Skip to main content
Erschienen in: Medical Oncology 4/2012

01.12.2012 | Original Paper

Adjuvant vaccination with melanoma antigen-pulsed dendritic cells in stage III melanoma patients

verfasst von: Sergiusz Markowicz, Zbigniew I. Nowecki, Piotr Rutkowski, Andrzej W. Lipkowski, Marzena Biernacka, Anna Jakubowska-Mucka, Tomasz Switaj, Aleksandra Misicka, Henryk Skurzak, Hanna Polowniak-Pracka, Jan Walewski

Erschienen in: Medical Oncology | Ausgabe 4/2012

Einloggen, um Zugang zu erhalten

Abstract

Dendritic cells may be successfully used to induce in vivo-specific anti-tumor responses when combined with the appropriate antigen in the appropriate context. The purpose of this study was to evaluate efficacy of peptide-loaded DC vaccine in high-risk stage III melanoma patients after lymph node dissection (LND). HLA-A2+, -A1+, or -A3+ melanoma patients (N = 22), stage III, N1b-N3, received 5–16 (median: 11) DC vaccines loaded with MHC class-I-restricted melanoma peptides respective to the patient’s haplotype, and with autologous tumor lysate, if available. Vaccinated patients were matched to unvaccinated stage III controls (22 of 869) by sex, number of metastatic lymph nodes, extracapsular involvement, LND type, Breslow stage, and ulceration. Vaccination elicited cutaneous delayed-type hypersensitivity (DTH) or/and IFN-γ-producing CD8+ cell response to melanoma peptides in 15 of 22 patients. Three-year overall survival (OS) rate was 68.2% in the vaccinated group versus 25.7% in the control group, P value accounting for matching: 0.0290. In a Cox regression model, hazard ratio (HR) for death of vaccinated patients was 0.31 [95% confidence interval (CI): 0.10–0.94]. The corresponding values for 3-year disease-free survival rate were 40.9 versus 14.5%, P = 0.1083; HR of recurrence for vaccinated, 0.46 (95% CI: 0.18–1.22). There was no grade >1 toxicity. The DC/peptide vaccine was well tolerated and elicited immune responses to melanoma antigens. Vaccinated patients had significantly longer OS after LND than the matched controls, but a significant improvement in the primary endpoint DFS was not achieved.
Literatur
1.
Zurück zum Zitat Mehta-Damani A, Markowicz S, Engleman EG. Generation of antigen-specific CD8+ CTLs from naive precursors. J Immunol. 1994;153:996–1003.PubMed Mehta-Damani A, Markowicz S, Engleman EG. Generation of antigen-specific CD8+ CTLs from naive precursors. J Immunol. 1994;153:996–1003.PubMed
2.
Zurück zum Zitat Mehta-Damani A, Markowicz S, Engleman EG. Generation of antigen-specific CD4+ T cell lines from naive precursors. Eur J Immunol. 1995;25:1206–11.PubMedCrossRef Mehta-Damani A, Markowicz S, Engleman EG. Generation of antigen-specific CD4+ T cell lines from naive precursors. Eur J Immunol. 1995;25:1206–11.PubMedCrossRef
3.
Zurück zum Zitat Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996;2:52–8.PubMedCrossRef Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996;2:52–8.PubMedCrossRef
4.
Zurück zum Zitat Timmerman JM, Czerwinski DK, Davis TA, et al. Idiotype pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood. 2002;99:1517–26.PubMedCrossRef Timmerman JM, Czerwinski DK, Davis TA, et al. Idiotype pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood. 2002;99:1517–26.PubMedCrossRef
5.
Zurück zum Zitat Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22.PubMedCrossRef Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22.PubMedCrossRef
6.
Zurück zum Zitat Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328–32.PubMedCrossRef Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328–32.PubMedCrossRef
7.
