Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis

  • Authors:
    • Munehiro Yoshitomi
    • Shigeru Yutani
    • Satoko Matsueda
    • Tetsuya Ioji
    • Nobukazu Komatsu
    • Shigeki Shichijo
    • Akira Yamada
    • Kyogo Itoh
    • Tetsuro Sasada
    • Hisafumi Kinoshita
  • View Affiliations

  • Published online on: December 20, 2011     https://doi.org/10.3892/etm.2011.424
  • Pages: 463-469
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Considering that the prognosis of patients with advanced biliary tract cancer (BTC) remains very poor, with a median survival of less than 1 year, new therapeutic approaches need to be developed. In the present study, a phase II clinical trial of personalized peptide vaccination (PPV) was conducted in advanced BTC patients to evaluate the feasibility of this treatment and to identify potential biomarkers. A maximum of 4 human leukocyte antigen-matched peptides, which were selected based on the pre-existing host immunity prior to vaccination, were subcutaneously administered (weekly for 6 consecutive weeks and bi-weekly thereafter) to 25 advanced BTC patients without severe adverse events. Humoral and/­or T cell responses specific to the vaccine antigens were substantially induced in a subset of the vaccinated patients. As shown by multivariate Cox regression analysis, lower interleukin-6 (IL-6) and higher albumin levels prior to vaccination and greater numbers of selected vaccine peptides were significantly favorable factors for overall survival [hazard ratio (HR)=1.123, 95% confidence interval (CI) 1.008‑1.252, P=0.035; HR=0.158, 95% CI 0.029-0.860, P=0.033; HR=0.258, 95% CI 0.098-0.682, P=0.006; respectively]. Based on the safety profile and substantial immune responses to vaccine antigens, PPV could be a promising approach for refractory BTC, although its clinical efficacy remains to be investigated in larger-scale prospective studies. The identified biomarkers are potentially useful for selecting BTC patients who would benefit from PPV.

Introduction

Biliary tract cancer (BTC) is one of the most aggressive types of cancer and has a very poor prognosis (1,2). Only 10% of newly diagnosed patients present with early-stage disease, which may be treated by a potentially radical excision of the tumor, and the remaining patients have unresectable disease with locally advanced and/or metastatic tumors. Recently, there have been substantial advances in treatment modalities, including systemic chemotherapies, for advanced BTC (14). For example, a randomized trial has suggested that cisplatin plus gemcitabine could be considered as a standard treatment option for patients with advanced BTC (3). In addition, a number of different targeted therapies for BTC have also been under investigation (14). Despite this progress, however, the prognosis of BTC patients remains very poor, with a median survival of less than 1 year. Therefore, further novel therapeutic approaches need to be developed.

We previously devised a new regime of peptide-based vaccination, known as ‘personalized peptide vaccination (PPV)’, in which vaccine antigens are selected and administered based on the pre-existing host immunity prior to vaccination (57). We reported favorable clinical and/or immune responses of this novel vaccination in various types of advanced cancer, including pancreatic, gastric, colorectal and prostate cancer, and glioblastoma (812). For example, a recently conducted randomized clinical trial of PPV for advanced prostate cancer patients showed a promising clinical outcome in the vaccinated group (11). In the present study, we addressed the feasibility of using PPV in advanced BTC patients in a small-scale phase II study. In addition, we identified potential biomarkers for predicting overall survival (OS) and selecting suitable patients for this treatment.

Patients and methods

Patients

Patients were eligible for inclusion in the present study if they had a histological diagnosis of BTC and showed positive humoral responses to at least two of the 31 different vaccine candidate peptides (Table I). Other inclusion criteria were as follows: age between 20 and 80 years; an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1; positive status for human leukocyte antigen (HLA)-A2, -A24, -A3 supertype (A3, A11, A31 or A33), or -A26; life expectancy of at least 12 weeks; negative status for hepatitis B and C virus; and adequate hematological, hepatic and renal function. Exclusion criteria included pulmonary, cardiac or other systemic diseases; an acute infection; a history of severe allergic reactions; pregnancy or nursing; and other inappropriate conditions for enrollment as judged by clinicians. The protocol was approved by the Kurume University Ethics Committee, and was registered in the UMIN Clinical Trials Registry (UMIN 2907). Following a full explanation of the protocol, written informed consent was obtained from all patients prior to enrollment.

Table I.

Peptide candidates for cancer vaccination.

Table I.

Peptide candidates for cancer vaccination.

