Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 6/2008

01-06-2008 | Original Article

Autologous versus allogeneic peptide-pulsed dendritic cells for anti-tumour vaccination: expression of allogeneic MHC supports activation of antigen specific T cells, but impairs early naïve cytotoxic priming and anti-tumour therapy

Authors: Alison Merrick, Rosa Maria Diaz, Dearbhaile O’Donnell, Peter Selby, Richard Vile, Alan Melcher

Published in: Cancer Immunology, Immunotherapy | Issue 6/2008

Login to get access

Abstract

Background

Dendritic cells (DC) pulsed with MHC class I-restricted tumour associated antigen (TAA) peptides have been widely tested in pre-clinical models and early clinical studies for their ability to prime cytotoxic T cell (CTL) responses. The effect of co-expression of allogeneic MHC antigens on DC immunogenicity has not been addressed, and has implications for the feasibility of clinical applications.

Objective

This study compared DC from autologous H-2b or semi-allogeneic F1 H-2bxk mice pulsed with the H-2b-restricted model ovalbumin (OVA) peptide SIINFEKL, and compared in vitro and in vivo their ability to (i) activate specific OT1 cells, (ii) prime naïve CTL, and (iii) protect against B16.OVA challenge. Peptide-pulsed autologous and allogeneic DC were also tested in naïve human CTL priming assays.

Results

Semi-allogeneic DC expressed higher levels of co-stimulatory molecules. On pulsing with SIINFEKL they triggered greater proliferation of OT1 cells in vitro and in vivo, but were less effective at naïve CTL priming and tumour protection. Autologous human DC were similarly more potent at naïve CTL priming against the melanoma-associated TAA MART-1 in vitro.

