1,888
Views
17
CrossRef citations to date
0
Altmetric
Review

Have lessons from past failures brought us closer to the success of immunotherapy in metastatic pancreatic cancer?

, , , &
Article: e1112942 | Received 16 Sep 2015, Accepted 22 Oct 2015, Published online: 08 Apr 2016

ABSTRACT

Pancreatic cancer is extremely resistant to chemo- and radiation-therapies due to its inherent genetic instability, the local immunosuppressive microenvironment and the remarkable desmoplastic stromal changes which characterize this cancer. Therefore, there is an urgent need for improvement on standard current therapeutic options. Immunotherapies aimed at harnessing endogenous antitumor immunity have shown promise in multiple tumor types. In this review, we give an overview of new immune-related therapeutic strategies currently being tested in clinical trials in pancreatic cancer. We propose that immunotherapeutic strategies in combination with current therapies may offer new hopes in this most deadly disease.

Introduction

Pancreatic cancer is characterized by an extremely poor survival rate, with up to 80% patients dying within the first year of diagnosis. In contrast to the positive outlook reported in other cancers in recent years, survival prospects have remained bleak and in Europe the trend is stubbornly negative in both sexes.Citation1 Indeed, the 5-y mortality rate for pancreatic cancer is over 95%.Citation2 Surgical resection is the only potentially curative treatment. However, overall survival (OS) rate even after surgery remains poor, as over 75% of patients with localized disease, amenable to complete surgical resection, die of local or metastatic recurrence within 5 yCitation3,4 Unfortunately, because pancreatic cancer is usually diagnosed at an advanced stage due to lack of specific symptomatology early in the disease, surgery is suitable for only a minority of patients. At the time of diagnosis, only 15–20% of patients present with operable disease whereas about 40% are found to have locally advanced, unresectable disease and approximately 45% have metastatic disease.Citation5

For patients unable to resort to surgery, these tumors represent particularly difficult therapeutic challenges, as they tend to be resistant to current chemo- and radiation-therapy strategies. This feature is due to the inherent genetic instability of pancreatic cancer cells, the immunosuppressive microenvironment at the tumor site and a remarkable desmoplastic reaction characterizing this cancer and renders it impenetrable to most chemotherapies. Ultimately, it is the development and spread of metastases, which leads to patients' death. Hence, controlling metastatic spread or increasing susceptibility of metastases to treatment may become an increasingly attractive avenue of research to improve survival.

Treatment options—chemotherapy

Gemcitabine monotherapy has been the standard treatment for pancreatic cancer since the late nineties. In a small pivotal clinical trial of 126 patients, treatment with gemcitabine, a nucleoside analog, was compared to bolus administration of 5-fluorouracil. Gemcitabine mediated a significant, albeit modest, effect (5.6 vs. 4.4 mo; p = 0.0025) on OS as well as improvements on quality of life, performance status and pain control.Citation6 The primary efficacy measure was clinical benefit response, which was a composite of measurements of pain (analgesic consumption and pain intensity), Karnofsky performance status and weight. Clinical benefit required a sustained ( ≥4 weeks) improvement in at least one parameter without worsening in any others.

Over the past two decades, gemcitabine has been the backbone for the addition of many compounds. Combination doublets of gemcitabine with other chemotherapeutic agents (capecitabine, irinotecan, oxaliplatin and cisplatin) have shown limited clinical effects over gemcitabine monotherapy.Citation7 A recent meta-analysis of 26 studies (with a total of over 8800 patients), reported a significantly lower objective response rate (ORR) (Relative Risk (RR) 0.72; 95% CI 0.63–0.83; p < 0 .001), and lower 1-y OS (RR 0.90; 95% CI 0.82–0.99; p = 0.04) of monotherapy compared only to doublet treatment with fluoropyrimidine, but at the cost of increased toxicity.Citation8 The addition of angiogenic inhibitors (bevacizumab, axitinib and aflibercept) has also failed to demonstrate any significant OS benefit.Citation9-11 The combination with erlotinib (Tarceva), a tyrosine kinase epidermal growth factor receptor inhibitor, has shown a very small clinical benefit in OS (6.4 vs. 6 mo; p = 0.028) and progression-free survival (PFS) (3.8 vs. 3.5 mo; p = 0.006) but at the expense of significant skin and gastrointestinal (GI) toxicities and considerable cost.Citation12 For these reasons, gemcitabine still is for most patients, especially those with poor performance status, the preferred and only treatment option.

In 2010, a randomized Phase III study, Prodige 4-ACCORD 11, reported on 336 untreated metastatic pancreatic ductal adenocarcinoma (PDAC) patients with good performance status (ECOG score of 0 or 1, normal bilirubin, good bone marrow and renal function) treated with FOLFIRINOX compared to gemcitabine alone.Citation13 Patients receiving the combination treatment had significantly longer OS (11.1 vs. 6.8 mo; Hazard ratio (HR)=0.57; 95% CI 0.45–0.73; p < 0 .001). Moreover, ORR (31.6% vs. 9.4%; p < 0 .001) and PFS were also significantly improved (6.4 vs. 3.3 mo; HR 0.47; 95% CI 0.37–0.59; p < 0 .001). Unfortunately, these improvements were countered by a raised incidence of various grade 3–4 toxicities, including febrile neutropenia (5.4% vs. 0.6%; p = 0.009), thrombocytopenia (9.1 vs. 2.4; p = 0.008), peripheral neuropathy (9% vs. 0%; p = 0.001), vomiting (14.5% vs. 4.7%; p = 0.002), diarrhea (12.7 vs. 1.2; p = 0.0001), thromboembolic events (6.6% vs. 4.1%).

The most recently FDA-approved treatment option for patients with advanced stage pancreatic cancer is Abraxane, albumin-bound paclitaxel (nab-paclitaxel) in combination with gemcitabine. A Phase III study (MPACT) reported in 2013 on the effect of gemcitabine plus nab-paclitaxel versus gemcitabine alone, in 861 untreated metastatic PDAC patients.Citation3 OS was significantly improved (8.5 vs. 6.7 mo; HR=0.72; 95% CI 0.62–0.83; p < 0 .001) as well as one-year survival rate (35% vs. 22.2%), PFS (5.5 vs. 3.7; HR=0.69; 95% CI 0.58–0.82; p < 0 .001) and ORR (23% vs. 7%; p < 0 .001). These significant improvements did not increase treatment-related deaths, which were similar in both groups (4% for each) but grade 3–4 neutropenia (38% vs. 20%), fatigue (17% vs. 7%), neuropathy (17% vs. <1 %) were all higher in the combination group. It is noteworthy to mention that in subgroup analyses of patients with poorer performance status (Karnofsky performance score of 70 and 80) and more bulky disease (liver metastases, >3 metastatic sites), the benefit afforded by this combination was greater.