Symbol for peptideOrigin proteinPosition of peptideAmino acid sequenceHLA type
CypB-129Cyclophilin B129–138KLKHYGPGWVA2, A3supa
Lck-246p56 lck246–254KLVERLGAAA2
Lck-422p56 lck422–430DVWSFGILLA2, A3sup
MAP-432ppMAPkkk432–440DLLSHAFFAA2, A26
WHSC2-103WHSC2103–111ASLDSDPWVA2, A3supa, A26
HNRPL-501HNRPL501–510NVLHFFNAPLA2, A26
UBE-43UBE2V43–51RLQEWCSVIA2
UBE-85UBE2V85–93LIADFLSGLA2
WHSC2-141WHSC2141–149ILGELREKVA2
HNRPL-140HNRPL140–148ALVEFEDVLA2
SART3-302SART3302–310LLQAEAPRLA2
SART3-309SART3309–317RLAEYQAYIA2
SART2-93SART293–101DYSARWNEIA24
SART3-109SART3109–118VYDYNCHVDLA24, A3supa, A26
Lck-208p56 lck208–216HYTNASDGLA24
PAP-213PAP213–221LYCESVHNFA24
PSA-248PSA248–257HYRKWIKDTIA24
EGFR-800EGF-R800–809DYVREHKDNIA24
MRP3-503MRP3503–511LYAWEPSFLA24
MRP3-1293MRP31293–1302NYSVRYRPGLA24
SART2-161SART2161–169AYDFLYNYLA24
Lck-486p56 lck486–494TFDYLRSVLA24
Lck-488p56 lck488–497DYLRSVLEDFA24
PSMA-624PSMA624–632TYSVSFDSLA24
EZH2-735EZH2735–743KYVGIEREMA24
PTHrP-102PTHrP102–111RYLTQETNKVA24
SART3-511SART3511–519WLEYYNLERA3supa
SART3-734SART3734–742QIRPIFSNRA3supa
Lck-90p56 lck90–99ILEQSGEWWKA3supa
Lck-449p56 lck449–458VIQNLERGYRA3supa
PAP-248PAP248–257GIHKQKEKSRA3supa

a A3sup, HLA-A3 supertype (A3, A11, A31 and A33). HLA, human leukocyte antigen.

Clinical protocol

This was an open-label phase II study, in which the primary and secondary end-points were to identify biomarkers for OS and to evaluate the safety of PPV in BTC patients, respectively. In this study, 31 peptides, whose safety and immunological effects had been confirmed in previously conducted clinical studies (612), were employed for vaccination [12 peptides for HLA-A2, 14 peptides for HLA-A24, 9 peptides for the HLA-A3 supertype (A3, A11, A31 or A33) and 4 peptides for HLA-A26] (Table I). The peptides were prepared under the conditions of Good Manufacturing Practice (GMP) by the PolyPeptide Laboratories (San Diego, CA, USA) and the American Peptide Company (Vista, CA, USA). The right peptides for vaccination to individual patients were selected, taking into consideration the pre-existing host immunity prior to vaccination, assessed by titers of IgG specific to each of the 31 different vaccine candidates, as reported previously (612). A maximum of 4 peptides (3 mg/each peptide), which were selected based on the results of HLA typing and peptide-specific IgG titers, were subcutaneously administered with incomplete Freund’s adjuvant (Montanide ISA51; Seppic, Paris, France) once a week for 6 consecutive weeks. After the first cycle of 6 vaccinations, up to 4 antigen peptides, which were reselected according to the titers of peptide-specific IgG at every cycle of 6 vaccinations, were administered every 2 weeks. Adverse events were monitored according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 3.0 (NCI-CTC Ver. 3.0). Complete blood counts and serum biochemistry tests were performed after every 6 vaccinations. The clinical responses were evaluated by the Response Evaluation Criteria in Solid Tumors (RECIST) in the vaccinated patients, whose radiological findings by computed tomography (CT) scan or magnetic resonance imaging (MRI) were available prior to and following vaccinations.

Measurement of humoral and T cell responses specific to the vaccine peptides

The humoral responses specific to the vaccine peptides were determined by peptide-specific IgG titers using a bead-based multiplex assay with the Luminex 200 system (Luminex, Austin, TX, USA), as reported previously (13). If peptide-specific IgG titers to at least one of the vaccine peptides in the post-vaccination plasma were more than 2-fold higher than those in the pre-vaccination plasma, the changes were considered to be significant.

