Introduction

Pancreatic cancer is the fourth leading cause of cancer-related death in the United States [13] and the fifth most common cause of such deaths in Japan [4]. Although surgical resection is considered to be the only curative therapy for pancreatic cancer, only 20 % of patients have resectable disease at the time of diagnosis [5, 6]. In addition, advanced pancreatic cancer patients exhibit a median survival time (MST) of approximately six months and a 5-year overall survival rate of less than 5 %, despite efforts to manage the tumors with chemotherapy, radiotherapy and other treatments [3, 58].

In 1997, Burris et al. reported that gemcitabine monotherapy is superior to fluorouracil (5-FU) monotherapy for treating pancreatic ductal adenocarcinoma (PDAC) [9]. Gemcitabine monotherapy has subsequently become the standard chemotherapy for PDAC, resulting in an MST of 5.65 months (Table 1). Currently, three protocols have proven to be superior to gemcitabine monotherapy. Combining gemcitabine with erlotinib improved the MST of PDAC to 6.24 months in the NCIC CTG PA3 trial [10], while combining gemcitabine with nab-paclitaxel improved the MST to 8.7 months in the MPACT trial [11]. FOLFIRINOX achieved the longest MST for PDAC (11.1 months) in the ACCORD11 trial [12], and the GEST study obtained similar clinical outcomes. S-1 is an oral fluoropyrimidine derivative that has been shown to be effective against various cancers, and a previous study found that it is at least as effective as gemcitabine against PDAC [13]. In addition, treatment with a combination of gemcitabine + S-1 has been demonstrated to result in an MST of 10.1 months [14]. Although these chemotherapies extend the survival period among PDAC patients, they also result in serious adverse events. Therefore, the optimal chemotherapy regimen for PDAC depends on the patient’s performance status.

Table 1 Chemotherapy for advanced pancreatic cancer

There have been numerous attempts to develop vaccine therapies for cancer over the past century [2, 3]. Although clinical trials of such vaccines have obtained promising results in specific patients, none of the tested vaccines has exhibited significant improvements in efficacy compared with established therapies. In addition, several issues must be resolved before vaccine therapies can be used in the clinical setting. Tumor-associated antigens (TAA) have been demonstrated to recognize specific human leukocyte antigens (HLA) [15]. Theoretically, the tumor lysate contains all of the antigens expressed by the tumor, and cytotoxic T lymphocytes (CTL) are capable of recognizing some of these antigens [16]. All vaccines for pancreatic cancer are based on the fact that CTL recognize TAA expressed on tumor cells and subsequently attack these cells. The question is how strongly and specifically each TAA stimulates CTL in vivo in the clinical setting. Immune tolerance can develop via various mechanisms, including the downregulation of the major histocompatibility complex (MHC) molecule expression, induction of T cell anergy, reductions in the number of immune effectors and increases in the number of regulatory T cells [17, 18], which may explain why no cancer vaccine therapy has been established as a standard treatment for advanced PDAC. Therefore, in this study, we comprehensively reviewed the clinical outcomes of vaccine therapy against advanced PDAC.

Peptide-based vaccines developed within the past few years

MUC1

Mucin 1, cell surface associated, (MUC1) is a type I transmembrane protein containing multiple tandem repeats of a 20-amino acid sequence. Several MUC1 peptides have been tested as vaccines in the clinical setting; however, most of them have failed to activate CTL [1921]. Ramanathan et al. [22]; Yamamoto et al. [23] injected pancreatic patients with a vaccine containing a 100-mer extracellular tandem repeat domain of MUC1 and Montanide ISA-51, and both studies obtained similar clinical responses; i.e., the authors detected cytokines (interferon (IFN)-γ or interleukin (IL)-4) and anti-MUC1 antibodies in the patients’ sera but did not observe any significant clinical effects. Another recent study involving a vaccine based on a different MUC1 epitope showed similar clinical outcomes, i.e., all seven patients had progressive disease (PD), although some of the patients exhibited immunological responses, such as IFN-γ and granzyme B secretion [24].

K-RAS mutants

K-RAS mutations are frequently found in patients with PDAC. Vaccines targeting mutations in codon 12 of the K-RAS gene have been tested as treatments for advanced [25] or postoperative [26] PDAC in the clinical setting. Gjertsen et al. [[21]] investigated the utility of a K-RAS peptide vaccine containing granulocyte–macrophage colony-stimulating factor (GM-CSF) in 10 patients who had undergone potentially curative resection (CTN RAS 95002) and 38 patients with advanced disease (CTN RAS 97004). In that study, one patient achieved a partial response (PR), which lasted for 28 months, and the MST of the immunological responders was 4.9 months, compared to 2.0 months for the non-responders.

