Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 9/2007

01-09-2007 | Original Article

Development of anti-PAX3 immune responses; a target for cancer immunotherapy

Authors: Nourredine Himoudi, Steven Nabarro, Mengyong Yan, Kimberly Gilmour, Adrian J. Thrasher, John Anderson

Published in: Cancer Immunology, Immunotherapy | Issue 9/2007

Login to get access

Abstract

PAX3 is overexpressed in several human cancers and is absent from normal adult human tissues. It is known to have an oncogenic function in human malignancy, and is therefore a promising target for cancer immunotherapy. We screened the murine and human PAX3 amino acid sequences for peptides that bind common MHC class I types, and identified murine GVFINGRPL and human KLTEARVQV sequences. Mice immunised with either a selected PAX3 peptide, or with a PAX3 expressing DNA vector, developed specific anti-PAX3 immune responses that inhibited tumour growth. The intensity of the immune response was significantly enhanced by pulsing of the peptide onto dendritic cells. Anti-PAX3 T cell lines were established from splenocytes of immunised mice. Intravenous administration of anti-PAX3 T cells caused regression of established tumours indicating a promising clinical application for anti-PAX3 immunotherapy. The human peptide stimulated growth of similar T cell lines from peripheral blood of three out of three normal human blood donors. These showed specific cytotoxicity against a range of human PAX3+ and HLA-A2+ cancer cell lines. Moreover, an anti-PAX3 response was detected as a component of the anti-tumour immune response in a patient treated with lysate pulsed dendritic cell vaccination. The ability to generate strong and specific anti PAX3 immune responses from the T cell repertoire in both mice and humans, provides evidence for PAX3 as a promising target for immunotherapy of cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dudley ME, Rosenberg SA (2003) Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat Rev Cancer 3(9):666–675PubMedCrossRef Dudley ME, Rosenberg SA (2003) Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat Rev Cancer 3(9):666–675PubMedCrossRef
2.
go back to reference Dudley ME et al (2005) Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 23(10):2346–2357PubMedCrossRef Dudley ME et al (2005) Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 23(10):2346–2357PubMedCrossRef
3.
go back to reference Correale P et al (1997) In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. J Natl Cancer Inst 89(4):293–300PubMedCrossRef Correale P et al (1997) In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. J Natl Cancer Inst 89(4):293–300PubMedCrossRef
4.
go back to reference Gao L et al (2000) Selective elimination of leukemic CD34 (+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood 95(7):2198–2203PubMed Gao L et al (2000) Selective elimination of leukemic CD34 (+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood 95(7):2198–2203PubMed
5.
go back to reference Overwijk WW et al (2003) Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8 + T cells. J Exp Med 198(4):569–580PubMedCrossRef Overwijk WW et al (2003) Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8 + T cells. J Exp Med 198(4):569–580PubMedCrossRef
6.
go back to reference Brentjens RJ et al (2003) Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 9(3):279–286PubMedCrossRef Brentjens RJ et al (2003) Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 9(3):279–286PubMedCrossRef
7.
go back to reference Goulding MD et al (1991) Pax-3, a novel murine DNA binding protein expressed during early neurogenesis. Embo J 10(5):1135–1147PubMed Goulding MD et al (1991) Pax-3, a novel murine DNA binding protein expressed during early neurogenesis. Embo J 10(5):1135–1147PubMed
8.
go back to reference Barr FG et al (1999) Predominant expression of alternative PAX3 and PAX7 forms in myogenic and neural tumor cell lines. Cancer Res 59(21):5443–5448PubMed Barr FG et al (1999) Predominant expression of alternative PAX3 and PAX7 forms in myogenic and neural tumor cell lines. Cancer Res 59(21):5443–5448PubMed
9.
go back to reference Parker CJ et al (2004) Expression of PAX 3 alternatively spliced transcripts and identification of two new isoforms in human tumors of neural crest origin. Int J Cancer 108(2):314–320PubMedCrossRef Parker CJ et al (2004) Expression of PAX 3 alternatively spliced transcripts and identification of two new isoforms in human tumors of neural crest origin. Int J Cancer 108(2):314–320PubMedCrossRef
