
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med
2011
;
364
:
1817
25
.
4.
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
.
5.
Nevala-Plagemann
C
,
Hidalgo
M
,
Garrido-Laguna
I
.
From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer
.
Nat Rev Clin Oncol
2020
;
17
:
108
23
.
6.
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
.
7.
Ischenko
I
,
D'Amico
S
,
Rao
M
,
Li
J
,
Hayman
MJ
,
Powers
S
, et al
.
KRAS drives immune evasion in a genetic model of pancreatic cancer
.
Nat Commun
2021
;
12
:
1482
.
8.
Dey
P
,
Li
J
,
Zhang
J
,
Chaurasiya
S
,
Strom
A
,
Wang
H
, et al
.
Oncogenic KRAS-driven metabolic reprogramming in pancreatic cancer cells utilizes cytokines from the tumor microenvironment
.
Cancer Discov
2020
;
10
:
608
25
.
9.
Wang
R
,
Dillon
CP
,
Shi
LZ
,
Milasta
S
,
Carter
R
,
Finkelstein
D
, et al
.
The transcription factor Myc controls metabolic reprogramming upon T-lymphocyte activation
.
Immunity
2011
;
35
:
871
82
.
10.
Sugiura
A
,
Rathmell
JC
.
Metabolic barriers to T-cell function in tumors
.
J Immunol
2018
;
200
:
400
7
.
11.
Yarchoan
M
,
Hopkins
A
,
Jaffee
EM
.
Tumor mutational burden and response rate to PD-1 inhibition
.
N Engl J Med
2017
;
377
:
2500
1
.
12.
Szeto
C
,
Gounder
MM
,
Parulkar
R
,
Nguyen
A
,
Rabizadeh
S
,
Reddy
SK
.
High correlation between TMB, expressed TMB, and neoantigen load using tumor: normal whole-exome DNA and matched whole transcriptome RNA sequencing
.
J Clin Oncol
2020
;
38
:
e15238
.
13.
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
.
14.
Yamamoto
K
,
Venida
A
,
Yano
J
,
Biancur
DE
,
Kakiuchi
M
,
Gupta
S
, et al
.
Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I
.
Nature
2020
;
581
:
100
5
.
15.
Hegde
S
,
Krisnawan
VE
,
Herzog
BH
,
Zuo
C
,
Breden
MA
,
Knolhoff
BL
, et al
.
Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer
.
Cancer Cell
2020
;
37
:
289
307
.
16.
Feig
C
,
Jones
JO
,
Kraman
M
,
Wells
RJ
,
Deonarine
A
,
Chan
DS
, et al
.
Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti–PD-L1 immunotherapy in pancreatic cancer
.
Proc Natl Acad Sci USA
2013
;
110
:
20212
7
.
17.
Chao
T
,
Furth
EE
,
Vonderheide
RH
.
CXCR2-dependent accumulation of tumor-associated neutrophils regulates T-cell immunity in pancreatic ductal adenocarcinoma
.
Cancer Immunol Res
2016
;
4
:
968
82
.
18.
Panni
RZ
,
Herndon
JM
,
Zuo
C
,
Hegde
S
,
Hogg
GD
,
Knolhoff
BL
, et al
.
Agonism of CD11b reprograms innate immunity to sensitize pancreatic cancer to immunotherapies
.
Sci Transl Med
2019
;
11
:
eaau9240
.
19.
Keir
ME
,
Butte
MJ
,
Freeman
GJ
,
Sharpe
AH
.
PD-1 and its ligands in tolerance and immunity
.
Annu Rev Immunol
2008
;
26
:
677
704
.
20.
Wei
SC
,
Levine
JH
,
Cogdill
AP
,
Zhao
Y
,
Anang
NAS
,
Andrews
MC
, et al
.
Distinct cellular mechanisms underlie anti–CTLA-4 and anti–PD-1 checkpoint blockade
.
Cell
2017
;
170
:
1120
33
.
21.
Waight
JD
,
Chand
D
,
Dietrich
S
,
Gombos
R
,
Horn
T
,
Gonzalez
AM
, et al
.
Selective FcγR co-engagement on APCs modulates the activity of therapeutic antibodies targeting T-cell antigens
.
