Loading

Journal of Cancer Biology and Research

Vaccine Therapy for Pancreatic Cancer: As a Novel Therapeutic Approach?

Review Article | Open Access | Volume 2 | Issue 1

  • 1. Department of Hepatobiliary and Pancreatic Surgery, Ningbo University, China
  • 2. Department of Pharmacology, Gandaki Medical College, Pokhara, Nepal
+ Show More - Show Less
Corresponding Authors
LU Cai-de, Department of Hepatobiliary and Pancreatic Surgery, Lihuili Hospital, Ningbo University, School of Medicine, Ningbo 315060, China
ABSTRACT

Pancreatic cancer is one of the deadliest human malignancies and little progress  has been achieved in its treatment over the past decades. Historically, chemotherapy  or radiotherapy did not provide significant survival benefit in advanced pancreatic  cancer. Thus, new therapeutic approaches are needed. As there is strong evidence that  pancreatic cancer elicits antitumor immune responses, scientists have tried to stimulate  the   of the immune system to fight against pancreatic cancer, but  has not reached to expected result. Pancreatic cancer activates both antitumor immune  responses and immunosuppressive mechanisms leading to tumor  and  progression. This action is achieved through mobilization and activation of immune  suppressive cells (CAFs), tolerogenic DCs, MDSCs, TAMs, Treg cells and cancer cellsderived soluble factors that promote the induction of tolerance through the generation  of CD4+a chain of IL-2R (CD25)+ fork head box P3 (Foxp3) subset. Vaccine therapy  relies on the administration of biological preparations that include an antigen that is  specifically expressed by malignant cells, boosting the natural ability of the immune  system to react against neoplastic cells. Potent vaccines stimulate antigen presentation  by dendritic cells, hence driving the expansion of antigen-specific effector and memory  T cells. In this paper we analyze recent preclinical and clinical efforts towards vaccine  therapy for pancreatic cancer designed to target pancreatic cancer-associated  antigens and to elicit an antitumor response in vivo.

KEYWORDS

 Pancreatic cancer ; Immunotherapy ;Dendritic cells ;Cancer vaccine ; MDSCs ; Treg.

CITATION

Yadav DK, Lu CD, Rajesh Kumar Y (2014) Vaccine Therapy for Pancreatic Cancer: As a Novel Therapeutic Approach? J Cancer Biol Res 2(1): 1046

ABBREVIATIONS

Pancreatic cancer-PC ; 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin-FOLFIRINOX; Myeloid-derived suppressor cells – MDSC; T regulatory cells- Treg ;Tumor microenvironment –TME; Tumor associated macrophages-TMAs; Natural killer Cells- NK cell; Cytotoxic T lymphocyte –CTL; Granulocyte-macrophage colony-stimulating factor- GM-CSF; Tumor associated antigensTAAs; Dendritic cell- DCs; Human leukocyte antigen –HLA; Major histocompatibility complex -MHC; C-phosphate-G – CpG.

Core Tip: This article includes vaccine therapy strategy for pancreatic cancer and different immunogenic obstacles due dysfunctional immune system in PC leading to tumor development, tumor progression, and resistance to therapy.

INTRODUCTION

Pancreatic Cancer (PC) is the fourth leading cause of cancer death in United States and seventh in China, with 45,220 new cases in 2013 and estimated 38,460 deaths in the USA [1,2].Treatment of PC has become multimodal with chemotherapy, radiation and surgical resection in the hope of long term survival. The only chance for cure and long -term survival is microscopic negative margins with R0 surgical resection [3]. However, only 10% to 20 % of patients with PC have resectable disease at the time of diagnosis, approximately 30 to 40 percent have locally advanced tumor and another 40 percent will have metastatic disease at the time of diagnosis, [1,4] and thus palliative chemotherapy remains the only option for most of these patients [5]. The overall fiveyear survival probability is less than 5% for all stages combined [1,3,4,6]. Even with advancement in surgical techniques most of the patients undergoing complete surgical resection experience a recurrence [7]. Several autopsy studies suggest that 8%–15% of PC patients die with locally advanced disease and without metastatic spread [8].

There are only a few chemotherapeutic agents that have shown to be effective against PC to date, such as FOLFIRINOX regiment and more recently combination of gemcetabine and erlotinib has shown somehow better result in treatment of PC [9,10]. The survival benefits for patients treated with these regimens are marginal and hence we are in urgent need of novel therapeutic approaches against PC.

As in recent years researchers has grown deeper understanding in the field of cancer immunology and their antigens that have given great hope for new alternative treatments for a variety of solid tumors including PC. Emerging evidence supports a critical role for the immune system in PC tumor development, progression and eradication [11,12]. There is strong recent evidence that classical anticancer treatments heavily rely on the immune system for their effectiveness [13- 15]. Desmoplasia and the Tumor Microenvironment (TME) are increasingly seen as major contributors to chemoresistance in PC [16]. Studies has shown that Tumor Associated Macrophages (TMAs) can influence the response of cancer cells to chemotherapy in the context of a process known as environment-mediated drug resistance [17,18]. Targeting specific immunotherapy could be revolutionary step in the treatment of PC. In support of the PC-specific immunotherapy approaches there are many data showing PC patients generates B and T cells specific to antigens expressed on autologous pancreatic tumor cells [19-21] . Antigens expressed in PC cell such as Carcinoembryonic Antigen (CEA) (over 90%), [22] wilms’ tumor gene 1 (WT1) (75%), [23] mucin 1 (MUC1) (over 85%), [24] survivin (77%), [25] human telomerase reverse transcriptase (hTERT) (88%), [26] HER-2/ neu (61.2%), [27] p53 (67%), [28] mutated K-RAS (73%) [29] and a-enolase [30] can be potential targets for immunotherapy. This review summarizes clinical and preclinical efforts towards vaccine strategies and clinical trials for PC.