10.
go back to reference Muratovska A et al (2003) Paired-Box genes are frequently expressed in cancer and often required for cancer cell survival. Oncogene 22(39):7989–7997PubMedCrossRef Muratovska A et al (2003) Paired-Box genes are frequently expressed in cancer and often required for cancer cell survival. Oncogene 22(39):7989–7997PubMedCrossRef
11.
go back to reference Galili N et al (1993) Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nat Genet 5(3):230–235PubMedCrossRef Galili N et al (1993) Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nat Genet 5(3):230–235PubMedCrossRef
12.
go back to reference Barr FG et al (1996) In vivo amplification of the PAX3-FKHR and PAX7-FKHR fusion genes in alveolar rhabdomyosarcoma. Hum Mol Genet 5(1):15–21PubMedCrossRef Barr FG et al (1996) In vivo amplification of the PAX3-FKHR and PAX7-FKHR fusion genes in alveolar rhabdomyosarcoma. Hum Mol Genet 5(1):15–21PubMedCrossRef
13.
go back to reference Xia SJ, Barr FG (2004) Analysis of the transforming and growth suppressive activities of the PAX3-FKHR oncoprotein. Oncogene 23(41):6864–6871PubMedCrossRef Xia SJ, Barr FG (2004) Analysis of the transforming and growth suppressive activities of the PAX3-FKHR oncoprotein. Oncogene 23(41):6864–6871PubMedCrossRef
14.
go back to reference Scheidler S et al (1996) The hybrid PAX3-FKHR fusion protein of alveolar rhabdomyosarcoma transforms fibroblasts in culture. Proc Natl Acad Sci USA 93(18):9805–9809PubMedCrossRef Scheidler S et al (1996) The hybrid PAX3-FKHR fusion protein of alveolar rhabdomyosarcoma transforms fibroblasts in culture. Proc Natl Acad Sci USA 93(18):9805–9809PubMedCrossRef
15.
go back to reference Keller C et al (2004) Pax3:Fkhr interferes with embryonic Pax3 and Pax7 function: implications for alveolar rhabdomyosarcoma cell of origin. Genes Dev 18(21):2608–2613PubMedCrossRef Keller C et al (2004) Pax3:Fkhr interferes with embryonic Pax3 and Pax7 function: implications for alveolar rhabdomyosarcoma cell of origin. Genes Dev 18(21):2608–2613PubMedCrossRef
16.
go back to reference Bernasconi M et al (1996) Induction of apoptosis in rhabdomyosarcoma cells through down-regulation of PAX proteins. Proc Natl Acad Sci USA 93(23):13164–13169PubMedCrossRef Bernasconi M et al (1996) Induction of apoptosis in rhabdomyosarcoma cells through down-regulation of PAX proteins. Proc Natl Acad Sci USA 93(23):13164–13169PubMedCrossRef
17.
go back to reference Scholl FA et al (2001) PAX3 is expressed in human melanomas and contributes to tumor cell survival. Cancer Res 61(3):823–826PubMed Scholl FA et al (2001) PAX3 is expressed in human melanomas and contributes to tumor cell survival. Cancer Res 61(3):823–826PubMed
18.
go back to reference Sadovnikova E et al (1998) Generation of human tumor-reactive cytotoxic T cells against peptides presented by non-self HLA class I molecules. Eur J Immunol 28(1):193–200PubMedCrossRef Sadovnikova E et al (1998) Generation of human tumor-reactive cytotoxic T cells against peptides presented by non-self HLA class I molecules. Eur J Immunol 28(1):193–200PubMedCrossRef
19.
go back to reference Evans R et al (1993) Intratumor gene expression after adoptive immunotherapy in a murine tumor model Regulation of messenger RNA levels associated with the differential expansion of tumor-infiltrating lymphocytes. J Immunol 150(1):177–184PubMed Evans R et al (1993) Intratumor gene expression after adoptive immunotherapy in a murine tumor model Regulation of messenger RNA levels associated with the differential expansion of tumor-infiltrating lymphocytes. J Immunol 150(1):177–184PubMed
20.
go back to reference Nabarro S et al (2005) Coordinated oncogenic transformation and inhibition of host immune responses by the PAX3-FKHR fusion oncoprotein. J Exp Med 202(10):1399–1410PubMedCrossRef Nabarro S et al (2005) Coordinated oncogenic transformation and inhibition of host immune responses by the PAX3-FKHR fusion oncoprotein. J Exp Med 202(10):1399–1410PubMedCrossRef
21.
go back to reference Kojima Y et al (2002) Adjuvant effect of multi-CpG motifs on an HIV-1 DNA vaccine. Vaccine 20(23–24):2857–2865PubMedCrossRef Kojima Y et al (2002) Adjuvant effect of multi-CpG motifs on an HIV-1 DNA vaccine. Vaccine 20(23–24):2857–2865PubMedCrossRef
22.
go back to reference Himoudi N et al (2002) Comparative vaccine studies in HLA-A2.1-transgenic mice reveal a clustered organization of epitopes presented in hepatitis C virus natural infection. J Virol 76(24):12735–12746PubMedCrossRef Himoudi N et al (2002) Comparative vaccine studies in HLA-A2.1-transgenic mice reveal a clustered organization of epitopes presented in hepatitis C virus natural infection. J Virol 76(24):12735–12746PubMedCrossRef
23.
go back to reference Schultze JL et al (1997) CD40-activated human B cells: an alternative source of highly efficient antigen presenting cells to generate autologous antigen-specific T cells for adoptive immunotherapy. J Clin Invest 100(11):2757–2765PubMedCrossRef Schultze JL et al (1997) CD40-activated human B cells: an alternative source of highly efficient antigen presenting cells to generate autologous antigen-specific T cells for adoptive immunotherapy. J Clin Invest 100(11):2757–2765PubMedCrossRef
24.
go back to reference WilleReece U et al (2006) Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primates. J Exp Med 203(5):1249–1258CrossRef WilleReece U et al (2006) Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primates. J Exp Med 203(5):1249–1258CrossRef
25.
go back to reference Speiser DE et al (2005) Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest 115(3):739–746PubMedCrossRef Speiser DE et al (2005) Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest 115(3):739–746PubMedCrossRef
26.
go back to reference May KF Jr et al (2002) Anti-4-1BB monoclonal antibody enhances rejection of large tumor burden by promoting survival but not clonal expansion of tumor-specific CD8+ T cells. Cancer Res 62(12):3459–3465PubMed May KF Jr et al (2002) Anti-4-1BB monoclonal antibody enhances rejection of large tumor burden by promoting survival but not clonal expansion of tumor-specific CD8+ T cells. Cancer Res 62(12):3459–3465PubMed
27.
go back to reference Hanson HL et al (2000) Eradication of established tumors by CD8+ T cell adoptive immunotherapy. Immunity 13(2):265–276PubMedCrossRef Hanson HL et al (2000) Eradication of established tumors by CD8+ T cell adoptive immunotherapy. Immunity 13(2):265–276PubMedCrossRef
28.
go back to reference Klebanoff CA et al (2004) IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci USA 101(7):1969–1974PubMedCrossRef Klebanoff CA et al (2004) IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci USA 101(7):1969–1974PubMedCrossRef
29.
go back to reference Ko K et al (2005) Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3 + CD25 + CD4 + regulatory T cells. J Exp Med 202(7):885–891PubMedCrossRef Ko K et al (2005) Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3 + CD25 + CD4 +  regulatory T cells. J Exp Med 202(7):885–891PubMedCrossRef
30.
go back to reference Gattinoni L et al (2005) Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 202(7):907–912PubMedCrossRef Gattinoni L et al (2005) Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 202(7):907–912PubMedCrossRef
31.
go back to reference Nakajima H et al (1997) The common cytokine receptor gamma chain plays an essential role in regulating lymphoid homeostasis. J Exp Med 185(2):189–195PubMedCrossRef Nakajima H et al (1997) The common cytokine receptor gamma chain plays an essential role in regulating lymphoid homeostasis. J Exp Med 185(2):189–195PubMedCrossRef
32.
go back to reference Schluns KS et al (2000) Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat Immunol 1(5):426–432PubMedCrossRef Schluns KS et al (2000) Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat Immunol 1(5):426–432PubMedCrossRef
33.
go back to reference Prlic M, Lefrancois L, Jameson SC (2002) Multiple choices: regulation of memory CD8 T cell generation and homeostasis by interleukin (IL)-7 and IL-15. J Exp Med 195(12):F49–F52PubMedCrossRef Prlic M, Lefrancois L, Jameson SC (2002) Multiple choices: regulation of memory CD8 T cell generation and homeostasis by interleukin (IL)-7 and IL-15. J Exp Med 195(12):F49–F52PubMedCrossRef
34.