Cancer Cell
2018
;
33
:
1033
47
.
22.
Arce Vargas
F
,
Furness
AJS
,
Litchfield
K
,
Joshi
K
,
Rosenthal
R
,
Ghorani
E
, et al
.
Fc effector function contributes to the activity of human anti–CTLA-4 antibodies
.
Cancer Cell
2018
;
33
:
649
63
.
23.
Royal
RE
,
Levy
C
,
Turner
K
,
Mathur
A
,
Hughes
M
,
Kammula
US
, et al
.
Phase II trial of single agent ipilimumab (anti–CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma
.
J Immunother
2010
;
33
:
828
33
.
24.
Kamath
SD
,
Kalyan
A
,
Kircher
S
,
Nimeiri
H
,
Fought
AJ
,
Benson
A
3rd
, et al
.
Ipilimumab and gemcitabine for advanced pancreatic cancer: a phase Ib study
.
Oncologist
2020
;
25
:
e808
e15
.
25.
O'Reilly
EM
,
Oh
DY
,
Dhani
N
,
Renouf
DJ
,
Lee
MA
,
Sun
W
, et al
.
Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase II randomized clinical trial
.
JAMA Oncol
2019
;
5
:
1431
8
.
26.
Melisi
D
,
Oh
DY
,
Hollebecque
A
,
Calvo
E
,
Varghese
A
,
Borazanci
E
, et al
.
Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti–PD-L1 antibody durvalumab in metastatic pancreatic cancer
.
J Immunother Cancer
2021
;
9
:
e002068
.
27.
Bahary
N
,
Wang-Gillam
A
,
Haraldsdottir
S
,
Somer
BG
,
Lee
JS
,
O'Rourke
MA
, et al
.
Phase II 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
:
4015
.
28.
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
.
29.
O'Hara
MH
,
O'Reilly
EM
,
Varadhachary
G
,
Wolff
RA
,
Wainberg
ZA
,
Ko
AH
, et al
.
CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: an open-label, multicenter, phase Ib study
.
Lancet Oncol
2021
;
22
:
118
31
.
30.
Hewitt
DB
,
Nissen
N
,
Hatoum
H
,
Musher
B
,
Seng
J
,
Coveler
AL
, et al
.
A phase III randomized clinical trial of chemotherapy with or without algenpantucel-L (HyperAcute-Pancreas) immunotherapy in subjects with borderline resectable or locally advanced unresectable pancreatic cancer
.
Ann Surg
2022
;
275
:
45
53
.
31.
Hecht
JR
,
Lonardi
S
,
Bendell
J
,
Sim
HW
,
Macarulla
T
,
Lopez
CD
, et al
.
Randomized phase III study of FOLFOX alone or with pegilodecakin as second-line therapy in patients with metastatic pancreatic cancer that progressed after gemcitabine (SEQUOIA)
.
J Clin Oncol
2021
;
39
:
1108
18
.
32.
Tempero
M
,
Oh
DY
,
Tabernero
J
,
Reni
M
,
Van Cutsem
E
,
Hendifar
A
, et al
.
Ibrutinib in combination with nab-paclitaxel and gemcitabine for first-line treatment of patients with metastatic pancreatic adenocarcinoma: phase III RESOLVE study
.
Ann Oncol
2021
;
32
:
600
8
.
33.
Bockorny
B
,
Macarulla
T
,
Semenisty
V
,
Borazanci
E
,
Feliu
J
,
Ponz-Sarvise
M
, et al
.
Motixafortide and pembrolizumab combined to nanoliposomal irinotecan, fluorouracil, and folinic acid in metastatic pancreatic cancer: the COMBAT/KEYNOTE-202 Trial
.
Clin Cancer Res
2021
;
27
:
5020
7
.
34.
Jacobetz
MA
,
Chan
DS
,
Neesse
A
,
Bapiro
TE
,
Cook
N
,
Frese
KK
, et al
.
Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer
.
Gut
2013
;
62
:
112
20
.
35.
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
.
36.
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
.
37.
Erez
N
,
Truitt
M
,
Olson
P
,
A
ST
,
Hanahan
D
.
Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner
.
Cancer Cell
2010
;
17
:
135
47
.
38.
Kraman
M
,
Bambrough
PJ
,
Arnold
JN
,
Roberts
EW
,
Magiera
L
,
Jones
JO
, et al
.
Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha
.
Science
2010
;
330
:
827
30
.
39.
Joyce
JA
,
Fearon
DT
.
T-cell exclusion, immune privilege, and the tumor microenvironment
.
Science
2015
;
348
:
74
80
.
40.
Biasci
D
,
Smoragiewicz
M
,
Connell
CM
,
Wang
Z
,
Gao
Y
,
Thaventhiran
JED
, et al
.
CXCR4 inhibition in human pancreatic and colorectal cancers induces an integrated immune response
.
Proc Natl Acad Sci USA
2020
;
117
:
28960
70
.
41.
Grauel
AL
,
Nguyen
B
,
Ruddy
D
,
Laszewski
T
,
Schwartz
S
,
Chang
J
, et al
.
TGFbeta-blockade uncovers stromal plasticity in tumors by revealing the existence of a subset of interferon-licensed fibroblasts
.
Nat Commun
2020
;
11
:
6315
.
42.
Hartmann
N
,
Giese
NA
,
Giese
T
,
Poschke
I
,
Offringa
R
,
Werner
J
, et al
.
Prevailing role of contact guidance in intrastromal T-cell trapping in human pancreatic cancer
.
Clin Cancer Res
2014
;
20
:
3422
33
.
43.
Ene-Obong
A
,
Clear
AJ
,
Watt
J
,
Wang
J
,
Fatah
R
,
Riches
JC
, et al
.
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
.
44.
von Bernstorff
W
,
Voss
M
,
Freichel
S
,
Schmid
A
,
Vogel
I
,
Johnk
C
, et al
.
Systemic and local immunosuppression in pancreatic cancer patients
.
Clin Cancer Res
2001
;
7
:
925s
32s
.
45.
Ozdemir
BC
,
Pentcheva-Hoang
T
,
Carstens
JL
,
Zheng
X
,
Wu
CC
,
Simpson
TR
, et al
.
Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival
.
Cancer Cell
2014
;
25
:
719
34
.
46.
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
.
47.
Nugent
FW
,
Cunningham
C
,
Barve
MA
,
Fisher
W
,
Patel
H
,
Meiri
E
, et al
.
Phase II study of talabostat/gemcitabine in Stage IV pancreatic cancer
.
J Clin Oncol
2007
;
25
:
4616
.
48.
Elyada
E
,
Bolisetty
M
,
Laise
P
,
Flynn
WF
,
Courtois
ET
,
Burkhart
RA
, et al
.
Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts
.
Cancer Discov
2019
;
9
:
1102
23
.
49.
Ohlund
D
,
Handly-Santana
A
,
Biffi
G
,
Elyada
E
,
Almeida
AS
,
Ponz-Sarvise
M
, et al
.
Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer
.
J Exp Med
2017
;
214
:
579
96
.
50.
Derynck
R
,
Turley
SJ
,
Akhurst
RJ
.
TGFβ biology in cancer progression and immunotherapy
.
Nat Rev Clin Oncol
2021
;
18
:
9
34
.
51.
Melisi
D
,
Garcia-Carbonero
R
,
Macarulla
T
,
Pezet
D
,
Deplanque
G
,
Fuchs
M
, et al
.
Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer
.
Br J Cancer
2018
;
119
:
1208
14
.
52.
Strauss
J
,
Heery
CR
,
Schlom
J
,
Madan
RA
,
Cao
L
,
Kang
Z
, et al
.
Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFβ, in advanced solid tumors
.
Clin Cancer Res
2018
;
24
:
1287
95
.
53.
Schnittert
J
,
Bansal
R
,
Prakash
J
.
Targeting pancreatic stellate cells in cancer
.
Trends Cancer
2019
;
5
:
128
42
.
54.
Froeling
FE
,
Feig
C
,
Chelala
C
,
Dobson
R
,
Mein
CE
,
Tuveson
DA
, et al
.
Retinoic acid–induced pancreatic stellate cell quiescence reduces paracrine Wnt-β-catenin signaling to slow tumor progression
.
Gastroenterology
2011
;
141
:
1486
97
,
97.e1–14
.
55.