These clinical developments of the last few years have provided added options for treatment of metastatic pancreatic cancer. However, any survival improvements have come at the expense of toxicity, which are somewhat limiting the general applicability of these therapies due to their effect on patients' performance score and added treatment costs due to toxicities to the health services. For these reasons, there is the urgent need for further therapeutic strategies to improve on patients' survival as well as quality of life. Additionally, the benefits of combining chemotherapy and, or radiotherapy with immune modulators to enhance response in patients has not yet been fully understood. Further investigation may provide much needed insight on effective therapeutic combinations and treatment schedules.

Treatment options—immunotherapy

Since the end of the 19th century, many attempts have been made to harness immunity in the battle against cancer. Following on from the early work of European physicians observing a correlation between severe inflammatory responses and cancer regression, William Coley was the first to systematically utilize this association in cancer therapy, by treating his patients with Coley Toxins, a preparation of killed Streptococcus pneumoniae and Serratia marcescens.Citation14 More recently, clinical responses have been well documented in some malignancies with Bacillus Calmette–Guerin (BCG), a strain of Mycobacterium bovis, currently approved for use in non-invasive transitional cell carcinoma of the bladder.Citation15 While medical oncologists have been skeptical of immunotherapy for some time due to the many negative results in solid malignancies, hematologists have successfully harnessed the power of the immune system to induce complete, long-term remissions in patients with leukemia.Citation16 Beyond the therapeutic failures, part of the immunotherapy skepticism in the oncology community was explained by the confusing diversity of strategies tested. Indeed, cancer immunotherapy strategies have included active (e.g., vaccines) and passive (e.g., monoclonal antibodies) immunotherapies which could be either specific (e.g., adoptive T cell) or non-specific (e.g., cytokines) for the cancer treated, the allogeneic transplantation of immunity (e.g., bone marrow, haplo-identical NKs) being a mix of all these strategies. To date, the use of immunotherapy in pancreatic cancer has been rather disappointing. However, recent advances in our understanding of molecular immunology and the interplay between the immune system and cancer have led to some exciting and promising developments. Here, we will review several different immunotherapy strategies used. Due to space limitations, passive immunotherapies are beyond the scope of this article.

Immune responses in pancreatic cancer

The clinical and pre-clinical data suggesting a major role for immunity in pancreatic cancer are now compelling. Pancreatic cancer patients are able to generate both B and T cells recognizing antigens (Ag) expressed on autologous pancreatic tumor cells.Citation17-20 These include Wilms' tumor gene 1 (WT1) (75% of patients),Citation21 mucin 1 (MUC1) (over 85% of patients),Citation22 human telomerase reverse transcriptase (hTERT) (88% of patients),Citation23 mutated K-RAS (73% of patients),Citation24 and carcinoembryonic antigen (CEA) (over 90% of patients).Citation25 Furthermore, sera from patients contain antibodies to tumor associated Ags, MUC-1 and mesothelin, in particular.Citation26,27 Interestingly, pre-invasive pancreatic lesions are characterized by infiltration of immune suppressor cells and absence of immune effector cells, suggesting that tumor immunity may be defective already from the inception of pancreatic cancer development.Citation28 The notion that defective immunological responses are responsible for cancer development is supported by evidence from animal models, which confirms the existence of immune surveillance mechanisms mediating responses which suppress cancer. For example, mice lacking interferon (IFN) Citation29 and perforin,Citation30 vital components for cytotoxic activity, are prone to develop cancer. Moreover, the adaptive immune system can recognize and eliminate malignant cells; in experimental models, it can limit growth of spontaneous and transplanted tumors.Citation31 Protective Ag-specific T cells can also be detected in human cancers.Citation32 However, their effects can be inhibited by the tumor microenvironment. In pancreatic cancer, tolerance to tumor Ag may occur due to Ag persistence, downregulation of major histocompatibility antigens (MHC) which prevents effective Ag presentation or increased infiltration of cells with immunosuppressive properties such as Ag-specific regulatory T (Treg) cells, tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs) and tumor-associated fibroblasts.Citation33,34 The accumulation of MDSCs and Tregs, as well as the alterations to checkpoint pathways which control immune responses development, have been shown to be closely related to the extent of disease, to correlate with disease stage and to predict survival.Citation35-38 Non-specific ‘innate’ tolerance can also be maintained by innate immune cells through the production of anti-inflammatory and immunosuppressive mediators and downregulation of Ag-presenting cell activity.Citation39

Inhibitors or agonists of checkpoint control

To target solid malignancies effectively, tumor-specific T cells must avoid negative regulatory signals that inhibit their activation or induce tolerance in the form of anergy or exhaustion. Cytotoxic T lymphocyte associated protein-4 (CTLA-4) and programmed cell death protein-1 (PD1) are major negative co-stimulatory molecules expressed on activated T cells.Citation40 Antibodies targeting these suppressive co-stimulatory receptors block inhibitory signals and prolong the life of activated T cells as well as induce T cell proliferation. The discovery of immune checkpoint blockade inhibitors is an exciting advance in the field of immunology that has pushed the clinical landscape to make significant progress in cancer immunotherapy.

Following on the clinical success of treatment with an anti-CTLA-4 inhibitor, ipilimumab, in melanoma,Citation41 this strategy was tested in Phase II clinical trials in advanced pancreatic cancer using ipilimumab (NCT01473940), pidilizumab (anti-PD1 mAb) (NCT01313416) and CP-870,893 (a selective agonist mAb of the CD40 receptor) (NCT00711191). Whereas results with ipilimumab have suggested no direct radiological tumor responses,Citation42 treatment with CP-870,893 in combination with gemcitabine led to activation of the immune system and tumor response in a small cohort of patients with unresectable pancreatic cancer.Citation38 Four patients out of 22 chemo-naive pancreatic cancer patients achieved a partial response, while 6 patients showed a PET response with more than 25% decrease in fluorodeoxyglucose uptake within the primary pancreatic tumor. However, responses observed in metastatic lesions were heterogeneous.Citation43 Several trials are now recruiting to investigate the combination of two checkpoint blockade inhibitors (CTLA-4 and PD1/PDL1 blockade) or combination with small molecule inhibitors to overcome the immunosuppressive tumor microenvironment. An ongoing study (NCT02301130) currently investigates the combination of mogamulizumab (an anti-CCR4 mAb) with either MEDI4736 (anti-B7H1 mAb) or tremelimumab (anti-CTLA-4 mAb) to overcome the immunosuppression in pancreatic cancer. This is a constantly evolving clinical research area aiming to find feasible combinations to restore and increase the activation of adaptive and innate immunity.

To date, it is still not understood why certain solid malignancies demonstrate a better clinical response to checkpoint blockade inhibitors than others. This appears to be particularly true for malignancies of the GI tract where anti-PD-1 mAb has activity in the esophageal and gastric cancers, but no activity in the colon (except those carrying microsatellite instabilities) and pancreatic cancers. More combination treatments need to be clinically investigated in this area to provide patients with suitable alternative options.