Human telomerase reverse transcriptase (hTERT)

Human telomerase reverse transcriptase (hTERT) is frequently expressed in cancer cells [27]. hTERT maintains functional telomeres at the end of chromosomes, which protect against cell senescence [28]. A vaccine against pancreatic cancer containing the telomerase peptide GV1001: hTERT (611-626) and GM-CSF was examined by Bernhardt et al. [29], who found the MST of the immunological responders and non-responders to be 7.2 and 2.9 months, respectively.

Vascular endothelial growth factor receptor 2 (VEGFR2)

Vascular endothelial growth factor (VEGF) plays an important role in the progression of PDAC. The type 2 VEGF receptor (VEGFR2) is expressed in PDAC and associated with tumor neovascularization. Miyazawa et al. [[30]] investigated the efficacy of combined treatment consisting of PDAC with a VEGFR2-169 peptide-based vaccine and gemcitabine chemotherapy and reported that one patient achieved a PR, while the disease control rate was 67 %. In addition, the MST was 7.7 months, although 15/18 patients were chemotherapy naive.

G17DT (gastrimmune)

Gastrin is expressed in PDAC and plays a role in regulating the autocrine, paracrine and endocrine systems [31]. The administration of the anti-gastrin immunogen G17DT results in increased serum antibody levels and reduced tumor growth in patients with gastrointestinal malignancies [32]. A randomized, double-blind, placebo-controlled multicenter trial of G17DT was also recently performed [33]. Although, among the intention to treat (ITT) population, no significant differences in MST were detected between the PDAC patients treated with G17DT and those given the placebo, the MST of the two groups differed significantly after excluding major protocol violators and censoring for chemotherapy.

Heat shock protein (HSP)

Heat shock protein (HSP) itself is not an immunogen; however, it acts as a chaperone or carrier of antigenic peptides and possesses a repertoire of cellular peptides for pancreatic cancer [34]. Furthermore, HSPPC-96 (Oncophage) has been tested as a vaccine in the adjuvant setting after complete resection of PDAC [35]. In the latter study, the MST of PDAC was reported to be 2.9 months after surgery; however, this did not result in further clinical studies because only two of 10 patients exhibited increased enzyme-linked immunospot (ELISPOT) reactivity.

Biological vaccines

Fowlpox viral vaccine

Carcinoembryonic antigen (CEA) and MUC1 are highly expressed in PDAC [36]. Viral vectors carrying CEA, MUC1 and TRICOM [a triad of costimulatory molecules: B7.1, intercellular adhesion molecule 1 (ICAM-1) and lymphocyte function-associated antigen 3 (LFA-3)] have been investigated as vaccines against advanced PDAC [37]. In one study, a vaccinia viral vector was used for the initial T cell priming, and a fowlpox viral vector was used for immune boosting. Although this treatment resulted in an MST of 6.3 months (1.5–21.1 months), the five patients who showed T cell responses achieved a longer survival period than the five patients who did not (15.1 and 3.9 months, respectively; P = 0.002) [38]. It should be noted that GM-CSF was used as a vaccine adjuvant in the latter trial (Table 2).

Table 2 Peptide-based vaccines and biological vaccines for advanced pancreatic cancer

Live-attenuated, double-deleted (LADD) Listeria monocytogene vaccine

ANZ-100 is a live-attenuated double-deleted Listeria monocytogene strain (LADD; Lm ΔactA/ΔinlB) found to induce a local proinflammatory response, resulting in the activation of innate and adaptive effector cells [39]. Mesothelin is expressed in PDAC and plays an important role in tumor progression [40]. CRS-207 is a LADD Lm strain that delivers mesothelin antigens into class I and II antigen-processing pathways [41]. In a study examining the utility of CRS-207 as a treatment for advanced cancer, three of the seven subjects with PDAC were long-term survivors, although the detection of a mesothelin-specific T cell response was not correlated with survival [41].

Recent vaccine therapies

WT1

Kobayashi et al. reported a retrospective analysis of 255 advanced PDAC patients who were treated with dendritic cell (DC) vaccines containing Wilms tumor 1 (WT1) and MUC1 after being recruited from seven institutions that followed a unified standard operating procedure. The MST of these patients was 9.9 months [42]. Nishida et al. also examined the utility of chemo-vaccine therapy in which a WT1-based vaccine was used in combination with the administration of 1,000 mg/m2 of gemcitabine weekly. The latter regimen resulted in an MST of 8.1 months among 31 advanced PDAC patients [43]. In addition, the MST of the immunological responders in these two studies was very similar (10.4 and 10.9 months, respectively) (Table 3).

Table 3 Recently developed peptide-based vaccines and multiple vaccines for advanced pancreatic cancer

KIF20A

Kinesin family member 20A (KIF20A) plays an important role in the trafficking of molecules and organelles [44] and is one of the molecules targeted by vaccines against PDAC. A KIF20A vaccine was recently tested using different regimens, including vaccine monotherapy [45] and chemo-vaccine therapy involving gemcitabine [46], and similar MST values were reported in both studies (4.7 and 5.8 months, respectively).

Carcinoembryonic antigen (CEA)

CEA is a 180-kDa immunoglobulin-like molecule expressed on the surface of 90 % of PDAC tumor cells [47]. CAP1-6D, a modified CEA peptide, was combined with Montanide/GM-CSF to produce a vaccine against pancreatic cancer that was subsequently tested in advanced PDAC patients [48]. The MST of the 19 patients was 11.1 months, and one patient, randomized into the 0.01 mg arm, achieved a complete response (CR).

Survivin2B

Survivin is a member of the inhibitors of apoptosis (IAP) family of proteins that protect apoptotic signals by inhibiting the caspase activity [49, 50]. Hence, survivin-expressing cancer cells escape from apoptosis and do not die. Using a peptide-binding assay, we found that the survivin2B 80–88 peptide induces a strong CTL response [51]. We also examined the effects of a survivin2B 80–88 peptide-based vaccine on various cancers in the clinical setting and obtained promising outcomes. In particular, the anti-tumor effect of the survivin2B 80–88 peptide was enhanced by combining it with incomplete Freud’s adjuvant and IFN-α injection. Our preliminary clinical study demonstrated a 66.6 % disease control rate in advanced PDAC patients (four of six patients) [52]. Moreover, the PDAC patients in our recent clinical phase I study exhibited an MST of 9.6 months.

Multiple vaccines

Personalized peptides

In a previous study, a set of 31 peptides was used to create personalized vaccines for advanced PDAC [53]. A maximum of four peptides were selected from among the 31-peptide set based on the results of HLA typing and the patients’ peptide-specific IgG titers. Eight patients received vaccine monotherapy, and 31 patients received chemo-vaccine therapy. In the chemo-vaccine therapy group, gemcitabine was administered in eight patients, S-1 was administered in six patients and gemcitabine + S-1 was given in eight patients. The overall MST was 9.6 months, although that of the patients who underwent monotherapy was 7.9 months. Yanagimoto et al. reported similar clinical outcomes for chemo-vaccine therapy involving personalized vaccines and gemcitabine based on the same regimen [54]. The MST of the patients in the latter study was 9.0 months, although that of the immunological responders was 15.5 months. None of the patients in either study achieved CR (Table 3).

Autologous tumor lysate combined with lymphokine-activated killer cell therapy

Kimura et al. treated 49 PDAC patients with vaccines based on five different peptides and autologous tumor lysate, although the vaccine preparation regimens and anti-tumor therapies varied in each case [16]. Two patients achieved CR after treatment with a combination of DC cell and lymphokine-activated killer cell (LAK) therapy. The MST of the patients treated with LAK + gemcitabine and S-1 was 16.9 months, whereas that of all patients was 12.0 months. It should be noted that the survival time was calculated from the day after the first vaccination, which may have resulted in a shorter survival time (by a couple of months) than would have been obtained using the methods employed in other studies. It is very difficult to evaluate the clinical results of this study due to the effects of the different therapeutic strategies used in each case. However, the fact that multiple patients achieved CR will encourage researchers to pursue this approach further.