Huang
L
,
Chen
Z
,
Williams
L
,
Chen
Y
,
Wang
Y
,
Kawakami
M
, et al
.
Abstract 3279: All-trans-retinoic acid (ATRA) markedly augments antitumor immunity
.
Cancer Res
2019
;
79
:
3279
.
56.
Kocher
HM
,
Basu
B
,
Froeling
FEM
,
Sarker
D
,
Slater
S
,
Carlin
D
, et al
.
Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer
.
Nat Commun
2020
;
11
:
4841
.
57.
Bockorny
B
,
Semenisty
V
,
Macarulla
T
,
Borazanci
E
,
Wolpin
BM
,
Stemmer
SM
, et al
.
BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial
.
Nat Med
2020
;
26
:
878
85
.
58.
Schmiechen
ZC
,
Stromnes
IM
.
Mechanisms governing immunotherapy resistance in pancreatic ductal adenocarcinoma
.
Front Immunol
2020
;
11
:
613815
.
59.
Candido
JB
,
Morton
JP
,
Bailey
P
,
Campbell
AD
,
Karim
SA
,
Jamieson
T
, et al
.
CSF1R(+) macrophages sustain pancreatic tumor growth through T-cell suppression and maintenance of key gene programs that define the squamous subtype
.
Cell Rep
2018
;
23
:
1448
60
.
60.
Saung
MT
,
Muth
S
,
Ding
D
,
Thomas
DL
2nd
,
Blair
AB
,
Tsujikawa
T
, et al
.
Targeting myeloid-inflamed tumor with anti–CSF-1R antibody expands CD137+ effector T cells in the murine model of pancreatic cancer
.
J Immunother Cancer
2018
;
6
:
118
.
61.
Zhu
Y
,
Knolhoff
BL
,
Meyer
MA
,
Nywening
TM
,
West
BL
,
Luo
J
, et al
.
CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models
.
Cancer Res
2014
;
74
:
5057
69
.
62.
32nd Annual Meeting and Pre-Conference Programs of the Society for Immunotherapy of Cancer (SITC 2017): Late-Breaking Abstracts
.
J Immunother Cancer
2017
;
5
:
89
.
63.
Wang-Gillam
A
,
O'Reilly
EM
,
Bendell
JC
,
Wainberg
ZA
,
Borazanci
EH
,
Bahary
N
, et al
.
A randomized phase II study of cabiralizumab (cabira) + nivolumab (nivo) ± chemotherapy (chemo) in advanced pancreatic ductal adenocarcinoma (PDAC)
.
J Clin Oncol
2019
;
37
:
TPS465
TPS
.
64.
Spits
H
,
de Waal Malefyt
R
.
Functional characterization of human IL10
.
Int Arch Allergy Immunol
1992
;
99
:
8
15
.
65.
Moore
KW
,
de Waal Malefyt
R
,
Coffman
RL
,
O'Garra
A
.
Interleukin-10 and the interleukin-10 receptor
.
Annu Rev Immunol
2001
;
19
:
683
765
.
66.
Hecht
JR
,
Naing
A
,
Falchook
GS
,
Patel
MR
,
Infante
JR
,
Aljumaily
R
, et al
.
Overall survival of PEGylated pegilodecakin with 5-FU/LV and oxaliplatin (FOLFOX) in metastatic pancreatic adenocarcinoma (PDAC)
.
J Clin Oncol
2018
;
36
:
4119
.
67.
Broz
ML
,
Binnewies
M
,
Boldajipour
B
,
Nelson
AE
,
Pollack
JL
,
Erle
DJ
, et al
.
Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T-cell immunity
.
Cancer Cell
2014
;
26
:
638
52
.
68.
Meyer
MA
,
Baer
JM
,
Knolhoff
BL
,
Nywening
TM
,
Panni
RZ
,
Su
X
, et al
.
Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance
.
Nat Commun
2018
;
9
:
1250
.
69.
Ruffell
B
,
Chang-Strachan
D
,
Chan
V
,
Rosenbusch
A
,
Ho
CM
,
Pryer
N
, et al
.
Macrophage IL10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL12 expression in intratumoral dendritic cells
.
Cancer Cell
2014
;
26
:
623
37
.
70.