Zurück zum Zitat Banchereau J, Palucka AK, Dhodapkar M, et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–8.PubMed Banchereau J, Palucka AK, Dhodapkar M, et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–8.PubMed
8.
Zurück zum Zitat O’Rourke MG, Johnson M, Lanagan C, et al. Durable complete clinical responses in a phase I/II trial using an autologous melanoma cell/dendritic cell vaccine. Cancer Immunol Immunother. 2003;52:387–95.PubMed O’Rourke MG, Johnson M, Lanagan C, et al. Durable complete clinical responses in a phase I/II trial using an autologous melanoma cell/dendritic cell vaccine. Cancer Immunol Immunother. 2003;52:387–95.PubMed
9.
Zurück zum Zitat O’Rourke MG, Johnson MK, Lanagan CM, et al. Dendritic cell immunotherapy for stage IV melanoma. Melanoma Res. 2007;17:316–22.PubMedCrossRef O’Rourke MG, Johnson MK, Lanagan CM, et al. Dendritic cell immunotherapy for stage IV melanoma. Melanoma Res. 2007;17:316–22.PubMedCrossRef
10.
Zurück zum Zitat Schadendorf D, Ugurel S, Schuler-Thurner B, et al. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol. 2006;17:563–70.PubMedCrossRef Schadendorf D, Ugurel S, Schuler-Thurner B, et al. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol. 2006;17:563–70.PubMedCrossRef
11.
Zurück zum Zitat Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P, Svane IM. Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother. 2009;58:1–14.PubMedCrossRef Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P, Svane IM. Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother. 2009;58:1–14.PubMedCrossRef
12.
Zurück zum Zitat Schadendorf D, Algarra SM, Bastholt L, et al. Immunotherapy of distant metastatic disease. Ann Oncol. 2009;20(Suppl 6):vi41–50.PubMedCrossRef Schadendorf D, Algarra SM, Bastholt L, et al. Immunotherapy of distant metastatic disease. Ann Oncol. 2009;20(Suppl 6):vi41–50.PubMedCrossRef
13.
Zurück zum Zitat Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15.PubMedCrossRef Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15.PubMedCrossRef
14.
Zurück zum Zitat Koos D, Josephs SF, Alexandrescu DT, et al. Tumor vaccines in 2010: need for integration. Cell Immunol. 2010;263:138–47.PubMedCrossRef Koos D, Josephs SF, Alexandrescu DT, et al. Tumor vaccines in 2010: need for integration. Cell Immunol. 2010;263:138–47.PubMedCrossRef
15.
Zurück zum Zitat Lesterhuis WJ, Aarntzen EH, De Vries IJ, et al. Dendritic cell vaccines in melanoma: from promise to proof? Crit Rev Oncol Hematol. 2008;66:118–34.PubMedCrossRef Lesterhuis WJ, Aarntzen EH, De Vries IJ, et al. Dendritic cell vaccines in melanoma: from promise to proof? Crit Rev Oncol Hematol. 2008;66:118–34.PubMedCrossRef
16.
Zurück zum Zitat Letsch A, Keilholz U, Fluck M, et al. Peptide vaccination after repeated resection of metastases can induce a prolonged relapse-free interval in melanoma patients. Int J Cancer. 2005;114:936–41.PubMedCrossRef Letsch A, Keilholz U, Fluck M, et al. Peptide vaccination after repeated resection of metastases can induce a prolonged relapse-free interval in melanoma patients. Int J Cancer. 2005;114:936–41.PubMedCrossRef
17.
Zurück zum Zitat Tuettenberg A, Becker C, Huter E, Knop J, Enk AH, Jonuleit H. Induction of strong and persistent MelanA/MART-1-specific immune responses by adjuvant dendritic cell-based vaccination of stage II melanoma patients. Int J Cancer. 2006;118:2617–27.PubMedCrossRef Tuettenberg A, Becker C, Huter E, Knop J, Enk AH, Jonuleit H. Induction of strong and persistent MelanA/MART-1-specific immune responses by adjuvant dendritic cell-based vaccination of stage II melanoma patients. Int J Cancer. 2006;118:2617–27.PubMedCrossRef
18.
Zurück zum Zitat Testori A, Rutkowski P, Marsden J, et al. Surgery and radiotherapy in the treatment of cutaneous melanoma. Ann Oncol. 2009;20(Suppl 6):vi22–9.PubMedCrossRef Testori A, Rutkowski P, Marsden J, et al. Surgery and radiotherapy in the treatment of cutaneous melanoma. Ann Oncol. 2009;20(Suppl 6):vi22–9.PubMedCrossRef
19.
Zurück zum Zitat Mortarini R, Anichini A, Di Nicola M, et al. Autologous dendritic cells derived from CD34+ progenitors and from monocytes are not functionally equivalent antigen-presenting cells in the induction of MelanA/Mart-127-35-specific CTLs from peripheral blood lymphocytes of melanoma patients with low frequency of CTL precursors. Cancer Res. 1997;57:5534–41.PubMed Mortarini R, Anichini A, Di Nicola M, et al. Autologous dendritic cells derived from CD34+ progenitors and from monocytes are not functionally equivalent antigen-presenting cells in the induction of MelanA/Mart-127-35-specific CTLs from peripheral blood lymphocytes of melanoma patients with low frequency of CTL precursors. Cancer Res. 1997;57:5534–41.PubMed
20.
Zurück zum Zitat Nowecki ZI, Rutkowski P, Michej W. The survival benefit to patients with positive sentinel node melanoma after completion lymph node dissection may be limited to the subgroup with a primary lesion Breslow thickness greater than 1.0 and less than or equal to 4 mm (pT2-pT3). Ann Surg Oncol. 2008;15:2223–34.PubMedCrossRef Nowecki ZI, Rutkowski P, Michej W. The survival benefit to patients with positive sentinel node melanoma after completion lymph node dissection may be limited to the subgroup with a primary lesion Breslow thickness greater than 1.0 and less than or equal to 4 mm (pT2-pT3). Ann Surg Oncol. 2008;15:2223–34.PubMedCrossRef
21.
Zurück zum Zitat Klein JP, Moeschberger ML. Survival analysis: techniques for censored and truncated data. New York: Springer; 1997. p. 282–6. Klein JP, Moeschberger ML. Survival analysis: techniques for censored and truncated data. New York: Springer; 1997. p. 282–6.
22.
Zurück zum Zitat Thompson LW, Garbee CF, Hibbitts S, et al. Competition among peptides in melanoma vaccines for binding to MHC molecules. J Immunother. 2004;27:425–31.PubMedCrossRef Thompson LW, Garbee CF, Hibbitts S, et al. Competition among peptides in melanoma vaccines for binding to MHC molecules. J Immunother. 2004;27:425–31.PubMedCrossRef
23.
Zurück zum Zitat Slingluff CL Jr, Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res. 2007;13:6386–95.PubMedCrossRef Slingluff CL Jr, Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res. 2007;13:6386–95.PubMedCrossRef
24.
Zurück zum Zitat Tsai V, Southwood S, Sidney J, et al. Identification of subdominant CTL epitopes of the gp100 melanoma-associated tumor antigen by primary in vitro immunization with peptide-pulsed dendritic cells. J Immunol. 1997;158:1796–802.PubMed Tsai V, Southwood S, Sidney J, et al. Identification of subdominant CTL epitopes of the gp100 melanoma-associated tumor antigen by primary in vitro immunization with peptide-pulsed dendritic cells. J Immunol. 1997;158:1796–802.PubMed
25.
Zurück zum Zitat Markowicz S, Engleman EG. Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest. 1990;85:955–61.PubMedCrossRef Markowicz S, Engleman EG. Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest. 1990;85:955–61.PubMedCrossRef
26.
Zurück zum Zitat Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. GM-CSF and TNF-α cooperate in the generation of dendritic Langerhans cells. Nature. 1992;360:258–61.PubMedCrossRef Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. GM-CSF and TNF-α cooperate in the generation of dendritic Langerhans cells. Nature. 1992;360:258–61.PubMedCrossRef