GVAX pancreas with CRS-207

GVAX is a series of irradiated GM-CSF-secreting allogeneic pancreatic cell lines that elicit broad antigenic responses. CRS-207 is a LADD Lm strain (Lm ΔactA/ΔinlB) that expresses mesothelin and stimulates the innate and adaptive immune systems. A phase II randomized control trial of GVAX pancreas combined with CRS-207 versus GVAX pancreas alone was presented at the 2014 American Society of Clinical Oncology (ASCO) Gastrointestinal Cancers Symposium [55]. Interestingly, the clinical results demonstrated that both treatments had dose-dependent survival benefits. The MST of the patients who received three or more rounds of vaccine therapy was 9.7 months, and the MST of the GVAX with CRS-207 arm was longer than that of the GVAX-alone arm (6.1 vs. 3.9 months; P = 0.01) [56].

Evaluation of therapeutic activity in solid tumors

The response of solid tumors is evaluated using either the WHO [57] or RECIST criteria [58]. The RECIST criteria were developed because the WHO criteria are quite complex and measuring all visible lesions in two dimensions is both time consuming and subject to measuring bias [59]. However, the use of immunotherapeutic agents in cancer patients is associated with the following problems: (a) The measurable anti-tumor activity can take longer to appear during immunotherapy than during cytotoxic therapy; (b) Responses to immunotherapy can occur after the standard criteria for progressive disease (PD) have been met; (c) Discontinuing immunotherapy may not be appropriate in some cases, unless PD is confirmed; (d) Allowing for “clinically insignificant” PD (e.g., small new lesions developing in the presence of other responsive lesions) is recommended; and (e) Durable stable disease (SD) may represent the anti-tumor activity [60]. Therefore, the immune-related response criteria (irRC) were developed to evaluate the immunotherapeutic activity in solid tumors [61]. The most important aspects of the irRC criteria are that (a) new lesions are not classified as PD and (b) two consecutive observations obtained at least four weeks apart are required to diagnose PD. However, the clinical utility of the irRC remains unclear and these criteria may require further optimization [61] (Table 4).

Table 4 Evaluation of therapeutic activity in solid tumors

Future research topics

Initial time point for survival assessments

The initial time point for survival assessments should be unified to allow clinical outcomes to be compared between studies. Most PDAC patients already have advanced disease at the time of diagnosis [6]. In addition, the adverse effects of chemotherapies differ markedly among the various regimens [8]. Therefore, the status of PDAC patients at the time point at which they are registered can differ both within and between clinical studies. Kobayashi et al. reported that the MST from the date of diagnosis and the MST from the first vaccination are very different (16.5 vs. 9.9 months) [42]. Therefore, MST data must be interpreted carefully.

Vaccine therapy and chemotherapy

The goal of vaccine therapy for cancer is to increase the native immunity of cancer patients. However, chemotherapy causes irreversible damage to proliferating cancer cells as well as immune cells, including T and B cells. Therefore, there is a conflict between the fundamental principles of these two treatments. Chemotherapy is currently the gold standard treatment for advanced PDAC. Although the biological mechanisms of vaccine therapy and chemotherapy conflict with each other, the anti-cancer activity of vaccine monotherapy or chemo-vaccine combination therapy should be greater than that of chemotherapy alone.

Slow clinical response to vaccine therapy

It is very hard to achieve a complete response (CR) with vaccine therapy alone. We reviewed 19 studies involving a total of 860 patients and found that CR responses were obtained in only three cases. Although none of these studies involved a large number of patients, the poor reported response rates are a concern. One of the patients who achieved a CR was administered CEA CAP1-6D + Montanide/GM-CSF therapy, while the other two were treated with WT1, Her2, CEA, MUC1, cancer antigen 125 and autologous tumor lysate vaccines combined with DC cell-based LAK therapy and chemotherapy. Immunological responses require a long time to control tumor growth and achieve remission. The primary goal of vaccine therapy is to achieve long-term SD [62]. Most previous clinical studies of PDAC involved patients with advanced disease for whom no other therapies were available. Therefore, vaccine therapy may be suitable for patients in other clinical stages or possibly a useful postoperative adjuvant therapy. The main advantage of vaccine therapy is that it has few adverse effects, although it has also demonstrated minimal clinical effects in previous trials. We are currently conducting a phase II study of the survivin2B 80–88 peptide + Montanide + IFN-β as a treatment for PDAC (SUCCESS, Study of Unresectable CanCEr with Survivin-2B peptide vaccine in Sapporo: UMIN000012146), in which half of the required patients have been recruited. The clinical results of the SUCCESS phase II study will be reported by the end of next year.