T cell responses specific to the vaccine peptides were evaluated by interferon (IFN)-γ ELISPOT assay (MBL, Nagoya, Japan) using peripheral blood mononuclear cells (PBMCs). Briefly, PBMCs (2.5x104 cells/well) were incubated in 384-well microculture plates (Iwaki, Tokyo, Japan) with 25 μl of medium (OpTmizer™ T Cell Expansion SFM; Invitrogen, Carlsbad, CA, USA) containing 10% FBS (MP Biologicals, Solon, OH, USA), recombinant human interleukin (IL)-2 (20 IU/ml; Serotec, Oxford, UK) and 10 μM of each peptide. Half of the medium was removed and replaced with new medium containing a corresponding peptide (20 μM) after 3 days of culture. After incubation for the following 6 days, the cells were harvested and tested for their ability to produce IFN-γ in response to either the corresponding peptides or a negative control peptide from human immunodeficiency virus (HIV). Antigen-specific IFN-γ secretion after an 18-h incubation was determined by ELISPOT assay with the Zeiss ELISPOT reader (Carl Zeiss MicroImaging Japan, Tokyo, Japan). Antigen-specific T cell responses were evaluated by the difference between the spot numbers (mean of duplicate samples) in response to the corresponding peptides and those in response to the control peptide. The differences of at least 10 spot numbers per 105 PBMCs were considered significant. If the spot numbers in response to at least one of the vaccine peptides in the post-vaccination PBMCs were more than 2-fold higher than those in the pre-vaccination PBMCs, the changes were considered significant.

Measurement of C-reactive protein (CRP), serum amyloid A (SAA) and cytokines

The levels of CRP, SAA and IL-6 in the plasma were examined by ELISA using kits from R&D Systems (Minneapolis, MN, USA), Invitrogen and eBioscience (San Diego, CA, USA), respectively. Bead-based multiplex assays were used to measure Th1/Th2 cytokines, including IL-2, IL-4, IL-5 and IFN-γ (Invitrogen) with the Luminex 200 system. Frozen plasma samples were thawed, diluted and assayed in duplicate in accordance with the manufacturer’s instructions. The mean of duplicate samples was used for statistical analysis.

Flow cytometric analysis of suppressive immune subsets in PBMCs

Suppressive immune subsets, myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) in PBMCs were examined by flow cytometry. For analysis of MDSCs, PBMCs (0.5x106 cells) were stained with the following monoclonal antibodies for 30 min at 4°C: anti-CD3-FITC, anti-CD56-FITC, anti-CD19-FITC, anti-CD33-APC, anti-HLA-DR-PE/Cy7 and anti-CD14-APC/Cy7 (all from Biolegend, San Diego, CA, USA). In the cell subpopulation negative for the lineage markers (CD3, CD19, CD56 and CD14) and HLA-DR, MDSCs were identified as positive for CD33. The frequency of MDSCs in the mononuclear cell gate defined by the forward scatter and side scatter was calculated. For analysis of Treg, PBMCs (1x106 cells) were stained with the cocktail of anti-CD4-FITC and anti-CD25-APC, and subsequently with anti-Foxp3-PE following fixation and permeabilization, according to the manufacturer’s instructions (eBioscience). The frequency of CD4+CD25+Foxp3+ cells in CD4+ cells was calculated. The samples were run on a FACSCanto II (BD Biosciences, San Diego, CA, USA), and data were analyzed using the Diva software (BD Biosciences).

Statistical methods

The two-sided Wilcoxon test was used to compare differences between pre- and post-vaccination measurements. OS time was calculated from the first day of peptide vaccination until the date of mortality or the last date when the patient was known to be alive. Predictive factors for OS were evaluated by univariate and multivariate analyses with the Cox proportional hazards regression model. P-values <0.05 were considered to indicate a statistically significant difference. All the statistical analyses were conducted using the SAS version 9.1 software (SAS Institute Inc., Cary, NC, USA).

Results

Patient characteristics

Between November 2008 and December 2010, 25 BTC patients were enrolled in the present study. Table II shows the clinicopathological characteristics of the enrolled patients. There were 18 male and 7 female subjects, with a median age of 59 years, ranging from 37 to 79 years. Primary sites of BTC were 7 gallbladder carcinomas, 11 extrahepatic and 6 intrahepatic cholangiocarcinomas, and 1 periampullary carcinoma. All the patients had advanced-stage cancer (stage IVa, n=5; stage IVb, n=9; recurrent, n=11). Prior to enrollment, 22 patients failed to respond to 1 (n=13) or 2 (n=9) regimen(s) of chemotherapy, whereas the remaining 3 patients did not tolerate chemotherapy due to adverse events. The median duration of chemotherapy prior to the PPV was 4 months, ranging from 2 to 27 months. The performance status at the time of enrollment was grade 0 (n=20) or grade 1 (n=5). The numbers of peptides vaccinated to the patients at the first cycle of vaccination were 4 peptides in 19 patients, 3 in 5 patients and 2 in 1 patient. The median number of vaccinations was 10, with a range of 2 to 24. During the PPV, 20 of 25 patients were treated in combination with chemotherapy, but the remaining 5 patients did not tolerate combined chemotherapy (patients 2, 9, 12, 13 and 25).