T cell therapies

Recently, cancer immunotherapy has focused on the activation of adaptive immunity. MUC-1-specific autologous T cells, isolated from patient peripheral blood mononuclear cells (PBMCs), were expanded by incubation with a MUC-1-presenting cell line prior to administration to pancreatic cancer patients. The mean survival time for unresectable patients in this study was 5 mo.Citation44 In a similar study, PBMC-derived mature DCs from a pancreatic cancer patient were pulsed with MUC-1 peptide. The pulsed DCs were administered in combination with MUC-1-specific T cells to patients with unresectable or recurrent pancreatic cancer. A complete response was observed in one patient with lung metastases and the mean survival time of the whole group was 9.8 mo, suggesting that the addition of pulsed DCs may have beneficial effects.Citation45

Chimeric antigen receptors

An alternative to Ag-specific expansion is the lentiviral transduction of patient T cells with a chimeric antigen receptor (CAR) specific for a tumor Ag. CARs are transmembrane proteins comprising an antibody-derived single-chain variable fragment (scFv) specific for a tumor Ag fused to a hinge region, a spacer, a membrane spanning element and signaling domain.Citation46 Often, the intracellular signaling domain contains the signaling motifs from multiple costimulatory molecules (41BB, OX40, CD28). This allows for both T cell receptor and costimulatory signaling cascades to be initiated, leading to optimal T cell activation. The resulting T cells recognize the tumor Ag in its native form and do not rely on its presentation by MHC class I, which is often downregulated in cancer. Several CARs have been created with specificity for mesothelin, CEA, MUC-1 and Her-2/neu and Phase I/II trials (NCT01583686, NCT02465983, NCT02349724) are ongoing.Citation47-49 Although this process results in a large population of Ag-specific T cells, adoptive immunotherapy is currently an expensive and time-consuming process in comparison with tumor Ag-based vaccine, systemic immune stimulation or monoclonal antibody therapies.

Vaccination therapies

Although Ag-specific immune responses can be detected in cancer patients undergoing tumor cell vaccination therapies, this approach has not delivered great successes in pancreatic cancer with the notable exception of post-surgical vaccination therapy which has shown a beneficial impact in pancreatic cancer patients with absent or minimal residual disease.Citation50 Many studies using whole-cell, DNA as well as peptide vaccines have been performed or are ongoing, but to date, there is no vaccine therapy showing benefit in metastatic pancreatic cancer. Most recently, a large Phase III trial (TELOVAC) comparing gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer failed to report any clinical benefit.Citation51 Although the TeloVac trial did not demonstrate any significant difference in OS and PFS between treatment arms, it demonstrated that the vaccine could prevent patient deterioration. Interestingly, further analysis of potential immunological biomarkers did demonstrate that baseline eotaxin levels predicted median OS in the concurrent gemcitabine and Capecitabine with GV1001 group. Whereas high eotaxin levels at baseline correlated with a longer OS in this study, sequential chemo-immunotherapy did not show any correlation between eotaxin levels and OS. A better understanding of the right immunological response profile would be advantageous for future clinical trial so recruitment of potential responders can be better guided.

For this reason, we will not expand further on vaccination therapies, aside from mentioning the case of a 77-y-old patient who was treated with survivin-based peptide vaccination and had a partial response in liver metastases at 6 mo and complete response at 8 mo.Citation52 However, the patient developed recurrent disease after being weaned off the vaccine therapy.

Utilizing bacteria for cancer therapy

The idea of harnessing immunity by inducing repeatedly infectious stimuli was born over 100 y ago. This concept was recently revisited with the suggestion that repeat exposures to microbes, which may induce non-specific acute inflammation and febrile episodes activate immune memory of antigenic changes important for cancer immunosurveillance.Citation53 For example, infections and acute inflammations generate abnormal Ags that activated DC carry to the draining lymph nodes, where they stimulate adaptive immunity and immune memory. This immune memory can be reactivated by tumor Ags and depending on the strength of the memory and of the reactivation, cancer cells may be either eliminated or kept in equilibrium. Two forms of bacterial formulations have been used in the clinic and need to be mentioned here.

Listeria monocytogenes

To date, this bacterium has been used either as a vehicle to deliver a specific epitope (mesothelin) via expression in live-attenuated Listeria monocytogenes.Citation54 or as a radioactive-labeled formulation.Citation55 While the latter has only shown efficacy in pre-clinical models, the live-attenuated L. monocytogenes-expressing mesothelin (CRS-207) in combination with low-dose cyclophosphamide and GVAX pancreas (granulocyte-macrophage colony-stimulating factor-secreting allogeneic pancreatic tumor cells) was compared to cyclophosphamide plus GVAX only, in 90 patients with metastatic pancreatic cancer. In the per-protocol analysis of patients who received at least three doses (two doses of cyclophsphamide/GVAX plus one of CRS-207 or three of cyclophsphamide/GVAX), OS was 9.7 vs. 4.6 mo (p = 0.02). Enhanced mesothelin-specific CD8+ T cell responses were associated with longer OS in both groups.Citation54 This treatment was given ‘breakthrough therapy’ designation by the FDA in 2014. Two clinical trials are planned, one investigating the addition of anti-PD-1 antibodies to the combination (NCT02243371), the other the addition of GVAX plus anti-CTLA-4 antibody in patients receiving FOLFIRINOX (NCT01896869).

In addition to the immune-related therapeutic strategies outlines in this review, we would like to mention the randomized, controlled STELLAR trial (Safety and Therapeutic Efficacy of Live-attenuated Listeria/GVAX with anti-PD-1 Regimen) which has enrolled approximately 88 patients with metastatic pancreatic cancer who have been treated previously with one line of chemotherapy. CRS-207/GVAX Pancreas vaccine and nivolumab treatment is compared to CRS-207/GVAX Pancreas vaccine alone in this Phase II trial for impact on OS. Secondary endpoints include evaluation of clinical and immune response and safety (NCT02243371).

Lastly, even though not immune-related the clinical development of evofosfamide both as a monotherapy and in combination with chemotherapy treatments needs mention. Evofosfamide is currently under evaluation in a Phase III trial (MAESTRO) in combination with gemcitabine versus gemcitabine and placebo in patients with locally advanced unresectable or metastatic pancreatic cancer. This Phase III trial is being conducted under Special Protocol Assessment (SPA) agreements with the FDA. Based on current projections, the number of protocol-specified events may be reached in the second half of 2015, with the results of the primary efficacy analyses available shortly thereafter. The FDA and the European Commission have granted evofosfamide Orphan Drug Designation for the treatment of pancreatic cancer.

Mycobacterium obuense

As above mentioned, mycobacteria have demonstrated antitumor activity in both pre-clinical and clinical settings and intra-vesical Mycobacterium bovis in the form of BCG is standard of care for non-muscle-invasive Bladder Cancers. Mycobacterium obuense another member of the Mycobacterium genus, is a non-pathogenic saprophytic mycobacteria. IMM-101 is derived from heat-killed Mycobacterium obuense (NCTC 13365), which can induce both innate and adaptive immune responses to boost antitumor immunity. Unlike L. monocytogenes, IMM-101 is not used as a vehicle to deliver either radioactive or tumor-specific Ags. Rather, IMM-101 contains a wide variety of pathogen-associated molecular patterns (PAMPs), including proteins, lipoproteins and carbohydrate Ags which may ensure activation of a wide pool of memory CD8+ cells, which while re-circulating may then amplify the immune response and target metastases and tumor Ags.

In preclinical models, treatment with IMM-101 appeared to increase the secretion of IFNγ and cytotoxic mediators such as perforin and granzyme B at the tumor site, the draining lymph nodes and in the spleen in tumor-bearing mice.Citation56 Most importantly, these modulatory effects are induced when the preparation is administered subcutaneous away from the tumor site. In a recent randomized, open-label, proof-of-concept, Phase II trial in advanced pancreatic cancer (IMAGE 1), the combination of IMM-101 with gemcitabine was tested (NCT01303172).Citation56 Patients with advanced pancreatic cancer and a WHO score of 0–2 were assigned randomly in a 2:1 ratio to receive IMM-101 plus gemcitabine or gemcitabine alone. A total of 110 patients were randomized, 75 to receive IMM-101 plus gemcitabine and 35 gemcitabine alone. In the pre-defined sub-group of patients with metastatic disease (n = 92), median OS was increased significantly by 59% to 7 mo in the IMM-101 plus gemcitabine group (n = 64) compared to 4.4 mo in the gemcitabine alone group (n = 28) (HR 0.54; 95% CI 0.33–0.87; p = 0.01). A highly significant 91% increase in median PFS from 2.3 mo in the gemcitabine group to 4.4 mo in the IMM-101 plus gemcitabine group was observed (HR 0.46; 95% CI 0.28–0.75; p = 0.001). Patients with locally advanced disease at the time of enrolment were eligible for the study and 18 such patients were included, 11 randomized to IMM-101 plus gemcitabine and 7 to gemcitabine alone. This sub-group was too small to draw firm conclusions but there was no evidence for a beneficial effect of IMM-101, which is consistent with preliminary findings from preclinical studies that indicated a more profound effect of IMM-101 on metastases than on the primary tumor.Citation56

The current challenge of immunotherapy in pancreatic cancer: finding synergistic combination therapies

Treatment of advanced pancreatic cancer has concentrated on single agent therapies or combination of compounds within the same class (e.g., cytotoxics). Perhaps, a polyvalent approach would be more appropriate for such a heterogeneous disease.Citation57 Chemotherapies exert various effects on the immune system that could be exploited to enhance the efficacy of immunotherapies. It had long been assumed that immune-stimulatory compounds could not be used in combination with immunosuppressive chemotherapies, but recent evidence has challenged this dogma. Chemotherapies could be used to condition the immune system and the tumor milieu to create an environment where immunotherapies have a better chance of success.

Three ways are proposed by which chemotherapy could promote immune responses and act in synergy with immunotherapiesCitation58: (a) deplete immune suppressive cells; (b) increase tumor immunogenicity by releasing tumor Ags due to cytotoxic activity; (c) direct activation of T cells. Any one of these effects could enhance the tumor-specific immune response elicited by immunotherapeutic agents, and some chemotherapies may even work through multiple mechanisms. A recent publication by García-Martínez and colleagues,Citation59 demonstrated that in a group of 121 neo-adjuvantly treated breast cancer patients, characterization of the immune cell subpopulation profiles by immunohistochemistry-based computerized analysis, identified groups of patients characterized by high response (in the pre-treatment setting) and poor prognosis (in the post-treatment setting). Similarly, immunologic factors were highly significant predictors of therapy response in the GeparSixto trial in breast cancer patients treated with carboplatin.Citation60

Cyclophosphamide has been described to affect subsets of CD4+ T cells.Citation61 In 2005, it was revealed that this drug efficiently depletes CD4+CD25+Treg cells at low doses Citation62 while leaving CD4+CD25 and CD8+ T cells unaffected. Gemcitabine has been proposed to deplete MDSCs. Both drugs have been reported to induce only a transient depletion of the respective cell populations.Citation63 Interestingly, since MHC class II positive MDSCs can promote Treg activation, gemcitabine could potentially reduce multiple suppressor cell types. Vincent et al. Citation63 examined gemcitabine, cyclophosphamide, 5-fluorouracil and paclitaxel among others in pre-clinical models of cancer. 5-fluorouracil induced apoptosis of GR+CD11b+ MDSC and was more potent than gemcitabine. 5-fluorouracil and gemcitabine have also been reported to increase immunological visibility of tumors by increasing expression of tumor Ags.Citation64 Recently, low-dose paclitaxel has been shown to have stimulatory effects on the immune system.Citation65 In murine models, paclitaxel is a ligand for TLR4 on DCs, mediating a direct effect on the immune system.Citation66 In addition, paclitaxel has also been shown to enhance activation of human DCs independent of TLR4 receptor engagement.Citation67 These findings supporting synergy between effects mediated by chemotherapy and the immune system, raise the question as to whether the success of FOLFIRINOX is potentially related to the combination of chemotherapy with additional, although not intentionally given for this purpose, immune modulation via G-CSF, which is administered to prevent the neutropenia caused by FOLFIRINOX.Citation13 Similarly, Abraxane might promote some of its efficacy through the immune modulatory effects of paclitaxel. These observations may lead the way for further investigating the right combination of immunotherapies and chemotherapy. The good results presented at ASCO 2015 of anti-PD-1 and anti-PD-L1 combinations with chemotherapy in non-small cell lung cancer (NSCLC) support this idea: such combinations had an ORR above 60%, that is higher than the sum of the ORR obtained with either therapy separately (ORR of around 30% for chemotherapy alone, and 20% for anti-PD-1/PD-L1 mAb).Citation68,69