Conclusion

The expression of allogeneic MHC antigens on peptide-pulsed DC impairs naïve CTL priming and anti-tumour effects, despite effective TAA presentation both in vitro and in vivo.
Literature
1.
go back to reference Banchereau J et al (2005) Immune and clinical outcomes in patients with stage IV melanoma vaccinated with peptide-pulsed dendritic cells derived from CD34+ progenitors and activated with type I interferon. J Immunother 28(5):505–516PubMedCrossRef Banchereau J et al (2005) Immune and clinical outcomes in patients with stage IV melanoma vaccinated with peptide-pulsed dendritic cells derived from CD34+ progenitors and activated with type I interferon. J Immunother 28(5):505–516PubMedCrossRef
2.
go back to reference Guermonprez P et al (2002) Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol 20:621–667PubMedCrossRef Guermonprez P et al (2002) Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol 20:621–667PubMedCrossRef
3.
go back to reference Rescigno M et al (1997) Dendritic cell maturation is required for initiation of the immune response. J Leukoc Biol 61(4):415–421PubMed Rescigno M et al (1997) Dendritic cell maturation is required for initiation of the immune response. J Leukoc Biol 61(4):415–421PubMed
4.
go back to reference Smits HH et al (2005) Different faces of regulatory DCs in homeostasis and immunity. Trends Immunol 26(3):123–129PubMedCrossRef Smits HH et al (2005) Different faces of regulatory DCs in homeostasis and immunity. Trends Immunol 26(3):123–129PubMedCrossRef
5.
go back to reference Ferlazzo G (2005) Natural killer and dendritic cell liaison: recent insights and open questions. Immunol Lett 101(1):12–17PubMedCrossRef Ferlazzo G (2005) Natural killer and dendritic cell liaison: recent insights and open questions. Immunol Lett 101(1):12–17PubMedCrossRef
6.
go back to reference Melcher A et al (2002) Dendritic cells for the immunotherapy of cancer. Clin Oncol (R Coll Radiol) 14(3):185–192 Melcher A et al (2002) Dendritic cells for the immunotherapy of cancer. Clin Oncol (R Coll Radiol) 14(3):185–192
7.
go back to reference Gong J et al (1997) Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat Med 3(5):558–561PubMedCrossRef Gong J et al (1997) Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat Med 3(5):558–561PubMedCrossRef
8.
go back to reference Phan V et al (2003) A new genetic method to generate and isolate small, short-lived but highly potent dendritic cell-tumor cell hybrid vaccines. Nat Med 9(9):1215–1219PubMedCrossRef Phan V et al (2003) A new genetic method to generate and isolate small, short-lived but highly potent dendritic cell-tumor cell hybrid vaccines. Nat Med 9(9):1215–1219PubMedCrossRef
9.
go back to reference Plautz GE et al (1993) Immunotherapy of malignancy by in vivo gene transfer into tumors. Proc Natl Acad Sci USA 90(10):4645–4649PubMedCrossRef Plautz GE et al (1993) Immunotherapy of malignancy by in vivo gene transfer into tumors. Proc Natl Acad Sci USA 90(10):4645–4649PubMedCrossRef
10.
go back to reference Errington F et al (2006) Allogeneic tumor cells expressing fusogenic membrane glycoproteins as a platform for clinical cancer immunotherapy. Clin Cancer Res 12(4):1333–1341PubMedCrossRef Errington F et al (2006) Allogeneic tumor cells expressing fusogenic membrane glycoproteins as a platform for clinical cancer immunotherapy. Clin Cancer Res 12(4):1333–1341PubMedCrossRef
11.
go back to reference Neves AR et al (2005) Dendritic cells derived from metastatic cancer patients vaccinated with allogeneic dendritic cell-autologous tumor cell hybrids express more CD86 and induce higher levels of interferon-gamma in mixed lymphocyte reactions. Cancer Immunol Immunother 54(1):61–66PubMedCrossRef Neves AR et al (2005) Dendritic cells derived from metastatic cancer patients vaccinated with allogeneic dendritic cell-autologous tumor cell hybrids express more CD86 and induce higher levels of interferon-gamma in mixed lymphocyte reactions. Cancer Immunol Immunother 54(1):61–66PubMedCrossRef
12.
go back to reference Hus I et al (2005) Allogeneic dendritic cells pulsed with tumor lysates or apoptotic bodies as immunotherapy for patients with early-stage B-cell chronic lymphocytic leukemia. Leukemia 19(9):1621–1627PubMedCrossRef Hus I et al (2005) Allogeneic dendritic cells pulsed with tumor lysates or apoptotic bodies as immunotherapy for patients with early-stage B-cell chronic lymphocytic leukemia. Leukemia 19(9):1621–1627PubMedCrossRef
13.
go back to reference Holtl L et al (2005) Allogeneic dendritic cell vaccination against metastatic renal cell carcinoma with or without cyclophosphamide. Cancer Immunol Immunother 54(7):663–670PubMedCrossRef Holtl L et al (2005) Allogeneic dendritic cell vaccination against metastatic renal cell carcinoma with or without cyclophosphamide. Cancer Immunol Immunother 54(7):663–670PubMedCrossRef
14.
go back to reference Gong J et al (2000) Fusions of human ovarian carcinoma cells with autologous or allogeneic dendritic cells induce antitumor immunity. J Immunol 165(3):1705–1711PubMed Gong J et al (2000) Fusions of human ovarian carcinoma cells with autologous or allogeneic dendritic cells induce antitumor immunity. J Immunol 165(3):1705–1711PubMed
15.
go back to reference Tanaka Y et al (2001) Vaccination with allogeneic dendritic cells fused to carcinoma cells induces antitumor immunity in MUC1 transgenic mice. Clin Immunol 101(2):192–200PubMedCrossRef Tanaka Y et al (2001) Vaccination with allogeneic dendritic cells fused to carcinoma cells induces antitumor immunity in MUC1 transgenic mice. Clin Immunol 101(2):192–200PubMedCrossRef
16.