Dysfunctional immune system in PC-tumor development and progression

It has been found that dysfunctional immune system in PC has a critical role for the development and progression of tumor [11,12]. It is known that both the innate and the adaptive immune system are active against human cancers [31]. However, cancer cells escape the innate and adaptive immune responses (immunosurveillance) by immunoselection (selection of nonimmunogenic tumor cell variants, also known as immunoediting) or immunosubversion (active suppression of the immune response) [32]. Anticancer function of the immune system is achieved by cytotoxic CD8 T cells, T helper-1 (Th1) cells, mature Dendritic Cells (DCs), activated pro-inflammatory macrophages (M1) and NK cells [33]. Under the tumor induced immunosuppressive environment T helper cells acquire a T helper cell type 2 phenotype (Th2), which does not support cytotoxic CD8 T cell responses and is resistance toward tumors, macrophages (M1) switch to the immunosuppressive M2 state [34], In addition, the environment in pancreatic cancer is consist of not only cancer cells but also immune suppressive cells such as Cancer-Associated Fibroblasts (CAFs), tolerogenic DCs, MyeloidDerived Suppressor Cells (MDSCs), immunosuppressive TumorAssociated Macrophages (TAMs), and T regulatory cells (Treg cells) which inhibit effector immune responses [35]. There are increasing evidences that cancer cells-derived soluble factors promote the induction of tolerance through the generation of CD4+α chain of IL-2R (CD25)+ forkhead box P3 (Foxp3)+ Treg subset, which is linked to compromised antitumor immune responses [36]. Pancreatic cancer cells modulate the immune system and avoid detection by effector immune by production of immune suppressive cytokines (e.g., TGF-β, IL-10, and IL-6), by expressing surface molecules that mediate immune suppression (e.g., Vascular Endothelial Growth Factors (VEGFs ), Fas Ligand (Fas-L), Programmed Death-1 Ligand (PD-L1), indolamine-2, and 3-dioxygenase (IDO), [35] and interference with MHC class I peptide presentation by down-regulation of MHC class I expression or disabling of the antigen degradation or antigen insertion into the MHC class I [34]. Thus, leading to tumor progression.

Cancer vaccines strategies

What determines whether cancer vaccines can become a success in human immunotherapy? Exactly the same as required for infectious diseases, cancer has to be immunogenic and activate cytotoxic T cell responses. Consequently, cancer cells have to possess immunogenic antigens susceptible of being targeted by vaccination. Cancer vaccines are biological preparations that involve administering a tumor antigen with the aim of stimulating tumor-specifc immunity. Antigen can be delivered by number of ways in form of wholecell recombinant vaccines, Dendritic Cell (DC) vaccines that combine antigen with DCs to present to white cells, DNA vaccine by inserting viral, bacterial or Yeast DNA into human or animal cells, or T-cell receptor peptide vaccines by inserting peptides to modulate cell-mediated immunity. To be considered an ideal tumor vaccine candidate, expression of the antigen must be restricted to the tumor or only minimally expressed elsewhere in the body. Vaccination against tumor antigens is an attractive approach to adjuvant treatment after surgery, when tumor-induced immune suppression is minimal [37]. Cancer vaccines were first approved for hepatocellular carcinoma and cervical cancer prevention. More recently, the first vaccine (Sipuleucel-T, Provenge) was approved for the treatment of hormone refractory prostate cancer [38].

Compared to all other standard modalities (surgery, chemotherapy, radiotherapy, and adaptive immunotherapy), an effective vaccine-based immune response against tumor may be the only cancer treatment with the potential benefit to last a lifetime. Theoretically, vaccinated patients could mount an immune response able to either cure tumor or keep it under constant restraint (i.e., immune surveillance), delaying tumor recurrence and prolonging survival.

For the development of efficient vaccine researchers have been using different strategies such as:

- Cancer vaccine should seek for Tumor specific antigens and distinct from self-proteins.

- Selection of the appropriate adjuvant, molecules that activate antigen-presenting cells to stimulate an antigen specific cytotoxic T lymphocyte (CTL) mediated immune responses [39].

- Effective vaccine should seek to provide long term memory to prevent tumor recurrence which can activate both innate and adaptive immune system [40].

- Efforts towards improving the clinical efficacy of immune therapy should involve strategies to neutralize or overcome immune suppression.

 

VACCINES FOR PANCREATIC CANCER

Several vaccine therapy strategies are being actively tested in clinical trials. An over view of clinical trials in provided in Table 1

Vaccine

Targeting Vaccines

Phase

Study year and Investigators

Number of Pt and Stage of Disease

Name of Antigen

Adjuvant therapy

Result

Peptide Vaccine

KRAS vaccines

 

I/II

Gjertsen MK, [49]

5 patients with histologically confirmed PC

Mutated K-ras

 

Resulted in an immune response in 2,

showed longer survival

 

Gjertsen MK, [50]

Weden S

[52]

48 patients ,10 Surgically resected pts

and 38 pts with advanced disease

Mutated K-ras

GM-CSF

Peptide-specific immunity in 58% of pts.

Responders median survival 148 vs 61 days

for non-responders. 20% long term survivors

 

Abou-Alfa GK,

[51]

24 patients with resected PC

Mutated K-ras

GM-CSF

Median recurrence free survival 8.6 months; Median overall survival 20.3 months

II

Estimated Study Completion Date: 2014 [53]

100 Patients following resection

Mutated K-ras

Gemcitabine

Ongoing

Gastrin vaccines

 

 

Gilliam AD,

[59]

154 patients with advanced PC, unwilling or unable to take

chemotherapy

Gastrin (G17DT) versus placebo

 

151 days G17DT

vs. 82 days placebo p=0.03

HSP-peptide complex vaccines

I

Maki RG

[60]

10 patients with resected PC

HSPCC-96

 

Median overall survival was 2.2 years

WT1 vaccine

 

Nishida S,

[61]

32 patients with advanced pancreatic cancer

WT1

Gemcitabine

Median survival time and 1-year survival rate were 8.1 months and 29%

cancer-testis antigens (CT) vaccine

 

I

Okuyama

[70]

9 patients with advanced pancreatic cancer

CT and VEGFRs

 

The median overall survival (OS) was 207 days.

VEGFRs vaccine

I/II

Gotoh M

[71]

17 Unresectable, recurrent or metastatic patients

VEGF-R1

VEGF-R2

Gemcitabine

Completed, result not reported yet

(hTERT) vaccine

I/II

Bernhardt SL [72]

48 patients with nonresectable PC

Telomerase

GM-CSF

24/38 With immune responses. Induction of

immune response correlated with improved

survival

III

Buanes T

[73]

178 patients with advanced PC

Telomerase

Gemcitabine

No overall survival benefit

I

Crocenzi T

[74]

11 patients with Locally Advanced PC

Telomerase

tadalafil

Ongoing

Her2/neu vaccine

I

Morse M

[75]

12 patients with Her2/neu overexpressing tumors, including PC

Her2/neu

 

Ongoing

Recombinant Vaccines

MUC-1 and CEA in poxvirus

TRICOM, MUC-1

and CEA in poxvirus

with GM-CSF

III

Arlen PM

[79]

255 Metastatic pts following gemcitabine failure

MUC-1, CEA

TRICOM,GM-CSF

No overall survival benefit

Listeria vaccines

Live attenuated

Listeria vaccine

(aNZ-100) vs Live

attenuated mesothelin expressing Listeria vaccine (CrS-207)

I

Le DT

[80]

28 patients with mesothelioma, lung, pancreas, or ovarian cancer liver metastasis

Mesothelin

 

37% of patients in CrS-207

arm live after 15 months

Lethally irradiated genetically engineered allogeneic whole tumor and listeria

II

Le DT

[81]

90 Pts with metastatic disease

Mesothelin and whole tumor

GM-CSF secreting allogeneic PC cells and cyclophosphamide

Ongoing

Antigen pulsed  DCs vaccines

 

MUC-1 pulsed  (DCs) vaccines

 

I/II

Lepisto AJ

[82]

12 Pts with surgically resected pancreatic (10 pts) or biliary (2 pts)

cancer

MUC-1

 

4 Out of 12 pts alive at 4 years

LAK cell pulsed  (DCs) vaccines

 

 

Kimura Y

[83]

49 patients with unrecetable PC

(Stage III,, IVa, IVB)

LAK

gemcitabine or S-1

Median overall survival of patients receiving DC vaccine and chemotherapy plus LAK cell therapy was longer than those receiving DC vaccine in combination with chemotherapy but no LAK cells

Whole tumor cell vaccines

Algenpantucel-L

II

Hardacre JM

[84]

70 patients with resected PC

 

whole tumor

algenpantucel-L with chemotherapy (gemcitabine and 5 FU)+ radiotherapy

12-month disease-free survival was 62 %, and the 12-month overall survival was 86 %

Algenpantucel-L

III

Multicentered [85]

722 patients with resected PC ( stage I,II)

 

whole tumor

algenpantucel-L with chemotherapy (gemcitabine and 5 FU)+ radiotherapy

Ongoing

Allogeneic GM-CSF

I

Jaffee EM

[86]

14 patients with resected PC

whole tumor

GM-CSF vaccine with chemoradiotherapy

3 patients disease free at least

25 months after diagnosis

 

II

Lutz E [87]

60 patients with resected PC

whole tumor

GM-CSF vaccine with chemotherapy (5FU) and radiotherapy

disease free survival of 17.3 months and an overall survival of 24.8 months

 

II

Laheru DA [88]

60 patients with resected PC

whole tumor

GM-CSF secreting allogeneic PC cells,cyclophosphamide and cetuximab

Completed, result not reported yet

 

II

Laheru DA

[89]

56 Pts with resected PC

whole tumor

GM-CSF secreting allogeneic PC cells

Ongoing

 

II

Laheru DA

[90]

87 patients with resected PC

whole tumor

GM-CSF secreting allogeneic PC cells and IV vs oral metronomic

cyclophosphamide

Ongoing

 

I

Herman J

[91]

18 Pts with resected PC

whole tumor

GM-CSF secreting allogeneic PC cells and cyclophosphamide followed by localized radiation (SBRT) and FOLFIRINOX

Ongoing

PC: Pancreatic Cancer; FOLFIRINOX: 5-Fluorouracil, Leucovorin, Irinotecan and Oxaliplatin; GM-CSF: Granulocyte-Macrophage Colony-Stimulating Factor; DCs: Dendritic Cell; CEA: Carcinoembryonic Antigen; MUC-1: Mucin 1; LAK: Lymphokine Activated Killer; hTERT: Telomerase Teverse Transcriptase; Her2/neu: Human Epidermal Growth Factor Receptor 2; VeGFrs: Vascular Endothelial Growth Factor Receptors; WT1: Wilms Tumor Gene; HSP: Heat Shock Protein

Peptide vaccines

Peptide-based cancer vaccines are preparations made from antigenic protein fragments (called epitopes), that represent the minimal immunogenic region of antigens [41,42], designed to enhance the T cell response, especially the CD8+ . Induction of CTLs needs peptides derived from TAAs to be presented on the surface of APCs (antigens presenting cells), such as DCs, in the context of HLA molecules.

KRAS vaccines: The association of mutant Kras with pancreatic cancer was established two decades ago [43,44]. The most common activating point mutation involves the KRAS2 oncogene, on chromosome 12p, in over 90% of PC [45]. This is the highest fraction of K-ras alteration found in any human tumor type. Recent tumor genome sequencing studies have established the prevalence of mutant Kras in Pancreatic Intraepithelial Neoplasia (PanINs), the most common precursor lesions and in pancreatic cancer with increased precision [46-48].In a Phase I/II study, the administration of synthetic KRAS-derived peptides to un resectable pancreatic cancer patients resulted in an immune response in 2 out of 5 individuals [49]. Since native epitopes have relatively low immunogenicity, granulocyte-macrophage colonystimulating factor (GM-CSF) was applied to achieve efficient vaccination in the study. Among 48 patients with pancreatic cancer (10 surgically resected and 38 with advanced disease), vaccination of mutant K-ras peptides in combination with GMCSF resulted in immune responses and prolonged survival [50]. Moreover, another group also reported that vaccination of 24 patients with resected pancreatic cancer with K-ras peptide in combination with GM-CSF proved to be safe without tumor regression and Median recurrence-free survival time was 8.6 mo and median overall survival time was 20.3 mo [51].In one clinical study vaccination with mutated K-ras resulted in 20% long term survivors [52]. A randomized phase II placebo controlled trial using recombinant mutated K-ras protein for vaccination in combination with gemcitabine, in patients with resected pancreatic cancer, is currently ongoing [53]. Lisiansky selectively kill Ras-transformed cells by over expressing the pro-apoptotic protein, p53 upregulated modulator of apoptosis (PUMA) under a Ras-responsive promoter, and assess it may become a useful, effective and safe approach to selectively target Ras-mutated tumor cells [54].

Survivin vaccines: Survivin is a member of the inhibitor apoptosis family, which is highly upregulated in most malignancies, including pancreatic cancer [55]. In a study, murine pancreatic and lymphoma models, survivin DNA vaccine showed significant slow tumor growth and longer survival compared with those vaccinated with vector DNA [56]. In a study, a survivin-derived peptide (AYACNTSTL) was used in combination with IFN α to vaccinate six patients who had advanced pancreatic cancers. Tetramer and enzyme-linked immunosorbent spot (ELISPOT) assays revealed that more than half of the patients had manifested immunological responses to vaccination, which were often accompanied by clinical benefits [57]. However, this vaccine still need to be tested in a large population.

Gastrin vaccines: Gastrin and cholecystokinin B receptor (CCKBR, also known as CCK-2) are upregulated and co-expressed in both pancreatic cell lines and human PDA specimens and have been implicated in autocrine, paracrine, and endocrine growth pathway [58]. In a randomized, double-blind, placebo-controlled, group-sequential multicenter trial of G17DT in patients with advanced pancreatic cancer unsuitable for or unwilling to take chemotherapy, resulted in a nearly 2-fold increase in median overall survival, as compared with placebo (151 vs. 82 d, respectively; p = 0.03). Anti-gastrin immune responses were noted in 73.8% of the patients and correlated with longer overall median survival versus non responders and placebo (176 vs. 63 vs. 83 days respectively, p = 0.003) [59]. Gastrin-based vaccines appear therefore to be well tolerated by and could represent a new therapeutic option for pancreatic cancer.

HSP-peptide complex vaccines: Heat Shock Protein (HSP), a component of HSP–Peptide Complex (PC), works as a peptide chaperone for stabilizing and delivering peptides. HSP-peptide complexes can be presented on MHC class I molecules on the cell surface. Tumor-derived HSP-peptide complexes have been shown to induce antitumor immune responses in preclinical studies. HSP96-peptides complexes produced from resected tumor tissues were the first to be employed in anticancer vaccines. A phase I pilot trial of patients with resected pancreatic cancer who received no adjuvant radiation or chemotherapy showed feasibility of preparing HSPCC-96 from the resected tumor. A total of 10 patients were vaccinated with 5 μg of autologous HSPPC-96 weekly for four doses. No dose limiting toxicities were observed. There was no correlation between survival and immune response, exhibiting a median overall survival of 2.2 years. Three of 10 patients were alive without disease at 2.6, 2.7 and 5-years follow up [60]. This study showed that vaccine preparation from resected tumor and administration were feasible. Further studies need to evaluate the clinical efficacy of HSP vaccines in patients with pancreatic cancer.

WT1, cancer-testis antigens (CT) and VEGFRs vaccines: Wilms Tumor gene (WT1) protein is an attractive target for cancer immunotherapy, in a study of 32 patients with advanced pancreatic cancer was treated with WT1 vaccine in combination with gemcitabine and was well tolerated. The association between longer survival and positive delayed-type hypersensitivity to WT1 peptide was statistically significant, and longer survivors featured a higher frequency of memory-phenotype WT1-specific cytotoxic T lymphocytes both before and after treatment [61]. Median survival time and 1-year survival rate were 8.1 months and 29% respectively.

The cancer-testis (CT) antigens are expressed by tumors of different histological types at varying frequencies (10-40%) [62]. CT antigens are absent in normal somatic cells in humans and rodents but expressed only in male germ cells (such as spermatogonial stem cells, spermatogonia, spermatocytes, spermatids and spermatozoa) during spermatogenesis in the testis (but not Sertoli and/or Leydig cells) [63-68]. Fifty two percentage of the analyzed pancreatic cancer tissue expressed at least one CT antigen [69]. Recently, in a Phase I clinical trial for advanced pancreatic cancer to investigate the safety, immunostimulatory effects, and antineoplastic activity of a multi-target vaccine composed of four distinct peptides derived from cancer-testis (CT) antigens and vascular endothelial growth factor receptors (VeGFrs) was well-tolerated, and no grade 3 or 4 adverse was observed [70]. The median overall survival (OS) of this cohort was 207 days.

Two vascular endothelial growth factor receptors (VeGFrs) peptide vaccine studies using VEGF-R1 and VEGF-R2 are completed but have not reported results yet [71].

Telomerase reverse transcriptase (hTERT) and Her2/ neu vaccines: Telomerase reverse transcriptase (hTERT) is a highly immunogenic antigen and has been the target in several vaccination studies. A small phase I/II study in patients with pancreatic cancer showed T cell responses in 63% of vaccinated patients, and prolonged survival in patients exhibiting T cell responses [72]. However, a large phase III study failed to show a survival benefit in pancreatic cancer [73]. Another study is evaluating radiation therapy, tadalafil, sargramostim, gemcitabine and telomerase vaccine (GV1001) in patients with unresectable pancreatic cancer and is currently active and waiting for result [74].Recently, vaccination against Her2/neu is also being tested in a phase I study in patients with pancreatic cancer [75].

Recombinant vaccines

Recombinant vaccines contain bacterial and viral antigen carriers thereby increasing DC activation and improving antigen presentation. They stimulate the innate immune response while efficiently recruiting and activating DCs. The most common carriers include Bacille Calmette-Guerin (BCG), Listeria Monocytogenes (LM), Salmonella and Poxviruses.

Carcinoembryonic antigen (CEA) and mucin 1 (MUC-1): The Carcinoembryonic Antigen (CEA) is an oncofetal antigen that is expressed highly in the majority of pancreatic cancers [76] and MUC-1 is a membrane bound glycoprotein known to promote pancreatic cancer epithelial to mesenchymal transition and invasiveness. It also induces CD8 T cell responses and the production of anti-MUC antibodies is associated with improved survival [77]. TRICOM is a poxvirus-based vaccine containing a combination of three T-cell costimulatory molecules: B7- 1, intercellular adhesion molecule 1 (ICAM-1) and leucocyte function associated antigen 3 (LFA-3). In a phase I study of TRICOM with CEA vaccine was conducted in 58 patients with metastatic pancreatic cancer by Marshall and colleagues. In this study, CEA-TRICOM vaccine was used with or without GM-CSF and only one patient had pancreatic cancer. This patient had progressed with increasing pain and CA 19-9 on previous CEA vaccination. After two vaccinations with CEA-TRICOM, CA 19-9 and pain levels decreased for almost 1 year [78]. This study showed that CEA-TRICOM vaccines were safe and generated significant CEA-specific immune response with associated clinical benefit. In a large randomized phase-III clinical trial of 255 patients vaccination with a MUC-1 and CEA expressing viral vector showed no overall survival benefit [79].

Listeria vaccines: Listeria monocytogenes (Lm)-based vaccines stimulate both innate and adaptive immunity. In one recent study, patients bearing hepatic metastases from mesothelioma, ovarian cancer, non-small cell lung carcinoma and PC were given this L. monocytogenes strain further engineered to express mesothelin, a cell surface molecule overexpressed by a large majority of PC, mesotheliomas, ovarian cancers, and non-small cell lung carcinomas [80].Thirty-seven percent of these patients survived 15 months or more. Half of them patients were those harboring metastatic PC, and immunological analyses revealed that they had developed listeriolysin O- and mesothelinspecific T-cell responses. While a mesothelin vaccine, using genetically modified live attenuated listeria as a vector for the antigen, has also entered clinical trial [81].

Antigen pulsed Dendritic cell (DCs) vaccines

DCs are the most potent Antigen Presenting Cells (APCs) that are capable of priming naïve T cells and can stimulate memory T cells and B cells to generate antigen specific response. Antigen pulsed DCs is another vaccination strategy where patient DCs are isolated, pulsed with peptides, autologous, or allogeneic tumor lysate, or transfected with RNA, and injected back to the patients. In a study of 12 patients (10 with pancreatic cancer), where MUC1 pulsed DCs were given as adjuvant therapy following resection, 4 of the 12 patients were alive at 4 years [82]. In the second study, a DC-based vaccine alone or combined with Lymphokine activated killer (LAK) cells was administered together with gemcitabine and/or S-1 to 49 patients with advanced pancreatic adenocarcinoma [83]. Median survival in these patients was 360 days. Patients receiving DC vaccine along with chemotherapy and LAK cell therapy had prolonged survival compared with patients who received DC vaccine and chemotherapy. Of all 49 patients, 2 had complete remission, 5 had partial remission and 10 had stable disease. Thus the study concluded that DC-based vaccine therapy with chemotherapy was shown to be safe and may induce responses.

Whole tumor cell vaccines

Whole tumor cell vaccines are another strategy that has shown promise in pancreatic cancer. Autologous whole tumor cells remain a potent vehicle for generating antitumor immunity. This is because tumor cells express all relevant candidate TAAs, including both known and unidentified. Two allogeneic whole cell based anticancer vaccines are currently being investigated for their safety and antineoplastic effects in PC patients.

Algenpantucel-L: Algenpantucel-L is the most clinically advanced and promising immunotherapy for pancreatic cancer. Algenpantucel-L (also known as hyperacute-pancreatic cancer vaccine) consists an 2 irradiated, live, human allogeneic pancreatic cancer cell lines that express murine α-1,3- galactosyltransferase, which is responsible for the synthesis of α-galactosylated epitopes on cell surface proteins. Hardacre and colleagues presented the results of an open-label, multiinstitutional Phase II clinical trial investigating Algenpantucel-L in combination with standard adjuvant chemoradiotherapy for the treatment of resected PC patients [84]. In this study 69 out of 73 patients were evaluable and they received 100 million cells (N= 43) or 300 million cells (N= 26) injected intradermally in up to 14 vaccinations. No serious adverse events were attributed to the immunotherapy. After a median follow-up of 21 months, the 12-month disease-free survival was 62 %, and the 12-month overall survival was 86 %. The most common adverse events were injection site pain and induration. The study concluded that the addition of algenpantucel-L to standard adjuvant therapy for resected pancreatic cancer may improve survival. A multi-institutional, phase 3 study is ongoing (ClinicalTrials.gov identifier, NCT01072981) [85].

Allogeneic GM-CSF vaccine therapy: Allogeneic Granulocyte–macrophage colony-stimulating factor (GM-CSF) vaccine therapy, called GVAX, has been tested in a variety of early phase clinical trials. In a phase I clinical study, tumor cells, which were modified to express the immunomodulating cytokine GMCSF, where given to 14 patients [86]. Three patients had delayedtype hypersensitivity responses to autologous tumor cells and those 3 patients had a longer disease free survival. Latter in In a phase-II study with a similar approach, 60 patients with resected pancreatic cancer were treated, yielding a disease free survival of 17.3 months and an overall survival of 24.8 months [87]. While the results did not superior result to previous study, other studies using similar approaches, or combining whole tumor vaccination with cyclophosphamide alone, or with conventional chemotherapy, are ongoing [88-91].

PERSPECTIVES

Although vaccine therapy as a single agent has encouraging results, clinical trials in PC patients have been underwhelming and disappointing. Most of these clinical studies identified a number of critical aspects that must be carefully considered for the design the next generation of cancer vaccines. As stated earlier dysfunctional immune system in PC further give rise to tumor induced immunosuppressive environment has a critical role for the development and progression of tumor. Which consist of not only cancer cells but also immune suppressive cells (CAFs, tolerogenic DCs, MDSCs, TAMs and Treg cells) and cancer cells-derived soluble factors that promote the induction of tolerance. So, novel vaccine therapy strategy should be designed for breaking immunosuppression within the tumor microenvironment, to inhibit immunologic checkpoint blockade and to modulate tumor microenvironment. Thus, this may require a combinatorial therapeutic approach which includes chemotherapy, radiation, surgery and immunotherapy.

REFERENCES

1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013; 63: 11-30.

2. Chen W, Zheng R, Zhang S, Zhao P, Li G, Wu L, et al. The incidences and mortalities of major cancers in China, 2009. Chin J Cancer. 2013; 32: 106-112.

3. Wellner UF, Makowiec F, Bausch D, Höppner J, Sick O, Hopt UT, et al. Locally advanced pancreatic head cancer: margin-positive resection or bypass? ISRN Surg. 2012; 2012: 513241.

4. Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011; 150: 466-473.

5. Shrikhande SV, Kleeff J, Reiser C, Weitz J, Hinz U, Esposito I, et al. Pancreatic resection for M1 pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2007; 14: 118-127.

6. Bose D, Katz MHG, Fleming JB. Pancreatic Adenocarcinoma. Feig BW, Ching CD, editors. In: The MD Anderson Surgical Oncology Hand book. 5th edn. Philadelphia: Lippincott Williams and Wilkins. 2012; 472–490.

7. Páez D, Labonte MJ, Lenz HJ. Pancreatic cancer: medical management (novel chemotherapeutics). Gastroenterol Clin North Am. 2012; 41: 189-209.

8. Iacobuzio-Donahue CA, Fu B, Yachida S, Luo M, Abe H, Henderson CM, et al. DPC4 Gene Status of the Primary Carcinoma Correlates with Patterns of Failure in Patients with Pancreatic Cancer. J Clin Oncol. 2009; 27: 1806-1813.

9. Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. Groupe Tumeurs Digestives of Unicancer; PRODIGE Intergroup.: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011; 364: 1817-1825.

10. Diaz Beveridge R, Alcolea V, Aparicio J, Segura Á, García J, Corbellas M, et al. Management of advanced pancreatic cancer with gemcitabine plus erlotinib: efficacy and safety results in clinical practice. JOP. 2014; 15: 19-24.

11. Yokokawa J, Palena C, Arlen P, Hassan R, Ho M, Pastan I, et al. Identification of novel human CTL epitopes and their agonist epitopes of mesothelin. Clin Cancer Res. 2005; 11: 6342-6351.

12. Johnston FM, Tan MC, Tan BR Jr, Porembka MR, Brunt EM, Linehan DC, et al. Circulating mesothelin protein and cellular antimesothelin immunity in patients with pancreatic cancer. Clinical Cancer Research. 2009; 15: 6511–6518.

13. Arce F, Kochan G, Breckpot K, Stephenson H, Escors D. Selective activation of intracellular signalling pathways in dendritic cells for cancer immunotherapy. Anticancer Agents Med Chem. 2012; 12: 29- 39.

14. Ghiringhelli F, Bruchard M, Apetoh L. Immune effects of 5-fluorouracil: Ambivalence matters. Oncoimmunology. 2013; 2: e23139.

15. Ghiringhelli F, Apetoh L. Chemotherapy and immunomodulation: from immunogenic chemotherapies to novel therapeutic strategies. Future Oncol. 2013; 9: 469-472.

16. Whatcott CJ, Posner RG, Von Hoff DD, Han H.; Desmoplasia and chemoresistance in pancreatic cancer. In: Grippo PJ, Munshi HG, editors. Pancreatic Cancer and Tumor Microenvironment. Trivandrum (India): Transworld Research Network; 2012. Chapter 8.

17. De Palma M, Lewis CE. Cancer: Macrophages limit chemotherapy. Nature. 2011; 472: 303-304.

18. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013; 73: 1128–1141.

19. Kubuschok B, Neumann F, Breit R, Sester M, Schormann C, Wagner C, et al. Naturally occurring T-cell response against mutated p21 Ras oncoprotein in pancreatic cancer. Clinical Cancer Research. 2006; 12: 1365–1372.

20. Yanagimoto H, Mine T, Yamamoto K, Satoi S, Terakawa N, Takahashi K, et al. Immunological evaluation of personalized peptide vaccination with gemcitabine for pancreatic cancer. Cancer Science. 2007; 98: 605–611.

21. Wenandy L, Sørensen RB, Sengeløv L, Svane IM, thor Straten P, Andersen MH. The immunogenicity of the hTERT540-548 peptide in cancer. Clin Cancer Res. 2008; 14: 4-7.

22. 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: 148-154.

23. Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, et al. Overexpression of the Wilms’ tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci. 2004; 95: 583-587.

24. Ueda M, Miura Y, Kunihiro O, Ishikawa T, Ichikawa Y, Endo I, et al. MUC1 overexpression is the most reliable marker of invasive carcinoma in intraductal papillary-mucinous tumor (IPMT). Hepatogastroenterology. 2005; 52: 398-403.

25. Wobser M, Keikavoussi P, Kunzmann V, Weininger M, Andersen MH, Becker JC. “Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLAA2 restricted peptide derived Central Lu et al. (2014) Email: J Cancer Biol Res 2(1): 1046 (2014) 8/9 from the universal tumor antigen surviving. Cancer Immunology, Immunotherapy. 2006; 55: 1294–1298.

26. Seki K, Suda T, Aoyagi Y, Sugawara S, Natsui M, Motoyama H, et al. Diagnosis of pancreatic adenocarcinoma by detection of human telomerase reverse transcriptase messenger RNA in pancreatic juice with sample qualification. Clinical Cancer Research. 2001; 7: 1976– 1981.

27. Komoto M, Nakata B, Amano R, Yamada N, Yashiro M, Ohira M, et al. HER2 overexpression correlates with survival after curative resection of pancreatic cancer. Cancer Sci. 2009; 100: 1243-1247.

28. Maacke H, Kessler A, Schmiegel W, Roeder C, Vogel I, Deppert W, et al. Overexpression of p53 protein during pancreatitis. Br J Cancer. 1997; 75: 1501-1504.

29. Gjertsen MK, Bakka A, Breivik J, Saeterdal I, Solheim BG, Søreide O, et al. Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet. 1995; 346: 1399-1400.

30. Cappello P, Tomaino B, Chiarle R, Ceruti P, Novarino A, Castagnoli C, et al. An integrated humoral and cellular response is elicited in pancreatic cancer by??-enolase, a novel pancreatic ductal adenocarcinoma associated antigen. International Journal of Cancer. 2009; 125: 639– 648.

31. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008; 454: 436-444.

32. 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.

33. Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death and Differentiation. 2014; 2: 15–25.

34. Sideras K, Braat H, Kwekkeboom J, van Eijck CH, Peppelenbosch MP, Sleijfer S, et al. Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies. Cancer Treat Rev. 2014; 40: 513-522.

35. Koido S, Homma S, Hara E, Namiki Y, Takahara A, Komita H, et al. Regulation of tumor immunity by tumor/dendritic cell fusions. Clin Dev Immunol. 2010; 2010: 516768.

36. Koido S, Homma S, Hara E, Mitsunaga M, Namiki Y, Takahara A, et al. In vitro generation of cytotoxic and regulatory T cells by fusions of human dendritic cells and hepatocellular carcinoma cells. J Transl Med. 2008; 6: 51.

37. Shakhar G, Ben-Eliyahu S. Potential prophylactic measures against postoperative immunosuppression: could they reduce recurrence rates in oncological patients? Annals of Surgical Oncology. 2003; 10: 972–992.

38. Palena C, Abrams SI, Schlom J, Hodge JW. Cancer vaccines: preclinical studies and novel strategies. Adv Cancer Res. 2006; 95: 115-145.

39. Palena C, Abrams SI, Schlom J, Hodge JW. Cancer vaccines: preclinical studies and novel strategies. Adv Cancer Res. 2006; 95: 115-145.

40. Pejawar-Gaddy S, Finn OJ. Cancer vaccines: accomplishments and challenges. Crit Rev Oncol Hematol. 2008; 67: 93-102.

41. Purcell AW, McCluskey J, Rossjohn J. More than one reason to rethink the use of peptides in vaccine design. Nat Rev Drug Discov. 2007; 6: 404-414.

42. Bijker MS, Melief CJ, Offringa R, van der Burg SH. Design and development of synthetic peptide vaccines: past, present and future. Expert Rev Vaccines. 2007; 6: 591-603.

43. Smit VT, Boot AJ, Smits AM, Fleuren GJ, Cornelisse CJ, Bos JL. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res. 1988; 16: 7773-7782.

44. Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-Kras genes. Cell. 1988; 53: 549-554.

45. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011; 11: 761-774.

46. Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012; 491: 399-405.

47. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008; 32: 1801–1806.

48. Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012; 142: 730–733.

49. Gjertsen MK, Bakka A, Breivik J, Saeterdal I, Solheim BG, Søreide O, et al. Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet. 1995; 346: 1399-1400.

50. Gjertsen MK, Buanes T, Rosseland AR, Bakka A, Gladhaug I, Sqreide O, et al. Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: Clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer. 2001; 92: 441-450.

51. Abou-Alfa GK, Chapman PB, Feilchenfeldt J, Brennan MF, Capanu M, Gansukh B, et al. Targeting mutated K-ras in pancreatic adenocarcinoma using an adjuvant vaccine. Am J Clin Oncol. 2011; 34: 321-325.

52. Wedén S, Klemp M, Gladhaug IP, Møller M, Eriksen JA, Gaudernack G, et al. Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int J Cancer. 2011; 128: 1120-1128.

53. Clinical Trials.gov Identifier: NCT00300950.

54. Lisiansky V, Naumov I, Shapira S, Kazanov D, Starr A, Arber N, et al. Gene therapy of pancreatic cancer targeting the K-Ras oncogene. Cancer Gene Ther. 2012; 19: 862-869.

55. Satoh K, Kaneko K, Hirota M, Masamune A, Satoh A, Shimosegawa T. Expression of survivin is correlated with cancer cell apoptosis and is involved in the development of human pancreatic duct cell tumors. Cancer. 2001; 92: 271-278.

56. Zhu K, Qin H, Cha SC, Neelapu SS, Overwijk W, Lizee GA, et al. Survivin DNA vaccine generated specific antitumor effects in pancreatic carcinoma and lymphoma mouse models. Vaccine. 2007; 25: 7955– 7961.

57. Kameshima H, Tsuruma T, Kutomi G, Shima H, Iwayama Y, Kimura Y, et al. Immunotherapeutic benefit of a-interferon (IFNa) in survivin2Bderived peptide vaccination for advanced pancreatic cancer patients. Cancer Sci. 2013; 104: 124-129.

58. Monstein HJ, Ohlsson B, Axelson J. Differential expression of gastrin, cholecystokinin-A and cholecystokinin-B receptor mRNA in human pancreatic cancer cell lines. Scand J Gastroenterol. 2001; 36: 738-743.

59. Gilliam AD, Broome P, Topuzov EG, Garin AM, Pulay I, Humphreys J, et al. An international multicenter randomized controlled trial of G17DT in patients with pancreatic cancer. Pancreas. 2012; 41: 374–379. Central Lu et al. (2014) Email: J Cancer Biol Res 2(1): 1046 (2014) 9/9

60. Maki RG, Livingston PO, Lewis JJ, Janetzki S, Klimstra D, Desantis D, et al. A phase I pilot study of autologous heat shock protein vaccine HSPPC-96 in patients with resected pancreatic adenocarcinoma. Dig Dis Sci. 2007; 52: 1964–1972.

61. Nishida S, Koido S, Takeda Y, Homma S, Komita H, Takahara A, et al. Wilms Tumor Gene (WT1) Peptide-based Cancer Vaccine Combined with Gemcitabine for Patients with Advanced Pancreatic Cancer. J Immunotherapy. 2014; 37: 105-114.

62. Van den Eynde BJ, Boon T. Tumor antigens recognized by T lymphocytes. Int J Clin Lab Res. 1997; 27: 81-86.

63. Scanlan MJ, Gure AO, Jungbluth AA, Old LJ, Chen YT. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev. 2002; 188: 22-32.

64. Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 2005; 5: 615- 625.

65. Mirandola L, J Cannon M, Cobos E, Bernardini G, Jenkins MR, Kast WM, et al. Cancer testis antigens: novel biomarkers and targetable proteins for ovarian cancer. Int Rev Immunol. 2011; 30: 127-137.

66. Cebon J. Cancer vaccines: Where are we going? Asia Pac J Clin Oncol. 2010; 6 Suppl 1: S9-15.

67. Akers SN, Odunsi K, Karpf AR. Regulation of cancer germline antigen gene expression: implications for cancer immunotherapy. Future Oncol. 2010; 6: 717-732.

68. Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 2009; 100: 2014-2021.

69. Schmitz-Winnenthal FH, Galindo-Escobedo LV, Rimoldi D, Geng W, Romero P, Koch M, et al. Potential target antigens for immunotherapy in human pancreatic cancer. Cancer Lett. 2007; 252: 290-298.

70. Okuyama R, Aruga A, Hatori T, Takeda K, Yamamoto M. Immunological responses to a multi-peptide vaccine targeting cancer-testis antigens and VEGFRs in advanced pancreatic cancer patients. OncoImmunology. 2013; 2: e27010.

71. ClinicalTrials.gov. Identifier: NCT00655785. 72.Bernhardt SL, Gjertsen MK, Trachsel S, Møller M, Eriksen JA, Meo M, et al. Telomerase peptide vaccination of patients with nonresectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer. 2006; 95: 1474–1482.

73. Buanes T, Maurel J, Liauw W, Hebbar M, Nemunaitis J. A randomized phase III study of gemcitabine (G) versus GV1001 in sequential combination with G in patients with unresectable and metastatic pancreatic cancer (PC). ASCO Annu Conf. 2009; 27: 4601.

74. ClinicalTrials.gov Identifier: NCT01342224.

75. ClinicalTrials.gov Identifier: NCT01526473.

76. Gunturu KS, Rossi GR, Saif MW. Immunotherapy Updates in pancreatic Cancer: are we there yet?. Ther Adv Med Oncol. 2013; 5: 81-89.

77. Hamanaka Y, Suehiro Y, Fukui M, Shikichi K, Imai K, Hinoda Y. Circulating anti-MUC1 IgG antibodies as a favorable prognostic factor for pancreatic cancer. Int J Cancer. 2003; 103: 97-100.

78. Marshall JL, Gulley JL, Arlen PM, Beetham PK, Tsang KY, Slack R, et al. Phase I study of sequential vaccinations with fowlpoxCEA(6D)- TRICOM alone and sequentially with vaccinia-CEA(6D)-TRICOM, with and without granulocyte-macrophage colony-stimulating factor, in patients with carcinoembryonic antigen-expressing carcinomas. J Clin Oncol. 2005; 23: 720–731.

79. Arlen PM, Gulley JL, Madan RA, Hodge JW, Schlom J. Preclinical and clinical studies of recombinant poxvirus vaccines for carcinoma therapy. Crit Rev Immunol. 2007; 27: 451-462.

80. Le DT, Brockstedt DG, Nir-Paz R, Hampl J, Mathur S, Nemunaitis J, et al. A live-attenuated Listeria vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction. Clin Cancer Res. 2012; 18: 858-868.

81. ClinicalTrials.gov. Identifier: NCT00655785.

82. Lepisto AJ, Moser AJ, Zeh H, Lee K, Bartlett D, McKolanis JR, et al. A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther. 2008; 6(B): 955–964.

83. Kimura Y, Tsukada J, Tomoda T, Takahashi H, Imai K, Shimamura K, et al. Clinical and immunologic evaluation of dendritic cell-based immunotherapy in combination with gemcitabine and/or S-1 in patients with advanced pancreatic carcinoma. Pancreas. 2012; 41: 195-205.

84. Hardacre JM, Mulcahy M, Small W, Talamonti M, Obel J, Krishnamurthi S, Rocha-Lima CS. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a phase 2 study. J Gastrointest Surg. 2013; 17: 94-100.

85. ClinicalTrials.gov. Identifier: NCT01072981.

86. Jaffee EM, Hruban RH, Biedrzycki B, Laheru D, Schepers K, Sauter PR, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factorsecreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Oncol. 2001; 19: 145-156.

87. Lutz E, Yeo CJ, Lillemoe KD, Biedrzycki B, Kobrin B, Herman J, et al. A lethally irradiated allogeneic granulocyte–macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A phase II trial of safety, efficacy, and immune activation. Ann Surg. 2011; 253: 328–335.

88. ClinicalTrials.gov Identifier: NCT00305760.

89. ClinicalTrials.gov Identifier: NCT00389610.

90. ClinicalTrials.gov Identifier: NCT00727441.

91. ClinicalTrials.gov Identifier: NCT01595321.

Yadav DK, Lu CD, Rajesh Kumar Y (2014) Vaccine Therapy for Pancreatic Cancer: As a Novel Therapeutic Approach? J Cancer Biol Res 2(1): 1046.

Received : 15 Apr 2014
Accepted : 12 May 2014
Published : 20 May 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X