Table II.

Characteristics of the enrolled patients.

Table II.

Characteristics of the enrolled patients.

Patient no.GenderAge (years)PSDisease typeStagePrevious treatment (months)aNo. of vaccinationsClinical responseOS (days)
1M590ICCRGEM + S-1 (2)18SD463
2F711GBCIVb-2NA57
3F591GBCIVbGEM→GEM + CDDP (8)4NA35
4M570ECCIVbGEM + S-1 (3)7NA116
5M750GBCIVbGEM→GEM + S-1 (2)5NA122
6M550PACRS-1→GEM (12)14SD234
7M650ECCRGEM→GEM + S-1 (4)6NA102
8M731ECCRGEM→S-1 (27)3NA51
9F371ECCIVbGEM + UFT→S-1 (7)3NA48
10F690ECCRGEM→S-1 (12)24bSD455c
11M620ECCIVaGEM→S-1 (6)8NA177
12M490GBCRGEM (6)7NA111
13F560ICCR-16SD222
14M620ECCRGEM + S-1(5)12PD286
15M530ICCIVbGEM (3)6SD84
16M750GBCRS-1 (2)6NA292
17M790ECCIVbS-1 (2)12NA355c
18M590ECCIVbGEM (2)13NA207
19F560GBCIVbGEM (2)7NA92
20M710ECCRGEM + S-1 (12)11NA163c
21M510ICCRGEM + S-1 (2)12SD179c
22M660ECCIVaGEM (3)17bSD179c
23M521ICCIVa5FU + CDDP→GEM + S-1 (14)10NA101
24M410ICCIVaGEM (4)19bPD428c
25F480GBCIVa-14bSD125c

a Duration of previous chemotherapy;

b under treatment;

c patients alive. M, male; F, female; PS, performance status; ICC, intrahepatic cholangiocarcionma; ECC, extrahepatic cholangiocarcinoma; GBC, gallbladder carcinoma; PAC, periampullary carcinoma; R, recurrent; GEM, gemcitabine; CDDP, cisplatin; UFT, tegafur-uracil; SD, stable disease; PD, progressive disease; OS, overall survival; NA, not assessed.

Of the 10 vaccinated patients whose radiological findings were available prior to and following the first cycle of vaccination, none had a complete response (CR) or partial response (PR). The best response was stable disease (SD) in 8 (80%) patients. The remaining 2 patients (20%) had progressive disease (PD) (Table II).

Toxicities

The overall toxicities are shown in Table III. The most frequent adverse events were dermatological reactions at the injection sites (n=17), hematological toxicity (n=14) and cholangitis (n=11). Severe adverse events (grade 3) were as follows: injection site reaction (n=1), gastrointestinal hemorrhage (n=2), gastrointestinal stricture (n=1), cholangitis (n=11), anemia (n=1), hyperbilirubinemia (n=1) and elevation of ALT (n=1) and ALP (n=1). According to an assessment by the independent safety evaluation committee in this trial, all of these severe adverse events, except for 1 case with a grade 3 injection site reaction, were due to cancer progression or other causes, rather than to the vaccinations themselves.

Table III.

Toxicities.

Table III.

Toxicities.

Grade 1Grade 2Grade 3Total
Injection site reaction115117
Gastrointestinal (GI)
  GI hemorrhage0022
  GI stricture0011
  Abdominal distension0101
  Constipation0101
  Ascites1001
Hepatobiliary
  Cholangitis001111
Pulmonary
  Pleural effusion1001
Cardiac general
  Hypertension0101
Blood/bone marrow
  Anemia91111
  Leukocytopenia1001
  Lymphopenia2002
Laboratory
  Hyperbilirubinemia1012
  AST elevation4105
  ALT elevation1113
  ALP elevation3216
  Hypoalbuminemia4307
  Hyperglycemia0303
  Hyponatremia1001
  Hypokalemia0101
  Hypercalcemia1102
  Creatinine elevation1001
Immune responses to the vaccine peptides

Both humoral and T cell responses specific to the vaccine peptides were analyzed in blood samples prior to and following vaccination (data not shown). Plasma samples were obtained from 25, 20 and 8 patients before and at the end of the first (6th vaccination) and second (12th vaccination) cycles of vaccination, respectively. The post-vaccination samples were not available in the patients who failed to complete the first or second cycle of 6 vaccinations due to disease progression. The IgG responses specific to at least one of the vaccine peptides were augmented in 7 of 20 patients (35%) and in 7 of 8 patients (88%) at the end of the first and second cycles of vaccination, respectively.

T cell responses to the vaccine peptides were measured by IFN-γ ELISPOT assay with PBMCs. PBMCs were available for this assay in 22, 17 and 7 patients prior to and at the end of the first and second cycle of vaccination, respectively. In the pre-vaccination samples, antigen-specific T cell responses were detectable in 5 patients (23%). Of the 17 patients who completed the first cycle of vaccination, 8 patients (47%) showed an induction of T cell responses to the vaccine peptides. At the end of the second cycle of vaccination, the antigen-specific T cell responses were induced in 4 of 7 patients (57%). It should be noted that 3 of the 4 patients with positive T cell responses at the end of the second cycle of vaccination showed reactivity to more than 2 peptides. Collectively, substantial increases in peptide-specific IgG titers and/or T cell responses following vaccination were observed in a subset of the vaccinated patients.

Cytokines and inflammation markers

We then measured several cytokines, including IL-2, IL-4, IL-5, IL-6, IFN-γ and the inflammation markers, CRP and SSA, in the plasma prior to and following the first cycle of vaccination. IL-6 was detect able in 17 of 25 patients (68%) prior to vaccination (median, 2 pg/ml; range, 0–21). Among the 20 plasma samples available at the end of the first cycle of vaccination, IL-6 levels were increased, decreased or unchanged in 12, 5 or 3 patients, respectively (median 3 pg/ml; range 0–43). There was no significant difference in the levels of IL-6 between pre- and post-vaccination samples (P=0.118, Wilcoxon test). Other cytokines, including IL-2, IL-4, IL-5 and IFN-γ, were rarely detectable in either pre- or post-vaccination plasma (data not shown).

The inflammation marker, CRP, was detectable in pre-vaccination plasma from all (100%) of the patients (median, 6.377 μg/ml; range, 0.043–8.891). Among the 20 plasma samples tested at the end of the first cycle of vaccination, plasma CRP levels were increased or decreased in 12 or 8 patients, respectively (median, 6.232 μg/ml; range, 1.331–17.332). Another inflammation marker, SAA, was also detected in pre-vaccination plasma from 21 (84%) of 25 patients (median, 113.486 μg/ml; range, 0–134.425). At the end of the first cycle of vaccination, plasma SAA levels were increased, decreased or unchanged in 12, 7 or 1 patients, respectively (median, 104.861 μg/ml; range, 0–138.917). There were no significant differences in the levels of CRP and SAA between pre- and post-vaccination samples (P=0.290 and P=0.252, respectively, Wilcoxon test).

Relationship between pre-vaccination clinical findings or laboratory data and OS

To identify potential biomarkers useful for selecting suitable patients for PPV, a Cox proportional hazards regression model was used with pre-vaccination clinical findings or laboratory data (Table IV). In the univariate analysis, IL-6, CRP, albumin, SAA and hemoglobin in pre-vaccination samples (P=0.002, P=0.004, P=0.008, P=0.031 and P=0.039, respectively), and the numbers of peptides selected for vaccination (P=0.039) were prognostic factors of OS. None of the other factors examined, such as age, gender, duration of previous chemotherapy, lymphocyte counts or frequencies of suppressive immune cell subsets (Treg and MDSCs) prior to vaccination, were statistically correlated with OS. Furthermore, multivariate Cox regression analysis was performed to define the clinical and laboratory features that were independently associated with OS by adjusting for possible confounding factors. Only the factors with a prognostic association in the univariate analysis, including IL-6, CRP, albumin, hemoglobin and the numbers of peptides selected for vaccination, were used for the multivariate analysis. SAA was not included for this analysis, since the levels of SAA were highly correlated with those of CRP (Pearson’s correlation co-efficient 0.707; P=0.0002). As shown in Table IV, lower IL-6 and higher albumin levels in pre-vaccination samples and greater numbers of antigen peptides selected for vaccination were significantly favorable factors for OS [hazard ratio (HR) = 1.123, 95% confidence interval (CI) 1.008–1.252, P=0.035; HR=0.158, 95% CI 0.029–0.860, P=0.033; HR=0.258, 95% CI 0.098–0.682, P=0.006; respectively]. However, the other factors had no significant association.

Table IV.

Univariate and multivariate analyses with pre-vaccination clinical findings and laboratory data.

Table IV.

Univariate and multivariate analyses with pre-vaccination clinical findings and laboratory data.

FactorUnivariate analysis
Multivariate analysis
Hazard ratio (95% CI)P-valueaHazard ratio (95% CI)P-valuea
Age0.986 (0.944–1.030)0.523
Gender1.673 (0.586–4.776)0.336
Duration of previous chemotherapy (months)1.056 (0.965–1.154)0.235
Lymphocyte count (x103/mm3)0.639 (0.202–2.023)0.446
Hemoglobin (g/dl)0.618 (0.392–0.976)0.039
Albumin (g/dl)0.158 (0.041–0.616)0.0080.158 (0.029–0.860)0.033
IL 6 (pg/ml)1.159 (1.055–1.274)0.0021.123 (1.008–1.252)0.035
CRP (μg/ml)1.533 (1.143–2.056)0.004
SAA (μg/ml)1.014 (1.001–1.027)0.031
MDSC (%)1.140 (0.823–1.580)0.432
Treg (%)0.823 (0.561–1.206)0.317
No. of selected peptides0.395 (0.163–0.953)0.0390.258 (0.098–0.682)0.006

a P-values determined by the Cox proportional hazard regression model. CI, confidence interval; IL-6, interleukin-6; CRP, C-reactive protein; SAA, serum amyloid A; MDSC, myeloid-derived suppressor cells; Treg, CD4+CD25+Foxp3+ regulatory T cells.

Relationship between post-vaccination clinical findings or laboratory data and OS

To further identify potential post-vaccination markers for predicting patient prognosis, the univariate and multivariate Cox analyses were also carried out with post-vaccination clinical findings or laboratory data from the patients who completed the first cycle of 6 vaccinations (n=20). In the univariate analysis, levels of albumin, IL-6, CRP and hemoglobin (P=0.003, P=0.005, P=0.027 and P=0.031, respectively) and the number of vaccine peptides (P=0.033) were prognostic of OS. In addition, although not statistically significant, positive humoral responses to the vaccine peptides had a tendency to be associated with OS (P=0.089) and were also used for the multivariate Cox analysis. The multivariate analysis demonstrated that, among these factors with a potentially prognostic association in the univariate analysis, lower IL-6 levels and greater numbers of vaccine peptides were significantly favorable factors for OS (HR=1.152, 95% CI 1.052–1.261, P=0.002; HR=0.120, 95% CI 0.027–0.540, P=0.006; respectively) (Table V). However, the other post-vaccination factors were not significantly associated with OS.

Table V.

Univariate and multivariate analyses with post-vaccination clinical findings and laboratory data.

Table V.

Univariate and multivariate analyses with post-vaccination clinical findings and laboratory data.

FactorUnivariate analysis
Multivariate analysis
Hazard ratio (95% CI)P-valueaHazard ratio (95% CI)P-valuea
Elevation of CTL responses0.530 (0.166–1.691)0.284
Elevation of humoral responses0.364 (0.114–1.165)0.089
Hemoglobin (g/dl)0.668 (0.463–0.965)0.031
Albumin (g/dl)0.173 (0.055–0.544)0.003
IL-6 (pg/ml)1.112 (1.033–1.198)0.0051.152 (1.052–1.261)0.002
CRP (μg/ml)1.217 (1.023–1.448)0.027
SAA (μg/ml)1.008 (0.995–1.021)0.234
No. of vaccinated peptides0.271 (0.082–0.899)0.0330.120 (0.027–0.540)0.006

a P-values determined by the Cox proportional hazard regression model. CI, confidence interval; IL-6, interleukin-6; CRP, C-reactive protein; SAA, serum amyloid A.

Discussion

For patients with advanced or recurrent BTC that are ineligible for surgery, various regimens of chemotherapeutic agents have been investigated (14). For example, a combination of chemotherapeutic agents, such as gemcitabine and cisplatin, has recently demonstrated a promising result (3). However, further treatment modalities for refractory patients who are unresponsive to or relapse following such regimens remain to be established. This is the first clinical report of refractory BTC patients who received PPV. Immune responses to the vaccine antigens, which have been reported to be significantly associated with clinical responses in previously conducted clinical trials of PPV (6,14), were substantially induced in a subset of the vaccinated patients. Toxicity of PPV mainly involved skin reactions at the injection sites, and no severe adverse events were observed. Based on the positive immune responses to vaccine antigens and the safety profile, PPV could be further investigated as one of the promising approaches for refractory BTC.

The most unique aspect of PPV is the ‘personalized’ selection of antigen peptides ideal for individual patients in consideration of the pre-existing host immunity prior to vaccination (57). In view of the heterogeneity and complexity of host immune responses against tumors, this approach appears to be more rational than vaccination with non-personalized ‘universal’ tumor antigens. Notably, in the present study, the number of selected and vaccinated peptides was significantly associated with OS in the multivariate analysis, suggesting that greater numbers of peptides would be required for better clinical responses, possibly due to the heterogeneity and complexity of host immune responses against tumors.

Cancer vaccines do not always elicit beneficial immune or clinical responses in treated patients. Therefore, identification of biomarkers for predicting clinical responses in vaccinated patients would be a significant issue in the clinical application of cancer vaccines (5,1517). At present, however, there is little information available regarding predictive biomarkers in patients undergoing cancer vaccines. In this study, the multivariate analysis demonstrated that lower IL-6 and higher albumin values, which may reflect less inflammation and better nutritional status, prior to vaccination were significantly favorable factors for OS. IL-6 is a multifunctional cytokine that regulates various aspects of immune responses, acute phase reactions and hematopoiesis. In particular, IL-6 has been reported to be deeply involved in cancer development, such as tumor cell growth and cancer-associated inflammation (18).

There have been a number of studies describing the correlation between IL-6 levels and prognosis in various types of cancer (1922). IL-6 has also been reported to be one of the critical cytokines for inducing suppressive immune cell subsets. For example, MDSCs and Th17, which are known to modulate antitumor immunity, were shown to be generated from their precursors in the presence of IL-6 and other cytokines (2325). Although the role of IL-6 in the immune response to cancer vaccines remains to be clarified, it is possible that the blockage of IL-6 signaling would be beneficial for enhancing the therapeutic efficacy of cancer vaccines.

In conclusion, the present study demonstrated that PPV induced substantial immune responses to vaccine antigens without severe adverse events in advanced BTC patients. In addition, the multivariate analysis suggested that lower plasma IL-6 and better nutritional status prior to vaccination and pre-existing immune responses to greater numbers of antigens may contribute to better responses to PPV. Therefore, the evaluation of these factors prior to vaccination may be useful for selecting patients who would benefit from PPV and defining eligibility and/or exclusion criteria for molecular-based personalized immunotherapy in BTC patients. Nevertheless, since this was a small study with a limited number of patients, all of whom received PPV, the clinical efficacy of PPV, as well as the clinical utility of the identified factors in refractory BTC patients remain to be confirmed in future larger-scale prospective trials conducted in defined patient populations with or without receiving PPV.

Acknowledgements

This study was supported by grants from the Regional Innovation Cluster Program of the Ministry of Education, Culture, Sports, Science and Technology of Japan, the Sendai-Kousei Hospital, the Kurozumi Medical Foundation and the Osaka Cancer Research Foundation.

References

1. 

P YachimskiDS PrattCholangiocarcinoma: natural history, treatment, and strategies for surveillance in high-risk patientsJ Clin Gastroenterol42178190200810.1097/MCG.0b013e31806daf8918209589

2. 

AF HezelV DeshpandeAX ZhuGenetics of biliary tract cancers and emerging targeted therapiesJ Clin Oncol2835313540201010.1200/JCO.2009.27.478720547994

3. 

J ValleH WasanDH PalmerCisplatin plus gemcitabine versus gemcitabine for biliary tract cancerN Engl J Med36212731281201010.1056/NEJMoa090872120375404

4. 

B GruenbergerJ SchuellerU HeubrandtnerCetuximab, gemcitabine, and oxaliplatin in patients with unresectable advanced or metastatic biliary tract cancer: a phase 2 studyLancet Oncol1111421148201010.1016/S1470-2045(10)70247-321071270

5. 

T SasadaN KomatsuS SuekaneA YamadaM NoguchiK ItohOvercoming the hurdles of randomised clinical trials of therapeutic cancer vaccinesEur J Cancer4615141519201010.1016/j.ejca.2010.03.01320413296

6. 

T MineY SatoM NoguchiHumoral responses to peptides correlate with overall survival in advanced cancer patients vaccinated with peptides based on pre-existing, peptide-specific cellular responsesClin Cancer Res10929937200410.1158/1078-0432.CCR-1117-3

7. 

K ItohA YamadaPersonalized peptide vaccines: a new therapeutic modality for cancerCancer Sci97970976200610.1111/j.1349-7006.2006.00272.x16984371

8. 

H YanagimotoH ShiomiS SatoiA phase II study of personalized peptide vaccination combined with gemcitabine for non-resectable pancreatic cancer patientsOncol Rep24795801201020664989

9. 

Y SatoT FujiwaraT MineImmunological evaluation of personalized peptide vaccination in combination with a 5-fluorouracil derivative (TS-1) for advanced gastric or colorectal carcinoma patientsCancer Sci9811131119200710.1111/j.1349-7006.2007.00498.x

10. 