go back to reference Siders WM et al (2003) Induction of specific antitumor immunity in the mouse with the electrofusion product of tumor cells and dendritic cells. Mol Ther 7(4):498–505PubMedCrossRef Siders WM et al (2003) Induction of specific antitumor immunity in the mouse with the electrofusion product of tumor cells and dendritic cells. Mol Ther 7(4):498–505PubMedCrossRef
17.
go back to reference Suzuki T et al (2005) Vaccination of dendritic cells loaded with interleukin-12-secreting cancer cells augments in vivo antitumor immunity: characteristics of syngeneic and allogeneic antigen-presenting cell cancer hybrid cells. Clin Cancer Res 11(1):58–66PubMed Suzuki T et al (2005) Vaccination of dendritic cells loaded with interleukin-12-secreting cancer cells augments in vivo antitumor immunity: characteristics of syngeneic and allogeneic antigen-presenting cell cancer hybrid cells. Clin Cancer Res 11(1):58–66PubMed
18.
go back to reference Wells JW et al (2007) Semi-allogeneic dendritic cells can induce antigen-specific T-cell activation, which is not enhanced by concurrent alloreactivity. Cancer Immunol Immunother 56(12):1861–1873PubMedCrossRef Wells JW et al (2007) Semi-allogeneic dendritic cells can induce antigen-specific T-cell activation, which is not enhanced by concurrent alloreactivity. Cancer Immunol Immunother 56(12):1861–1873PubMedCrossRef
19.
go back to reference Tirapu I et al (2004) Improving efficacy of interleukin-12-transfected dendritic cells injected into murine colon cancer with anti-CD137 monoclonal antibodies and alloantigens. Int J Cancer 110(1):51–60PubMedCrossRef Tirapu I et al (2004) Improving efficacy of interleukin-12-transfected dendritic cells injected into murine colon cancer with anti-CD137 monoclonal antibodies and alloantigens. Int J Cancer 110(1):51–60PubMedCrossRef
20.
go back to reference Chakraborty NG et al (2004) Regulatory T-cell response and tumor vaccine-induced cytotoxic T lymphocytes in human melanoma. Hum Immunol 65(8):794–802PubMedCrossRef Chakraborty NG et al (2004) Regulatory T-cell response and tumor vaccine-induced cytotoxic T lymphocytes in human melanoma. Hum Immunol 65(8):794–802PubMedCrossRef
21.
go back to reference Babatz J et al (2006) Induction of cellular immune responses against carcinoembryonic antigen in patients with metastatic tumors after vaccination with altered peptide ligand-loaded dendritic cells. Cancer Immunol Immunother 55(3):268–276PubMedCrossRef Babatz J et al (2006) Induction of cellular immune responses against carcinoembryonic antigen in patients with metastatic tumors after vaccination with altered peptide ligand-loaded dendritic cells. Cancer Immunol Immunother 55(3):268–276PubMedCrossRef
22.
go back to reference Wierecky J et al (2006) Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res 66(11):5910–5918PubMedCrossRef Wierecky J et al (2006) Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res 66(11):5910–5918PubMedCrossRef
23.
go back to reference Melcher A et al (1999) Adoptive transfer of immature dendritic cells with autologous or allogeneic tumor cells generates systemic antitumor immunity. Cancer Res 59(12):2802–2805PubMed Melcher A et al (1999) Adoptive transfer of immature dendritic cells with autologous or allogeneic tumor cells generates systemic antitumor immunity. Cancer Res 59(12):2802–2805PubMed
24.
go back to reference Gallucci S, Lolkema M, Matzinger P (1999) Natural adjuvants: endogenous activators of dendritic cells. Nat Med 5(11):1249–1255PubMedCrossRef Gallucci S, Lolkema M, Matzinger P (1999) Natural adjuvants: endogenous activators of dendritic cells. Nat Med 5(11):1249–1255PubMedCrossRef
25.
go back to reference Inaba K et al (1992) Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med 176(6):1693–1702PubMedCrossRef Inaba K et al (1992) Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med 176(6):1693–1702PubMedCrossRef
26.
go back to reference Petersen MS et al (2000) Strain-specific variations in the development of dendritic cells in murine bone-marrow cultures. Scand J Immunol 51(6):586–594PubMedCrossRef Petersen MS et al (2000) Strain-specific variations in the development of dendritic cells in murine bone-marrow cultures. Scand J Immunol 51(6):586–594PubMedCrossRef
27.
go back to reference Gallucci S, Matzinger P (2001) Danger signals: SOS to the immune system. Curr Opin Immunol 13(1):114–119PubMedCrossRef Gallucci S, Matzinger P (2001) Danger signals: SOS to the immune system. Curr Opin Immunol 13(1):114–119PubMedCrossRef
28.
go back to reference Yasuda T et al (2007) Superior anti-tumor protection and therapeutic efficacy of vaccination with allogeneic and semiallogeneic dendritic cell/tumor cell fusion hybrids for murine colon adenocarcinoma. Cancer Immunol Immunother 56(7):1025–1036PubMedCrossRef Yasuda T et al (2007) Superior anti-tumor protection and therapeutic efficacy of vaccination with allogeneic and semiallogeneic dendritic cell/tumor cell fusion hybrids for murine colon adenocarcinoma. Cancer Immunol Immunother 56(7):1025–1036PubMedCrossRef
Metadata
Title
Autologous versus allogeneic peptide-pulsed dendritic cells for anti-tumour vaccination: expression of allogeneic MHC supports activation of antigen specific T cells, but impairs early naïve cytotoxic priming and anti-tumour therapy
Authors
Alison Merrick
Rosa Maria Diaz
Dearbhaile O’Donnell
Peter Selby
Richard Vile
Alan Melcher
Publication date
01-06-2008
Publisher
Springer-Verlag
Published in
Cancer Immunology, Immunotherapy / Issue 6/2008
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-007-0426-9

Other articles of this Issue 6/2008

Cancer Immunology, Immunotherapy 6/2008 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine