A phase I/IIa study of adjuvant immunotherapy with tumour antigen-pulsed dendritic cells in patients with hepatocellular carcinoma

Jeong-Hoon Lee, Yoon Lee, Minjong Lee, Min Kyu Heo, Jae-Sung Song, Ki-Hwan Kim, Hyunah Lee, Nam-Joon Yi, Kwang-Woong Lee, Kyung-Suk Suh, Yong-Soo Bae, Yoon Jun Kim, Jeong-Hoon Lee, Yoon Lee, Minjong Lee, Min Kyu Heo, Jae-Sung Song, Ki-Hwan Kim, Hyunah Lee, Nam-Joon Yi, Kwang-Woong Lee, Kyung-Suk Suh, Yong-Soo Bae, Yoon Jun Kim

Abstract

Background: To date, no adjuvant treatment has been shown to have a clear benefit in patients with hepatocellular carcinoma (HCC). In this prospective phase I/IIa study, we evaluated the safety and efficacy of adjuvant dendritic cell (DC) therapy in HCC patients who received primary treatment for HCC.

Methods: Twelve HCC patients who had no viable tumour after primary treatments were included. Dendritic cell vaccines pulsed with cytoplasmic transduction peptide-attached alpha-fetoprotein, glypican-3 and melanoma-associated antigen 1 recombinant fusion proteins were injected subcutaneously near to inguinal lymph nodes. Adverse effects, time to progression (TTP), and associated immune responses were evaluated after DC vaccination.

Results: Nine of 12 patients had no tumour recurrence up to 24 weeks after DC vaccination. Among a total of 144 adverse events, 129 events (89.6%) were regarded as adverse drug reactions, all of which were grade 1 or 2. The majority of patients showed enhanced anti-tumour immune responses after DC vaccination. Recurrence-free patients exhibited relatively stronger anti-tumour immune responses than patients who developed recurrence after DC vaccination, as evidenced by lymphocyte proliferation and IFN-γ ELISPOT assays. The median time of TTP was 36.6 months in the DC-vaccination group and 11.8 months in the control group (hazard ratio, 0.41; 95% confidence interval, 0.18-0.95; P=0.0031 by log-rank test).

Conclusions: Adjuvant DC vaccine for HCC was safe and well tolerated in phase I/IIa study, and preliminary efficacy data are encouraging to warrant further clinical study in patients with HCC after primary treatments.

Conflict of interest statement

YL, MKHeo, and J-SS are CreaGene Inc. employees. Y-SB has been a consultant for JW CreaGene Inc. All remaining authors have declared no conflicts of interest.

Figures

Figure 1
Figure 1
Study design. (A) Study design for vaccination and evaluation. CT=computed tomography; MRI=magnetic resonance imaging; DC=dendritic cell. (B) Flow chart for tolerated dose-finding study. DLT=dose-limiting toxicity; TD=tolerated dose.
Figure 2
Figure 2
Surface phenotypes and T-cell stimulation capacity of TAA-pulsed mDCs in the preclinical study, which were performed as described in the Supplementary Method. (A) Representative FACS data (left) and statistical analysis (right) of the surface phenotype of imDCs and TAA-pulsed mDCs. (B) CD8+ T-cell population (left) and the levels of IFN-γ and IL-4 (right) were assessed from the co-culture of autologous T cells with TAA-pulsed or Ag-unpulsed mDCs at a 10 : 1 ratio in the presence of IL-7 (Peprotech). Activated T cells were restimulated three times every 7–8 days with a same set of TAA-pulsed DCs at the same ratio as described in the Supplementary Method. (C) From the activated T cells, the CTL was assessed with MHC-matched HepG2 target cells at different effector/target (E/T) ratios as described in the Supplementary Method. Data are presented as mean±s.d. of nine samples pooled from three individual experiments. *P<0.05 and **P<0.01 compared with control CTLs prepared by stimulation of T cells with Ag-unpulsed mDCs, Student's t-test.
Figure 3
Figure 3
Immunological analysis after DC vaccination. (A) Antigen-specific lymphocyte proliferation assay was performed during and after DC vaccinations using the autologous PBMCs obtained from 12 patients (upper), and the results were further analysed in recurrence-free patients (lower left) and recurrent patients (lower right). The proliferation was determined by 3H-thymidine incorporation (dpm) using a liquid scintillation counter. Data are presented as mean±s.e., *P<0.05. (B) ELISPOT assay was performed with the PBMCs obtained from each patient at the indicated time points after the start of DC vaccination. The results from all (12) patients (upper), the nine recurrence-free patients (lower left) and the three recurrent patients (lower right) are presented as mean±s.e.
Figure 4
Figure 4
CT and MRI scan data of two representative patients (pt4 and pt6) before and after one cycle of antigen-pulsed DC vaccination. Treatment records of each patient are summarised.
Figure 5
Figure 5
TTP and RFS (starting from primary treatments rather than baseline radiographic tumour assessments) of patients treated with DC vaccine during the trial period and 4-year follow-up study in comparison with historical controls (n=31). The composition of 31 patients with HCC for historical control group was summarised in Table 5. A statistically significant difference in survival was noted (P<0.05).