go back to reference Judge AD et al (2002) Interleukin 15 controls both proliferation and survival of a subset of memory-phenotype CD8 (+) T cells. J Exp Med 196(7):935–946PubMedCrossRef Judge AD et al (2002) Interleukin 15 controls both proliferation and survival of a subset of memory-phenotype CD8 (+) T cells. J Exp Med 196(7):935–946PubMedCrossRef
35.
go back to reference Zeng R et al (2005) Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. J Exp Med 201(1):139–148PubMedCrossRef Zeng R et al (2005) Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. J Exp Med 201(1):139–148PubMedCrossRef
36.
go back to reference Loser K et al (2005) An important role of CD80/CD86-CTLA-4 signaling during photocarcinogenesis in mice. J Immunol 174(9):5298–5305PubMed Loser K et al (2005) An important role of CD80/CD86-CTLA-4 signaling during photocarcinogenesis in mice. J Immunol 174(9):5298–5305PubMed
37.
go back to reference Mokyr MB et al (1998) Realization of the therapeutic potential of CTLA-4 blockade in low-dose chemotherapy-treated tumor-bearing mice. Cancer Res 58(23):5301–5304PubMed Mokyr MB et al (1998) Realization of the therapeutic potential of CTLA-4 blockade in low-dose chemotherapy-treated tumor-bearing mice. Cancer Res 58(23):5301–5304PubMed
38.
go back to reference Bristol JA, Schlom J, Abrams SI (1999) Persistence, immune specificity, and functional ability of murine mutant ras epitope-specific CD4 (+) and CD8 (+) T lymphocytes following in vivo adoptive transfer. Cell Immunol 194(1):78–89PubMedCrossRef Bristol JA, Schlom J, Abrams SI (1999) Persistence, immune specificity, and functional ability of murine mutant ras epitope-specific CD4 (+) and CD8 (+) T lymphocytes following in vivo adoptive transfer. Cell Immunol 194(1):78–89PubMedCrossRef
39.
go back to reference van den Broeke LT et al (2006) Identification and epitope enhancement of a PAX-FKHR fusion protein breakpoint epitope in alveolar rhabdomyosarcoma cells created by a tumorigenic chromosomal translocation inducing CTL capable of lysing human tumors. Cancer Res 66(3):1818–1823PubMedCrossRef van den Broeke LT et al (2006) Identification and epitope enhancement of a PAX-FKHR fusion protein breakpoint epitope in alveolar rhabdomyosarcoma cells created by a tumorigenic chromosomal translocation inducing CTL capable of lysing human tumors. Cancer Res 66(3):1818–1823PubMedCrossRef
40.
go back to reference Rodeberg DA et al (2006) Generation of tumoricidal PAX3 peptide antigen specific cytotoxic T lymphocytes. Int J Cancer 119(1):126–132PubMedCrossRef Rodeberg DA et al (2006) Generation of tumoricidal PAX3 peptide antigen specific cytotoxic T lymphocytes. Int J Cancer 119(1):126–132PubMedCrossRef
41.
go back to reference Davis RJ, Barr FG (1997) Fusion genes resulting from alternative chromosomal translocations are overexpressed by gene-specific mechanisms in alveolar rhabdomyosarcoma. Proc Natl Acad Sci USA 94(15):8047–8051PubMedCrossRef Davis RJ, Barr FG (1997) Fusion genes resulting from alternative chromosomal translocations are overexpressed by gene-specific mechanisms in alveolar rhabdomyosarcoma. Proc Natl Acad Sci USA 94(15):8047–8051PubMedCrossRef
42.
go back to reference Goldstein M et al (2006) Novel genes implicated in embryonal, alveolar, and pleomorphic rhabdomyosarcoma: a cytogenetic and molecular analysis of primary tumors. Neoplasia 8(5):332–343PubMedCrossRef Goldstein M et al (2006) Novel genes implicated in embryonal, alveolar, and pleomorphic rhabdomyosarcoma: a cytogenetic and molecular analysis of primary tumors. Neoplasia 8(5):332–343PubMedCrossRef
43.
go back to reference He SJ et al (2005) Transfection of melanoma cells with antisense PAX3 oligonucleotides additively complements cisplatin-induced cytotoxicity. Mol Cancer Ther 4(6):996–1003PubMedCrossRef He SJ et al (2005) Transfection of melanoma cells with antisense PAX3 oligonucleotides additively complements cisplatin-induced cytotoxicity. Mol Cancer Ther 4(6):996–1003PubMedCrossRef
Metadata
Title
Development of anti-PAX3 immune responses; a target for cancer immunotherapy
Authors
Nourredine Himoudi
Steven Nabarro
Mengyong Yan
Kimberly Gilmour
Adrian J. Thrasher
John Anderson
Publication date
01-09-2007
Publisher
Springer-Verlag
Published in
Cancer Immunology, Immunotherapy / Issue 9/2007
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-007-0294-3

Other articles of this Issue 9/2007

Cancer Immunology, Immunotherapy 9/2007 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