In situ immunization: back to the future

One striking observation that can be made when looking at the clinical development of immunotherapies is the fact that although they rely on disruptive mechanisms of action, people keep administering them like conventional therapies (e.g., anti-CTLA-4 or anti-PD-1 administered intra-venously (IV) every 3 weeks). Therefore, it is not surprising that systemic activation of the immune system (e.g., sub-cutaneous IL-2 or ipilimumab+nivolumab) generate a very high level of drug related grade 3–4 off-target toxicities. Intra-tumoral injections of immunostimulatory products could be a good way to use a tumor as its own vaccine and to locally prime an antitumor immune response which could subsequently act on distant, non-treated tumor sites.Citation70 This in situ immunization (also called in situ vaccination) strategy is likely what William Coley was doing at the end of 19th century, and long-term tumor responses were reported in multiple, injectable, cancer types.Citation71,72 Besides, bladder cancer use, intra-tumoral BCG has also demonstrated significant antitumor activity in melanoma and squamous cell carcinoma of the head and neck.Citation73-75 A small clinical report of intra-peritoneal BCG also reported activity in pancreatic cancers.Citation76 Interestingly, the study of the effects of mycobacteria in murine tumor models led to the identification of Toll-like receptor 9 (TLR-9) agonists (CpG-rich DNA motifs).Citation77,78 Recently, the combination of intra-tumoral CpG combined with low dose irradiation (2 × 2 Gy) has demonstrated antitumor activity in patients with B cell and T cell lymphoma, including activity at distant, non-treated, tumor sites.Citation79,80 Also, clinical reports have shown that radiotherapy could overcome anti-CTLA-4 resistance in patients with metastatic melanoma, inducing tumor responses in both irradiated and non-irradiated lesions, through the so-called ‘abscopal effect.’Citation81,82 More recently, the combination of local radiotherapy with systemic (s.c.) GM-CSF has been shown to generate abscopal activity in NSCLC, thymic and breast cancers.Citation83 Radiotherapy could indeed have a more important role to play in the immunotherapy of metastatic pancreatic cancer. Currently, its role (low dose per fraction, 6 weeks treatment) is for local consolidation following chemotherapy for borderline resectable or locally advanced tumors, with the sole intention of causing cell death by oxidative effects and DNA double strand breaks. Ionizing radiations have the ability to convert the irradiated tumor into an ‘immunogenic hub’—acting in effect like an autologous tumor ‘vaccine.’Citation84 At higher doses per fraction, the radiobiology changes due to alternative modes of action.Citation84,85 After radiation exposure, tumor cell death includes apoptosis and necrosis as well as autophagy and mitotic catastrophe. Importantly, radiation has been shown to induce an immunogenic cell death, characterized by three molecular signals that promote uptake of dying cells by DCs, cross-presentation of the tumor-derived Ags to T cells and activation of antitumor T cells, exposure of calreticulin on the tumor cell surface, release of high-mobility group protein B1 (HMGB1), and release of ATP.Citation86 Furthermore among genes that are upregulated post-radiation are those controlling expression of growth factors, cytokines, chemokines, and cell surface receptors that modulate the interaction of the tumor with the immune system.Citation87,88 Thus, radiation may have important systemic effects beyond its local actions.

In addition to excellent local control of disease, high-dose per fraction radiotherapy—stereotactic body radiotherapy (SBRT)—also appears to impact disease outside the irradiated volume. This is likely to be an example of the abscopal effect, resulting from the stimulation of T cell immunity by tumor Ags released by SBRT, leading to the eradication of occult regional micro-metastases. Significant induction of low-density lipoprotein (LDL)-enriched ceramide, secretory sphingomyelinase (S-SMase), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and TNF-α in serum from patients treated with SBRT, suggests these bystander effects may have a role in overall tumor response. In view of these encouraging results, the combination of SBRT and immunotherapy in humans is currently being investigated in several studies and should include pancreatic cancer. However, the identification of the right irradiation dose and regimen for optimal immune activation remains unclear and pre-clinical models have brought contradictory results so far.Citation89

Only 20–30% of patients generate objective responses in many cancer types with anti-PD-1/PD-L1 therapy and no activity has been reported so far in pancreatic cancers. Therefore, the current challenge in cancer immunotherapy is to overcome primary resistance to immune checkpoint blockade therapy. One way could be to increase the intra-tumoral concentration of these immunostimulatory monoclonal antibodies. This could be a good way to increase T cell activation in situ while preventing systemic exposure and off-target toxicity. Interestingly, a recent report at ASCO 2015 has shown strong activity of in situ ipilimumab with IL-2 with abscopal effect seen in 75% of patients with metastatic melanoma.Citation90 It becomes clear now that the in vivo activity of immune checkpoint targeted monoclonal antibodies rely on the presence of FcγR positive cells within the tumor micro-environment (which are mostly myeloid cells, notably macrophages) (see Citation91-92 for review). A good way to switch myeloid cells from a tolerogenic phenotype to an activated Ag-presenting cell phenotype (MHC class I & II high, upregulation of CD80/86) is to stimulate them with PAMPs. Therefore, it would make sense to combine intra-tumoral injections of PAMPs with immune checkpoint targeted antibodies. Indeed, several pre-clinical results have demonstrated the ability of either TLR agonists or oncolytic virus (providers of PAMPs) to overcome immune checkpoint blockade resistance.Citation93,94 This strategy is currently tested in several ongoing clinical trials () and should be specifically developed in patients with pancreatic cancers where the stroma modification seems critical for efficient immunotherapy.

Closing remarks

Therapeutic modalities to treat pancreatic cancer are ever expanding and include surgery, radiotherapy, chemotherapy and now immunotherapy. To obtain clinically effective and meaningful antitumor responses, the successful execution of several interventions will be required. Preclinical studies suggest that immunotherapy combinations targeting distinct steps of antitumor immunity might be synergistic, resulting in stronger and more sustained responses that accomplish durable tumor destruction. Targeting all parts of immune activation, depletion of immunosuppressor cells, enhancing Ag release and presentation and activation of adaptive immunity is crucial to efficient cancer immunotherapy. Bacterial formulations like IMM-101, which do not follow a ‘classic’ approach, offer the benefits of a multitude of immune modulation pathways. This diversity of responses may carry the key for tumor control and overcoming resistance to treatments. Indeed, this approach demonstrates the importance of combining immunotherapy with chemotherapy in the metastatic pancreatic cancer setting, where smaller metastatic lesions lacking the dense desmoplastic stroma of the primary tumor may be more amenable to treatment. Controlling metastatic disease will be the key to achieve better survival outcomes for patients with pancreatic cancer.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References

  • Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2014. Ann Oncol. 2014; 25:1650-6; PMID:24759568
  • Seufferlein T, Bachet JB, Van Cutsem E, Rougier P. Pancreatic adenocarcinoma: ESMO-ESDO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2012; 23 Suppl 7:vii33-40
  • Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Eng J Med 2013; 369:1691-703; http://dx.doi.org/10.1056/NEJMoa1304369
  • Hidalgo M. Pancreatic cancer. N Eng J Med 2010; 362:1605-17
  • Sener SF, Fremgen A, Menck HR, Winchester DP. Pancreatic cancer: a report of treatment and survival trends for 100,313 patients diagnosed from 1985-1995, using the National Cancer Database. J Am College Surgeons 1999; 189:1-7; http://dx.doi.org/10.1016/S1072-7515(99)00075-7
  • Burris HA, 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 1997; 15:2403-13
  • Heinemann V, Haas M, Boeck S. Systemic treatment of advanced pancreatic cancer. Cancer Treatment Rev 2012; 38:843-53; http://dx.doi.org/10.1016/j.ctrv.2011.12.004
  • Sun C, Ansari D, Andersson R, Wu DQ. Does gemcitabine-based combination therapy improve the prognosis of unresectable pancreatic cancer?. World J Gastroenterol 2012; 18:4944-58; http://dx.doi.org/10.3748/wjg.v18.i35.4944
  • Van Cutsem E, Vervenne WL, Bennouna J, Humblet Y, Gill S, Van Laethem JL, Verslype C, Scheithauer W, Shang A, Cosaert J et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol 2009; 27:2231-7; http://dx.doi.org/10.1200/JCO.2008.20.0238
  • Kindler HL, Niedzwiecki D, Hollis D, Sutherland S, Schrag D, Hurwitz H, Innocenti F, Mulcahy MF, O'Reilly E, Wozniak TF et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol 2010; 28:3617-22; http://dx.doi.org/10.1200/JCO.2010.28.1386
  • Kindler HL, Wroblewski K, Wallace JA, Hall MJ, Locker G, Nattam S, Agamah E, Stadler WM, Vokes EE. Gemcitabine plus sorafenib in patients with advanced pancreatic cancer: a phase II trial of the University of Chicago Phase II Consortium. Invest New Drugs 2012; 30:382-6; PMID:20803052; http://dx.doi.org/10.1007/s10637-010-9526-z
  • Boeck S, Hausmann A, Reibke R, Schulz C, Heinemann V. Severe lung and skin toxicity during treatment with gemcitabine and erlotinib for metastatic pancreatic cancer. Anti-Cancer Drugs 2007; 18:1109-11; PMID:17704662
  • Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C et al. FOLFIRINOX vs. gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011; 364:1817-25; PMID:21561347; http://dx.doi.org/10.1056/NEJMoa1011923
  • Coley WB II. Hawkins on Tubercular Peritonitis. Annals of surgery 1893; 17:462-4
  • Herr HW, Schwalb DM, Zhang ZF, Sogani PC, Fair WR, Whitmore WF Jr, Oettgen HF. Intravesical bacillus Calmette-Guerin therapy prevents tumor progression and death from superficial bladder cancer: ten-year follow-up of a prospective randomized trial. J Clin Oncol 1995; 13:1404-8
  • Collins RH, Jr, Sackler M, Pitcher CJ, Waldrop SL, Klintmalm GB, Jenkins R, Picker LJ. Immune reconstitution with donor-derived memory/effector T cells after orthotopic liver transplantation. Exp Hematol 1997; 25:147-59; PMID:9015215
  • Kubuschok B, Neumann F, Breit R, Sester M, Schormann C, Wagner C, Sester U, Hartmann F, Wagner M, Remberger K et al. Naturally occurring T-cell response against mutated p21 ras oncoprotein in pancreatic cancer. Clin Cancer Res. 2006; 12:1365-72; http://dx.doi.org/10.1158/1078-0432.CCR-05-1672
  • Wenandy L, Sorensen RB, Svane IM, Thor Straten P, Andersen MH. RhoC a new target for therapeutic vaccination against metastatic cancer. Cancer Immunol Immunother 2008; 57:1871-8; PMID:18415097; http://dx.doi.org/10.1007/s00262-008-0517-2
  • Wenandy L, Sorensen RB, Sengelov L, Svane IM, Thor Straten P, Andersen MH. The immunogenicity of the hTERT540-548 peptide in cancer. Clin Cancer Res 2008; 14:4-7; http://dx.doi.org/10.1158/1078-0432.CCR-07-4590
  • Dodson LF, Hawkins WG, Goedegebuure P. Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 2011; 3:517-37; PMID:21463193; http://dx.doi.org/10.2217/imt.11.10
  • Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, Abeno S, Ikeba A, Takashima S, Tsujie M et al. Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci 2004; 95:583-7; PMID:15245594; http://dx.doi.org/10.1111/j.1349-7006.2004.tb02490.x
  • Ueda M, Miura Y, Kunihiro O, Ishikawa T, Ichikawa Y, Endo I, Sekido H, Togo S, Shimada H. MUC1 overexpression is the most reliable marker of invasive carcinoma in intraductal papillary-mucinous tumor (IPMT). Hepato-gastroenterology 2005; 52:398-403; PMID:15816444
  • Seki K, Suda T, Aoyagi Y, Sugawara S, Natsui M, Motoyama H, Shirai Y, Sekine T, Kawai H, Mita Y et al. Diagnosis of pancreatic adenocarcinoma by detection of human telomerase reverse transcriptase messenger RNA in pancreatic juice with sample qualification. Clin Cancer Res 2001; 7:1976-81
  • Gjertsen MK, Bakka A, Breivik J, Saeterdal I, Solheim BG, Søreide O, Thorsby E, Gaudernack G. Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet 1995; 346:1399-400; PMID:7475823; http://dx.doi.org/10.1016/S0140-6736(95)92408-6
  • Yamaguchi K, Enjoji M, Tsuneyoshi M. Pancreatoduodenal carcinoma: a clinicopathologic study of 304 patients and immunohistochemical observation for CEA and CA19-9. J Surg Oncol 1991; 47:148-54; PMID:2072697; http://dx.doi.org/10.1002/jso.2930470303
  • Kotera Y, Fontenot JD, Pecher G, Metzgar RS, Finn OJ. Humoral immunity against a tandem repeat epitope of human mucin MUC-1 in sera from breast, pancreatic, and colon cancer patients. Cancer Res 1994; 54:2856-60
  • Johnston FM, Tan MC, Tan BR, Jr, Porembka MR, Brunt EM, Linehan DC, Simon PO Jr, Plambeck-Suess S, Eberlein TJ, Hellstrom KE et al. Circulating mesothelin protein and cellular antimesothelin immunity in patients with pancreatic cancer. Clin Cancer Res 2009; 15:6511-8; http://dx.doi.org/10.1158/1078-0432.CCR-09-0565
  • Clark CE, Beatty GL, Vonderheide RH. Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer. Cancer Letters 2009; 279:1-7; PMID:19013709; http://dx.doi.org/10.1016/j.canlet.2008.09.037
  • Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nature Rev Immunol 2006; 6:836-48; http://dx.doi.org/10.1038/nri1961
  • Swann JB, Hayakawa Y, Zerafa N, Sheehan KC, Scott B, Schreiber RD, Hertzog P, Smyth MJ. Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J Immunol 2007; 178:7540-9; PMID:17548588; http://dx.doi.org/10.4049/jimmunol.178.12.7540
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331(6024):1565-70; PMID:21436444; http://dx.doi.org/10.1126/science.1203486
  • Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, McMichael AJ, Davis MM. Phenotypic analysis of antigen-specific T lymphocytes. Science 1996; 274:94-6; PMID:8810254; http://dx.doi.org/10.1126/science.274.5284.94
  • Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012; 21:822-35; PMID:22698406; http://dx.doi.org/10.1016/j.ccr.2012.04.025
  • Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-α. Science 2010; 330:827-30; PMID:21051638; http://dx.doi.org/10.1126/science.1195300
  • Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 2009; 58:49-59; http://dx.doi.org/10.1007/s00262-008-0523-4
  • Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res 2006; 12:5423-34
  • Nomi T, Sho M, Akahori T, Hamada K, Kubo A, Kanehiro H, Nakamura S, Enomoto K, Yagita H, Azuma M et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res 2007; 13:2151-7
  • Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011; 331:1612-6; PMID:21436454
  • Mantovani A, Romero P, Palucka AK, Marincola FM. Tumour immunity: effector response to tumour and role of the microenvironment. Lancet 2008; 371:771-83; PMID:18275997
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Rev Cancer 2012; 12:252-64
  • Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Eng J Med 2010; 363:711-23
  • Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, Sherry RM, Topalian SL, Yang JC, Lowy I et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010; 33:828-33; PMID:20842054
  • Beatty GL, Torigian DA, Chiorean EG, Saboury B, Brothers A, Alavi A, Troxel AB, Sun W, Teitelbaum UR, Vonderheide RH et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res 2013; 19:6286-95
  • Kawaoka T, Oka M, Takashima M, Ueno T, Yamamoto K, Yahara N, Yoshino S, Hazama S. Adoptive immunotherapy for pancreatic cancer: cytotoxic T lymphocytes stimulated by the MUC1-expressing human pancreatic cancer cell line YPK-1. Oncol Rep 2008; 20:155-63; PMID:18575732
  • Kondo H, Hazama S, Kawaoka T, Yoshino S, Yoshida S, Tokuno K, Takashima M, Ueno T, Hinoda Y, Oka M. Adoptive immunotherapy for pancreatic cancer using MUC1 peptide-pulsed dendritic cells and activated T lymphocytes. Anticancer Res 2008; 28:379-87; PMID:18383873
  • Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM, Varela-Rohena A, Haines KM, Heitjan DF, Albelda SM et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 2009; 106:3360-5; PMID:19211796
  • Shirasu N, Shibaguci H, Kuroki M, Yamada H, Kuroki M. Construction and molecular characterization of human chimeric T-cell antigen receptors specific for carcinoembryonic antigen. Anticancer Res 2010; 30:2731-8; PMID:20683006
  • Wilkie S, Burbridge SE, Chiapero-Stanke L, Pereira AC, Cleary S, van der Stegen SJ, Spicer JF, Davies DM, Maher J. Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J Biol Chem 2010; 285:25538-44; PMID:20562098
  • Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, Zhong X, Sadelain M, Eshhar Z, Rosenberg SA, Morgan RA. A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol 2009; 183:5563-74; PMID:19843940
  • Weden S, Klemp M, Gladhaug IP, Møller M, Eriksen JA, Gaudernack G, Buanes T. Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int J Cancer J Int Du Cancer 2011; 128:1120-8
  • Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, Propper D, Coxon F, Ross P, Madhusudan S, Roques T et al. Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncol 2014; 15:829-40; PMID:24954781; http://dx.doi.org/10.1016/S1470-2045(14)70236-0
  • Wobser M, Keikavoussi P, Kunzmann V, Weininger M, Andersen MH, Becker JC. Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunol Immunother 2006; 55:1294-8; PMID:16315030; http://dx.doi.org/10.1007/s00262-005-0102-x
  • Finn OJ. Vaccines for cancer prevention: a practical and feasible approach to the cancer epidemic. Cancer Immunol Res 2014; 2:708-13; PMID:25092812; http://dx.doi.org/10.1158/2326-6066.CIR-14-0110
  • Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, Morse M, Zeh H, Cohen D, Fine RL 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; PMID:25584002; http://dx.doi.org/10.1200/JCO.2014.57.4244
  • Quispe-Tintaya W, Chandra D, Jahangir A, Harris M, Casadevall A, Dadachova E, Gravekamp C. Nontoxic radioactive Listeria(at) is a highly effective therapy against metastatic pancreatic cancer. Proc Natl Acad Sci U S A 2013; 110:8668-73; PMID:23610422; http://dx.doi.org/10.1073/pnas.1211287110
  • Passero FC, Jr, Saif MW. Advancements in the Management of Pancreatic Cancer: 2015 ASCO Gastrointestinal Cancers Symposium (San Francisco, CA, USA. January 15-17, 2015). JOP 2015; 16:99-103; PMID:25791541
  • Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012; 491:399-405; PMID:23103869; http://dx.doi.org/10.1038/nature11547
  • Zitvogel L, Apetoh L, Ghiringhelli F, André F, Tesniere A, Kroemer G. The anticancer immune response: indispensable for therapeutic success?. J Clin Invest 2008; 118:1991-2001; PMID:18523649; http://dx.doi.org/10.1172/JCI35180
  • Garcia-Martinez E, Gil GL, Benito AC, González-Billalabeitia E, Conesa MA, García García T, García-Garre E, Vicente V, Ayala de la Peña F. Tumor-infiltrating immune cell profiles and their change after neoadjuvant chemotherapy predict response and prognosis of breast cancer. Breast Cancer Res 2014; 16:488; PMID:25432519; http://dx.doi.org/10.1186/s13058-014-0488-5
  • Denkert C, von Minckwitz G, Brase JC, Sinn BV, Gade S, Kronenwett R, Pfitzner BM, Salat C, Loi S, Schmitt WD et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol 2015; 33:983-91; http://dx.doi.org/10.1200/JCO.2014.58.1967
  • Berd D, Mastrangelo MJ. Effect of low dose cyclophosphamide on the immune system of cancer patients: depletion of CD4+, 2H4+ suppressor-inducer T-cells. Cancer Res 1988; 48:1671-5; PMID:2830969
  • Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood 2005; 105:2862-8; PMID:15591121; http://dx.doi.org/10.1182/blood-2004-06-2410
  • Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, Martin F, Apetoh L, Rébé C, Ghiringhelli F. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 2010; 70:3052-61; PMID:20388795; http://dx.doi.org/10.1158/0008-5472.CAN-09-3690
  • Correale P, Cusi MG, Del Vecchio MT, Aquino A, Prete SP, Tsang KY, Micheli L, Nencini C, La Placa M, Montagnani F et al. Dendritic cell-mediated cross-presentation of antigens derived from colon carcinoma cells exposed to a highly cytotoxic multidrug regimen with gemcitabine, oxaliplatin, 5-fluorouracil, and leucovorin, elicits a powerful human antigen-specific CTL response with antitumor activity in vitro. J Immunol 2005; 175:820-8; PMID:16002679; http://dx.doi.org/10.4049/jimmunol.175.2.820
  • Yuan L, Wu L, Chen J, Wu Q, Hu S. Paclitaxel acts as an adjuvant to promote both Th1 and Th2 immune responses induced by ovalbumin in mice. Vaccine 2010; 28:4402-10; PMID:20434553; http://dx.doi.org/10.1016/j.vaccine.2010.04.046
  • Pfannenstiel LW, Lam SS, Emens LA, Jaffee EM, Armstrong TD. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol 2010; 263:79-87; PMID:20346445; http://dx.doi.org/10.1016/j.cellimm.2010.03.001
  • John J, Ismail M, Riley C, Askham J, Morgan R, Melcher A, Pandha H. Differential effects of Paclitaxel on dendritic cell function. BMC Immunol 2010; 11:14; PMID:20302610; http://dx.doi.org/10.1186/1471-2172-11-14
  • Liu SV, Powderly JD, Camidge DR, Ready N, Heist RS, Hodi S, Giaccone G, Liu B, Wallin J, Funke RF et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with platinum-based doublet chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2015; 33(suppl; abstr 8030)
  • Papadimitrakopoulou V, Patnaik A, Borghaei H, Stevenson J, Gandhi L, Gubens MA, Yang JC, Sequist LV, Ge JY, Bourque J et al. Pembrolizumab (pembro; MK-3475) plus platinum doublet chemotherapy (PDC) as front-line therapy for advanced non-small cell lung cancer (NSCLC): KEYNOTE-021 Cohorts A and C. J Clin Oncol 2015; 33(suppl; abstr 8031)
  • Marabelle A, Kohrt H, Caux C, Levy R. Intratumoral immunization: a new paradigm for cancer therapy. Clin Cancer Res 2014; 20:1747-56; http://dx.doi.org/10.1158/1078-0432.CCR-13-2116
  • Coley WB. The Treatment of Malignat Tumors By Repeated Inoculations of Erysipelas: With A Report of Ten Original Cases. Am J Med Sci 1893; 105:487-510; http://dx.doi.org/10.1097/00000441-189305000-00001
  • Nauts HC, Swift WE, Coley BL. The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, M.D., reviewed in the light of modern research. Cancer Res 1946; 6:205-16; PMID:21018724
  • Cohen MH, Jessup JM, Felix EL, Weese JL, Herberman RB. Intralesional treatment of recurrent metastatic cutaneous malignant melanoma: a randomized prospective study of intralesional Bacillus Calmette-Guerin versus intralesional dinitrochlorobenzene. Cancer 1978; 41:2456-63; PMID:657108; http://dx.doi.org/10.1002/1097-0142(197806)41:6%3c2456::AID-CNCR2820410654%3e3.0.CO;2-B
  • Morton DL, Eilber FR, Holmes EC, Hunt JS, Ketcham AS, Silverstein MJ, Sparks FC. BCG immunotherapy of malignant melanoma: summary of a seven-year experience. Annals Surg 1974; 180:635-43; http://dx.doi.org/10.1097/00000658-197410000-00029
  • Bier J, Kleinschuster S, Bier H, Rapp H. Intratumor immunotherapy with BCG cell wall preparations: development of a new therapy approach for head-neck tumors. Arch Oto-Rhino-Laryngol 1982; 236:245-55; http://dx.doi.org/10.1007/BF00454216
  • Falk RE, MacGregor AB, Landi S, Ambus U, Langer B. Immunostimulation with intraperitoneally administered bacille Calmette Guerin for advanced malignant tumors of the gastrointestinal tract. Surg Gynecol Obstet 1976; 142:363-8
  • Shimada S, Yano O, Inoue H, Kuramoto E, Fukuda T, Yamamoto H, Kataoka T, Tokunaga T. Antitumor activity of the DNA fraction from Mycobacterium bovis BCG. II. Effects on various syngeneic mouse tumors. J Natl Cancer Inst 1985; 74:681-8; PMID:3856070
  • Tokunaga T, Yamamoto T, Yamamoto S. How BCG led to the discovery of immunostimulatory DNA. Japanese J Infect Dis 1999; 52:1-11
  • Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, Kim YH, Hoppe RT, Knox SJ, Shin LK et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol 2010; 28:4324-32; http://dx.doi.org/10.1200/JCO.2010.28.9793
  • Kim YH, Gratzinger D, Harrison C, Brody JD, Czerwinski DK, Ai WZ, Morales A, Abdulla F, Xing L, Navi D et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study. Blood. Blood 2012; 119:355-63; PMID:22045986; http://dx.doi.org/10.1182/blood-2011-05-355222
  • Hiniker SM, Chen DS, Knox SJ. Abscopal effect in a patient with melanoma. N Eng J Med 2012; 366:2035; author reply -6; http://dx.doi.org/10.1056/NEJMc1203984
  • Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, Mu Z, Rasalan T, Adamow M, Ritter E et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 2012; 366:925-31; PMID:22397654; http://dx.doi.org/10.1056/NEJMoa1112824
  • Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, Friedman K, Ponzo F, Babb JS, Goldberg J et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 2015; 16:795-803; PMID:26095785; http://dx.doi.org/10.1016/S1470-2045(15)00054-6
  • Bindea G, Mlecnik B, Angell HK, Galon J. The immune landscape of human tumors: Implications for cancer immunotherapy. Oncoimmunology 2014; 3:e27456; PMID:24800163; http://dx.doi.org/10.4161/onci.27456
  • Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, Tosolini M, Camus M, Berger A, Wind P et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313:1960-4; PMID:17008531; http://dx.doi.org/10.1126/science.1129139
  • Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. Biomed Res Int 2014; 2014:751392; PMID:25110692
  • Olmos D, Brewer D, Clark J, Danila DC, Parker C, Attard G, Fleisher M, Reid AH, Castro E, Sandhu SK et al. Prognostic value of blood mRNA expression signatures in castration-resistant prostate cancer: a prospective, two-stage study. Lancet Oncol 2012; 13:1114-24; PMID:23059046; http://dx.doi.org/10.1016/S1470-2045(12)70372-8
  • Ross RW, Galsky MD, Scher HI, Magidson J, Wassmann K, Lee GS, Katz L, Subudhi SK, Anand A, Fleisher M et al. A whole-blood RNA transcript-based prognostic model in men with castration-resistant prostate cancer: a prospective study. Lancet Oncol 2012; 13:1105-13; PMID:23059047; http://dx.doi.org/10.1016/S1470-2045(12)70263-2
  • Marabelle A, Filatenkov A, Sagiv-Barfi I, Kohrt H. Radiotherapy and toll-like receptor agonists. Seminars Radiat Oncol 2015; 25:34-9; http://dx.doi.org/10.1016/j.semradonc.2014.07.006
  • Bowen CR, Meek S, Williams M, Grossmann KF, Andtbacka RHI, Bowles TL, Hyngstrom JR, Leachman SA, Grossman D, Holmen SL et al. A phase I study of intratumoral injection of ipilimumab and interleukin-2 in patients with unresectable stage III-IV melanoma. J Clin Oncol 2015; 33(suppl; abstr 3018)
  • Marabelle A, Kohrt H, Levy R. New insights into the mechanism of action of immune checkpoint antibodies. Oncoimmunology 2014; 3:e954869; PMID:25610751; http://dx.doi.org/10.4161/21624011.2014.954869
  • DiLillo DJ, Ravetch JV. Fc-Receptor Interactions Regulate Both Cytotoxic and Immunomodulatory Therapeutic Antibody Effector Functions. Cancer Immunol Res 2015; 3:704-13; PMID:26138698; http://dx.doi.org/10.1158/2326-6066.CIR-15-0120
  • Zamarin D, Holmgaard RB, Subudhi SK, Park JS, Mansour M, Palese P, Merghoub T, Wolchok JD, Allison JP. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med 2014; 6:226ra32; PMID:24598590; http://dx.doi.org/10.1126/scitranslmed.3008095
  • Houot R, Levy R. T-cell modulation combined with intratumoral CpG cures lymphoma in a mouse model without the need for chemotherapy. Blood 2009; 113:3546-52; PMID:18941113; http://dx.doi.org/10.1182/blood-2008-07-170274