References

    1. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature 392(6673): 245–252.
    1. Beyer M, Schultze JL (2006) Regulatory T cells in cancer. Blood 108(3): 804–811.
    1. Bolondi L, Sofia S, Siringo S, Gaiani S, Casali A, Zironi G, Piscaglia F, Gramantieri L, Zanetti M, Sherman M (2001) Surveillance programme of cirrhotic patients for early diagnosis and treatment of hepatocellular carcinoma: a cost effectiveness analysis. Gut 48(2): 251–259.
    1. Britten CM, Janetzki S, Butterfield LH, Ferrari G, Gouttefangeas C, Huber C, Kalos M, Levitsky HI, Maecker HT, Melief CJ, O'Donnell-Tormey J, Odunsi K, Old LJ, Ottenhoff TH, Ottensmeier C, Pawelec G, Roederer M, Roep BO, Romero P, van der Burg SH, Walter S, Hoos A, Davis MM (2012) T cell assays and MIATA: the essential minimum for maximum impact. Immunity 37(1): 1–2.
    1. Bruix J, Sherman M American Association for the Study of Liver Diseases (2011) Management of hepatocellular carcinoma: an update. Hepatology 53(3): 1020–1022.
    1. Butterfield LH, Ribas A, Dissette VB, Lee Y, Yang JQ, De la Rocha P, Duran SD, Hernandez J, Seja E, Potter DM, McBride WH, Finn R, Glaspy JA, Economou JS (2006) A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four alpha-fetoprotein peptides. Clin Cancer Res 12(9): 2817–2825.
    1. den Brok MH, Nierkens S, Figdor CG, Ruers TJ, Adema GJ (2005) Dendritic cells: tools and targets for antitumor vaccination. Expert Rev Vaccines 4(5): 699–710.
    1. Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ (2009) Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 58(1): 49–59.
    1. European Association for The Study of The Liver, European Organisation for Research Treatment of Cancer (2012) EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol 56(4): 908–943.
    1. Greene FL. American Joint Committee on Cancer, American Cancer Society (2002) AJCC Cancer Staging Manual 6th edn. Springer-Verlag: New York.
    1. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140(6): 883–899.
    1. Hermans IF, Ritchie DS, Yang J, Roberts JM, Ronchese F (2000) CD8+ T cell-dependent elimination of dendritic cells in vivo limits the induction of antitumor immunity. J Immunol 164(6): 3095–3101.
    1. Holtl L, Rieser C, Papesh C, Ramoner R, Herold M, Klocker H, Radmayr C, Stenzl A, Bartsch G, Thurnher M (1999) Cellular and humoral immune responses in patients with metastatic renal cell carcinoma after vaccination with antigen pulsed dendritic cells. J Urol 161(3): 777–782.
    1. Kim D, Jeon C, Kim JH, Kim MS, Yoon CH, Choi IS, Kim SH, Bae YS (2006) Cytoplasmic transduction peptide (CTP): new approach for the delivery of biomolecules into cytoplasm in vitro and in vivo. Exp Cell Res 312(8): 1277–1288.
    1. Kim JH, Lee Y, Bae YS, Kim WS, Kim K, Im HY, Kang WK, Park K, Choi HY, Lee HM, Baek SY, Lee H, Doh H, Kim BM, Kim CY, Jeon C, Jung CW (2007) Phase I/II study of immunotherapy using autologous tumor lysate-pulsed dendritic cells in patients with metastatic renal cell carcinoma. Clin Immunol 125(3): 257–267.