T HattoriT MineN KomatsuImmunological evaluation of personalized peptide vaccination in combination with UFT and UZEL for metastatic colorectal carcinoma patientsCancer Immunol Immunother5818431852200910.1007/s00262-009-0695-6

11. 

M NoguchiT KakumaH UemuraA randomized phase II trial of personalized peptide vaccine plus low dose estramustine phosphate (EMP) versus standard dose EMP in patients with castration resistant prostate cancerCancer Immunol Immunother5910011009201010.1007/s00262-010-0822-4

12. 

M TerasakiS ShibuiY NaritaPhase I trial of a personalized peptide vaccine for patients positive for human leukocyte antigen – A24 with recurrent or progressive glioblastoma multiformeJ Clin Oncol293373442011

13. 

N KomatsuS ShichijoM NakagawaK ItohNew multiplexed flow cytometric assay to measure anti-peptide antibody: a novel tool for monitoring immune responses to peptides used for immunizationScand J Clin Lab Invest64535545200410.1080/0036551041000700815370458

14. 

M NoguchiT MineN KomatsuAssessment of immunological biomarkers in patients with advanced cancer treated by personalized peptide vaccinationCancer Biol Ther1012661279201110.4161/cbt.10.12.1344820935522

15. 

ML DisisImmunologic biomarkers as correlates of clinical response to cancer immunotherapyCancer Immunol Immunother60433442201110.1007/s00262-010-0960-821221967

16. 

LH ButterfieldAK PaluckaCM BrittenRecommendations from the iSBTc-SITC/FDA/NCI Workshop on Immunotherapy BiomarkersClin Cancer Res1730643076201110.1158/1078-0432.CCR-10-223421558394

17. 

A HoosAM EggermontS JanetzkiImproved endpoints for cancer immunotherapy trialsJ Natl Cancer Inst10213881397201010.1093/jnci/djq31020826737

18. 

WE NauglerM KarinThe wolf in sheep’s clothing: the role of interleukin-6 in immunity, inflammation and cancerTrends Mol Med141091192008

19. 

G ScambiaU TestaP Benedetti PaniciPrognostic significance of interleukin 6 serum levels in patients with ovarian cancerBr J Cancer71354356199510.1038/bjc.1995.717841052

20. 

J NakashimaM TachibanaY HoriguchiSerum interleukin 6 as a prognostic factor in patients with prostate cancerClin Cancer Res627022706200010914713

21. 

S OkadaT OkusakaH IshiiElevated serum interleukin-6 levels in patients with pancreatic cancerJpn J Clin Oncol281215199810.1093/jjco/28.1.129491135

22. 

JS GoydosAM BrumfieldE FrezzaA BoothMT LotzeSE CartyMarked elevation of serum interleukin-6 in patients with cholangiocarcinoma: validation of utility as a clinical markerAnn Surg227398404199810.1097/00000658-199803000-000129527063

23. 

I MarigoE BosioS SolitoTumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factorImmunity32790802201010.1016/j.immuni.2010.05.01020605485

24. 

MG LechnerDJ LiebertzAL EpsteinCharacterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cellsJ Immunol18522732284201010.4049/jimmunol.1000901

25. 

W ZouNP RestifoT(H)17 cells in tumour immunity and immunotherapyNat Rev Immunol10248256201010.1038/nri274220336152

Related Articles

Journal Cover

March 2012
Volume 3 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yoshitomi M, Yutani S, Matsueda S, Ioji T, Komatsu N, Shichijo S, Yamada A, Itoh K, Sasada T, Kinoshita H, Kinoshita H, et al: Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis. Exp Ther Med 3: 463-469, 2012
APA
Yoshitomi, M., Yutani, S., Matsueda, S., Ioji, T., Komatsu, N., Shichijo, S. ... Kinoshita, H. (2012). Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis. Experimental and Therapeutic Medicine, 3, 463-469. https://doi.org/10.3892/etm.2011.424
MLA
Yoshitomi, M., Yutani, S., Matsueda, S., Ioji, T., Komatsu, N., Shichijo, S., Yamada, A., Itoh, K., Sasada, T., Kinoshita, H."Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis". Experimental and Therapeutic Medicine 3.3 (2012): 463-469.
Chicago
Yoshitomi, M., Yutani, S., Matsueda, S., Ioji, T., Komatsu, N., Shichijo, S., Yamada, A., Itoh, K., Sasada, T., Kinoshita, H."Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis". Experimental and Therapeutic Medicine 3, no. 3 (2012): 463-469. https://doi.org/10.3892/etm.2011.424