Engelhardt
JJ
,
Boldajipour
B
,
Beemiller
P
,
Pandurangi
P
,
Sorensen
C
,
Werb
Z
, et al
.
Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells
.
Cancer Cell
2012
;
21
:
402
17
.
71.
Wachsmann
MB
,
Pop
LM
,
Vitetta
ES
.
Pancreatic ductal adenocarcinoma: a review of immunologic aspects
.
J Investig Med
2012
;
60
:
643
63
.
72.
Ma
HS
,
Poudel
B
,
Torres
ER
,
Sidhom
JW
,
Robinson
TM
,
Christmas
B
, et al
.
A CD40 agonist and PD-1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T cell–mediated anticancer activity
.
Cancer Immunol Res
2019
;
7
:
428
42
.
73.
Morrison
AH
,
Diamond
MS
,
Hay
CA
,
Byrne
KT
,
Vonderheide
RH
.
Sufficiency of CD40 activation and immune checkpoint blockade for T-cell priming and tumor immunity
.
Proc Natl Acad Sci USA
2020
;
117
:
8022
31
.
74.
Soong
RS
,
Song
L
,
Trieu
J
,
Lee
SY
,
He
L
,
Tsai
YC
, et al
.
Direct T cell activation via CD40 ligand generates high avidity CD8+ T cells capable of breaking immunologic tolerance for the control of tumors
.
PLoS One
2014
;
9
:
e93162
.
75.
O'Hara
MH
,
O'Reilly
EM
,
Wolff
RA
,
Wainberg
ZA
,
Ko
AH
,
Rahma
OE
, et al
.
Gemcitabine (Gem) and nab-paclitaxel (NP) ± nivolumab (nivo) ± CD40 agonistic monoclonal antibody APX005M (sotigalimab), in patients (Pts) with untreated metastatic pancreatic adenocarcinoma (mPDAC): Phase (Ph) II final results
.
J Clin Oncol
2021
;
39
:
4019
.
76.
Bajor
DL
,
Gutierrez
M
,
Vaccaro
GM
,
Masood
A
,
Brown-Glaberman
U
,
Grilley-Olson
JE
, et al
.
Preliminary results of a phase I study of sea-CD40, gemcitabine, nab-paclitaxel, and pembrolizumab in patients with metastatic pancreatic ductal adenocarcinoma (PDAC)
.
J Clin Oncol
2022
;
40
:
559
.
77.
Ho
WW
,
Gomes-Santos
IL
,
Aoki
S
,
Datta
M
,
Kawaguchi
K
,
Talele
NP
, et al
.
Dendritic cell paucity in mismatch repair-proficient colorectal cancer liver metastases limits immune checkpoint blockade efficacy
.
Proc Natl Acad Sci USA
2021
;
118
:
e2105323118
.
78.
Ward
JE
,
McNeel
DG
.
GVAX: an allogeneic, whole-cell, GM-CSF–secreting cellular immunotherapy for the treatment of prostate cancer
.
Expert Opin Biol Ther
2007
;
7
:
1893
902
.
79.
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
.
80.
Le
DT
,
Picozzi
VJ
,
Ko
AH
,
Wainberg
ZA
,
Kindler
H
,
Wang-Gillam
A
, et al
.
Results from a phase IIb, randomized, multicenter study of GVAX pancreas and CRS-207 compared with chemotherapy in adults with previously treated metastatic pancreatic adenocarcinoma (ECLIPSE Study)
.
Clin Cancer Res
2019
;
25
:
5493
502
.
81.
Mahalingam
D
,
Wilkinson
GA
,
Eng
KH
,
Fields
P
,
Raber
P
,
Moseley
JL
, et al
.
Pembrolizumab in combination with the oncolytic virus pelareorep and chemotherapy in patients with advanced pancreatic adenocarcinoma: a phase Ib study
.
Clin Cancer Res
2020
;
26
:
71
81
.
82.
Bazan-Peregrino
M
,
Garcia-Carbonero
R
,
Laquente
B
,
Alvarez
R
,
Mato-Berciano
A
,
Gimenez-Alejandre
M
, et al
.
VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects
.
J Immunother Cancer
2021
;
9
:
e003254
.
83.