    1. Korangy F, Hochst B, Manns MP, Greten TF (2010) Immune responses in hepatocellular carcinoma. Dig Dis 28(1): 150–154.
    1. Lai EC, Fan ST, Lo CM, Chu KM, Liu CL, Wong J (1995) Hepatic resection for hepatocellular carcinoma. An audit of 343 patients. Ann Surg 221(3): 291–298.
    1. Lappin MB, Weiss JM, Delattre V, Mai B, Dittmar H, Maier C, Manke K, Grabbe S, Martin S, Simon JC (1999) Analysis of mouse dendritic cell migration in vivo upon subcutaneous and intravenous injection. Immunology 98(2): 181–188.
    1. Lee WC, Wang HC, Hung CF, Huang PF, Lia CR, Chen MF (2005) Vaccination of advanced hepatocellular carcinoma patients with tumor lysate-pulsed dendritic cells: a clinical trial. J Immunother 28(5): 496–504.
    1. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D (1998) Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med 4(3): 328–332.
    1. Okada N, Tsujino M, Hagiwara Y, Tada A, Tamura Y, Mori K, Saito T, Nakagawa S, Mayumi T, Fujita T, Yamamoto A (2001) Administration route-dependent vaccine efficiency of murine dendritic cells pulsed with antigens. Br J Cancer 84(11): 1564–1570.
    1. Palmer DH, Midgley RS, Mirza N, Torr EE, Ahmed F, Steele JC, Steven NM, Kerr DJ, Young LS, Adams DH (2009) A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology 49(1): 124–132.
    1. Prins RM, Craft N, Bruhn KW, Khan-Farooqi H, Koya RC, Stripecke R, Miller JF, Liau LM (2006) The TLR-7 agonist, imiquimod, enhances dendritic cell survival and promotes tumor antigen-specific T cell priming: relation to central nervous system antitumor immunity. J Immunol 176(1): 157–164.
    1. Samuel M, Chow PK, Chan Shih-Yen E, Machin D, Soo KC (2009) Neoadjuvant and adjuvant therapy for surgical resection of hepatocellular carcinoma. Cochrane Database Syst Rev, (1): CD001199.
    1. Schwartz JD, Schwartz M, Mandeli J, Sung M (2002) Neoadjuvant and adjuvant therapy for resectable hepatocellular carcinoma: review of the randomised clinical trials. Lancet Oncol 3(10): 593–603.
    1. Small EJ, Fratesi P, Reese DM, Strang G, Laus R, Peshwa MV, Valone FH (2000) Immunotherapy of hormone-refractory prostate cancer with antigen-loaded dendritic cells. J Clin Oncol 18(23): 3894–3903.
    1. Tada F, Abe M, Hirooka M, Ikeda Y, Hiasa Y, Lee Y, Jung NC, Lee WB, Lee HS, Bae YS, Onji M (2012) Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Int J Oncol 41(5): 1601–1609.
    1. Ungefroren H, Sebens S, Seidl D, Lehnert H, Hass R (2011) Interaction of tumor cells with the microenvironment. Cell Commun Signal 9: 18.
    1. Verslype C, Rosmorduc O, Rougier P ESMO Guidelines Working Group (2012) Hepatocellular carcinoma: ESMO-ESDO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 23(Suppl 7): vii41–vii48.
    1. Yuen MF, Cheng CC, Lauder IJ, Lam SK, Ooi CG, Lai CL (2000) Early detection of hepatocellular carcinoma increases the chance of treatment: Hong Kong experience. Hepatology 31(2): 330–335.
    1. Zamarron BF, Chen W (2011) Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 7(5): 651–658.

Source: PubMed

3
Suscribir