Hingorani
SR
,
Wang
L
,
Multani
AS
,
Combs
C
,
Deramaudt
TB
,
Hruban
RH
, et al
.
Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice
.
Cancer Cell
2005
;
7
:
469
83
.
84.
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
.
85.
Catenacci
DV
,
Junttila
MR
,
Karrison
T
,
Bahary
N
,
Horiba
MN
,
Nattam
SR
, et al
.
Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer
.
J Clin Oncol
2015
;
33
:
4284
92
.
86.
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
.
87.
Shibuya
KC
,
Goel
VK
,
Xiong
W
,
Sham
JG
,
Pollack
SM
,
Leahy
AM
, et al
.
Pancreatic ductal adenocarcinoma contains an effector and regulatory immune cell infiltrate that is altered by multimodal neoadjuvant treatment
.
PLoS One
2014
;
9
:
e96565
.
88.
Hidalgo
M
,
Amant
F
,
Biankin
AV
,
Budinska
E
,
Byrne
AT
,
Caldas
C
, et al
.
Patient-derived xenograft models: an emerging platform for translational cancer research
.
Cancer Discov
2014
;
4
:
998
1013
.
89.
Garrido-Laguna
I
,
Uson
M
,
Rajeshkumar
NV
,
Tan
AC
,
de Oliveira
E
,
Karikari
C
, et al
.
Tumor engraftment in nude mice and enrichment in stroma-related gene pathways predict poor survival and resistance to gemcitabine in patients with pancreatic cancer
.
Clin Cancer Res
2011
;
17
:
5793
800
.
90.
Choi
Y
,
Lee
S
,
Kim
K
,
Kim
SH
,
Chung
YJ
,
Lee
C
.
Studying cancer immunotherapy using patient-derived xenografts (PDXs) in humanized mice
.
Exp Mol Med
2018
;
50
:
1
9
.
91.
Gonzalez
L
,
Strbo
N
,
Podack
ER
.
Humanized mice: novel model for studying mechanisms of human immune-based therapies
.
Immunol Res
2013
;
57
:
326
34
.
92.
Fabre
M
,
Ferrer
C
,
Dominguez-Hormaetxe
S
,
Bockorny
B
,
Murias
L
,
Seifert
O
, et al
.
OMTX705, a novel FAP-targeting ADC demonstrates activity in chemotherapy and pembrolizumab-resistant solid tumor models
.
Clin Cancer Res
2020
;
26
:
3420
30
.
93.
Jiang
X
,
Seo
YD
,
Chang
JH
,
Coveler
A
,
Nigjeh
EN
,
Pan
S
, et al
.
Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the immune microenvironment
.
Oncoimmunology
2017
;
6
:
e1333210
.
94.
Schmid
MC
,
Avraamides
CJ
,
Foubert
P
,
Shaked
Y
,
Kang
SW
,
Kerbel
RS
, et al
.
Combined blockade of integrin-α4β1 plus cytokines SDF-1α or IL1β potently inhibits tumor inflammation and growth
.
Cancer Res
2011
;
71
:
6965
75
.
95.
Park
H
,
Bendell
JC
,
Messersmith
WA
,
Rasco
DW
,
Bono
JSD
,
Strickler
JH
, et al
.
Preliminary clinical and biologic results of GB1275, a first-in-class oral CD11b modulator, alone and with pembrolizumab, in advanced solid tumors (KEYNOTE A36)
.
J Clin Oncol
2021
;
39
:
2505
.
96.
Duan
X
,
Deng
L
,
Chen
X
,
Lu
Y
,
Zhang
Q
,
Zhang
K
, et al
.
Clinical significance of the immunostimulatory MHC class I chain-related molecule A and NKG2D receptor on NK cells in pancreatic cancer
.
Med Oncol
2011
;
28
:
466
74
.
97.
Lee
J
,
Kang
TH
,
Yoo
W
,
Choi
H
,
Jo
S
,
Kong
K
, et al
.
An antibody designed to improve adoptive NK-cell therapy inhibits pancreatic cancer progression in a murine model
.
Cancer Immunol Res
2019
;
7
:
219
29
.
98.
Janakiram
NB
,
Mohammed
A
,
Bryant
T
,
Ritchie
R
,
Stratton
N
,
Jackson
L
, et al
.
Loss of natural killer T cells promotes pancreatic cancer in LSL-Kras(G12D/+) mice
.
Immunology
2017
;
152
:
36
51
.
99.
Tanne
AJ
,
Galand
C
,
Abou-Slaybi
A
,
Wilkens
M
,
Marques
M
,
Ng
S
, et al
.
Abstract 1878: Fc-enhanced anti–CTLA-4 antibody, AGEN1181: new mechanistic insights for potent antitumor immunity and combination potential in treatment-resistant solid tumors
.
Cancer Res
2021
;
81
:
1878
.
100.
El-Khoueiry
A
,
Bullock
A
,
Tsimberidou
A
,
Mahadevan
D
,
Wilky
B
,
Twardowski
P
, et al
.
479 AGEN1181, an Fc-enhanced anti–CTLA-4 antibody, alone and in combination with balstilimab (anti–PD-1) in patients with advanced solid tumors: initial phase I results
.
J Immunother Cancer
2021
;
9
:
A509
A
.
101.
Freed-Pastor
WA
,
Lambert
LJ
,
Ely
ZA
,
Pattada
NB
,
Bhutkar
A
,
Eng
G
, et al
.
The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer
.
Cancer Cell
2021
;
39
:
1342
60
.
102.
Grell
P
,
Lin
C-C
,
Milella
M
,
Chee
CE
,
Sivakumar
S
,
Peltola
KJ
, et al
.
Phase II study of the anti-TGFβ monoclonal antibody (mAb) NIS793 with and without the PD-1 inhibitor spartalizumab in combination with nab-paclitaxel/gemcitabine (NG) versus NG alone in patients (pts) with first-line metastatic pancreatic ductal adenocarcinoma (mPDAC)
.
J Clin Oncol
2021
;
39
:
TPS4173
TPS
.
103.
Bendell
JC
,
Manji
GA
,
Pant
S
,
Lai
DW
,
Colabella
J
,
Berry
W
, et al
.
A phase I study to evaluate the safety and tolerability of AB680 combination therapy in participants with gastrointestinal malignancies
.
J Clin Oncol
2020
;
38
:
TPS788
TPS
.
104.
Riquelme
E
,
Zhang
Y
,
Zhang
L
,
Montiel
M
,
Zoltan
M
,
Dong
W
, et al
.
Tumor microbiome diversity and composition influence pancreatic cancer outcomes
.
Cell
2019
;
178
:
795
806
.
105.
Luchini
C
,
Brosens
LAA
,
Wood
LD
,
Chatterjee
D
,
Shin
JI
,
Sciammarella
C
, et al
.
Comprehensive characterization of pancreatic ductal adenocarcinoma with microsatellite instability: histology, molecular pathology and clinical implications
.
Gut
2021
;
70
:
148
56
.
106.
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
.
107.
Lawlor
RT
,
Mattiolo
P
,
Mafficini
A
,
Hong
SM
,
Piredda
ML
,
Taormina
SV
, et al
.
Tumor mutational burden as a potential biomarker for immunotherapy in pancreatic cancer: systematic review and still-open questions
.
Cancers
2021
;
13
:
3119
.
108.
Andre
T
,
Shiu
KK
,
Kim
TW
,
Jensen
BV
,
Jensen
LH
,
Punt
C
, et al
.
Pembrolizumab in microsatellite instability-high advanced colorectal cancer
.
N Engl J Med
2020
;
383
:
2207
18
.
109.
Palacio
S
,
McMurry
HS
,
Ali
R
,
Donenberg
T
,
Silva-Smith
R
,
Wideroff
G
, et al
.
DNA damage repair deficiency as a predictive biomarker for FOLFIRINOX efficacy in metastatic pancreatic cancer
.
J Gastrointest Oncol
2019
;
10
:
1133
9
.
110.
Perkhofer
L
,
Gout
J
,
Roger
E
,
Kude de Almeida
F
,
Baptista Simoes
C
,
Wiesmuller
L
, et al
.
DNA damage repair as a target in pancreatic cancer: state-of-the-art and future perspectives
.
Gut
2021
;
70
:
606
17
.
111.
Terrero
G
,
Datta
J
,
Dennison
J
,
Sussman
DA
,
Lohse
I
,
Merchant
NB
, et al
.
Ipilimumab/Nivolumab therapy in patients with metastatic pancreatic or biliary cancer with homologous recombination deficiency pathogenic germline variants
.
JAMA Oncol
2022
;
8
:
1
3
.
112.
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
.
113.
Collisson
EA
,
Bailey
P
,
Chang
DK
,
Biankin
AV
.
Molecular subtypes of pancreatic cancer
.
Nat Rev Gastroenterol Hepatol
2019
;
16
:
207
20
.
114.
Bigot
F
,
Castanon
E
,
Baldini
C
,
Hollebecque
A
,
Carmona
A
,
Postel-Vinay
S
, et al
.
Prospective validation of a prognostic score for patients in immunotherapy phase I trials: The Gustave Roussy Immune Score (GRIm-Score)
.
Eur J Cancer
2017
;
84
:
212
8
.
115.
Yu
J
,
Green
MD
,
Li
S
,
Sun
Y
,
Journey
SN
,
Choi
JE
, et al
.
Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T-cell elimination
.
Nat Med
2021
;
27
:
152
64
.
116.
Wang
C
,
Sandhu
J
,
Ouyang
C
,
Ye
J
,
Lee
PP
,
Fakih
M
.
Clinical response to immunotherapy targeting programmed cell death receptor 1/programmed cell death ligand 1 in patients with treatment-resistant microsatellite stable colorectal cancer with and without liver metastases
.
JAMA Netw Open
2021
;
4
:
e2118416
.
117.
Parikh
AR
,
Szabolcs
A
,
Allen
JN
,
Clark
JW
,
Wo
JY
,
Raabe
M
, et al
.
Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial
.
Nat Cancer
2021
;
2
:
1124
35
.
118.
Allison
JP
.
Nobel Lecture
.
2022
Sat. 16 Apr 2022
.
Nobel Prize Outreach AB 2022
.
119.
Chen
TT
.
Designing late-stage randomized clinical trials with cancer immunotherapy: Can we make it simpler?
Cancer Immunol Res
2018
;
6
:
250
4
.
120.
Wolchok
JD
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
J-J
,
Rutkowski
P
,
Lao
CD
, et al
.
Long-term outcomes with nivolumab plus ipilimumab or nivolumab alone versus ipilimumab in patients with advanced melanoma
.
J Clin Oncol
2022
;
40
:
127
37
.
121.
Heumann
T
,
Azad
N
.
Next-generation immunotherapy for pancreatic ductal adenocarcinoma: navigating pathways of immune resistance
.
Cancer Metastasis Rev
2021
;
40
:
837
62
.
122.
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
.
123.
Grothey
A
,
Van Cutsem
E
,
Sobrero
A
,
Siena
S
,
Falcone
A
,
Ychou
M
, et al
.
Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicenter, randomized, placebo-controlled, phase III trial
.
Lancet
2013
;
381
:
303
12
.
124.
Fukuoka
S
,
Hara
H
,
Takahashi
N
,
Kojima
T
,
Kawazoe
A
,
Asayama
M
, et al
.
Regorafenib Plus Nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603)
.
J Clin Oncol
2020
;
38
:
2053
61
.
125.
Frei
E
3rd
,
Freireich
EJ
.
Progress and perspectives in the chemotherapy of acute leukemia
.
Adv Chemother
1965
;
2
:
269
98
.
126.
Hunger
SP
,
Mullighan
CG
.
Acute lymphoblastic leukemia in children
.
N Engl J Med
2015
;
373
:
1541
52
.
127.
Einhorn
LH
.
Legends in urology
.
Can J Urol
2011
;
18
:
5764
5
.
128.
Bayat Mokhtari
R
,
Homayouni
TS
,
Baluch
N
,
Morgatskaya
E
,
Kumar
S
,
Das
B
, et al
.
Combination therapy in combating cancer
.
Oncotarget
2017
;
8
:
38022
43
.
129.
Scarlett
UK
,
Chang
DC
,
Murtagh
TJ
,
Flaherty
KT
.
High-throughput testing of novel-novel combination therapies for cancer: an idea whose time has come
.
Cancer Discov
2016
;
6
:
956
62
.