27.
Zurück zum Zitat Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP, Rivoltini L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol. 2007;18:226–32.PubMedCrossRef Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP, Rivoltini L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol. 2007;18:226–32.PubMedCrossRef
28.
Zurück zum Zitat Eggermont AM. Immunostimulation versus immunosuppression after multiple vaccinations: the woes of therapeutic vaccine development. Clin Cancer Res. 2009;15:6745–7.PubMedCrossRef Eggermont AM. Immunostimulation versus immunosuppression after multiple vaccinations: the woes of therapeutic vaccine development. Clin Cancer Res. 2009;15:6745–7.PubMedCrossRef
29.
Zurück zum Zitat Faries MB, Hsueh EC, Ye X, Hoban M, Morton DL. Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine. Clin Cancer Res. 2009;15:7029–35.PubMedCrossRef Faries MB, Hsueh EC, Ye X, Hoban M, Morton DL. Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine. Clin Cancer Res. 2009;15:7029–35.PubMedCrossRef
30.
Zurück zum Zitat Slingluff CL Jr, Petroni GR, Olson WC, et al. Effect of granulocyte/macrophage colony-stimulating factor on circulating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine: outcome of a multicenter randomized trial. Clin Cancer Res. 2009;15:7036–44.PubMedCrossRef Slingluff CL Jr, Petroni GR, Olson WC, et al. Effect of granulocyte/macrophage colony-stimulating factor on circulating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine: outcome of a multicenter randomized trial. Clin Cancer Res. 2009;15:7036–44.PubMedCrossRef
31.
Zurück zum Zitat Gunturu KS, Meehan KR, Mackenzie TA, et al. Cytokine working group study of lymphodepleting chemotherapy, interleukin-2, and granulocyte-macrophage colony-stimulating factor in patients with metastatic melanoma: clinical outcomes and peripheral-blood cell recovery. J Clin Oncol. 2010;28:1196–202.PubMedCrossRef Gunturu KS, Meehan KR, Mackenzie TA, et al. Cytokine working group study of lymphodepleting chemotherapy, interleukin-2, and granulocyte-macrophage colony-stimulating factor in patients with metastatic melanoma: clinical outcomes and peripheral-blood cell recovery. J Clin Oncol. 2010;28:1196–202.PubMedCrossRef
32.
Zurück zum Zitat Santiago-Schwarz F, Belilos E, Diamond B, Carsons SE. TNF in combination with GM-CSF enhances the differentiation of neonatal cord blood stem cells into dendritic cells and macrophages. J Leuk Biol. 1992;52:274–81. Santiago-Schwarz F, Belilos E, Diamond B, Carsons SE. TNF in combination with GM-CSF enhances the differentiation of neonatal cord blood stem cells into dendritic cells and macrophages. J Leuk Biol. 1992;52:274–81.
33.
Zurück zum Zitat Reid CDL, Stackpoole A, Meager A, Tikerpae J. Interactions of tumor necrosis factor with granulocyte-macrophage colony-stimulating factor and other cytokines in the regulation of dendritic cell growth in vitro from early bipotent CD34+ progenitors in human bone marrow. J Immunol. 1992;149:2681–8.PubMed Reid CDL, Stackpoole A, Meager A, Tikerpae J. Interactions of tumor necrosis factor with granulocyte-macrophage colony-stimulating factor and other cytokines in the regulation of dendritic cell growth in vitro from early bipotent CD34+ progenitors in human bone marrow. J Immunol. 1992;149:2681–8.PubMed
34.
Zurück zum Zitat Young JW, Szabolcs P, Moore MAS. Identification of dendritic cell colony-forming units among normal human CD34+ bone marrow progenitors that are expanded by c-kit-ligand and yield pure dendritic cell colonies in the presence of granulocyte/macrophage colony-stimulating factor and tumor necrosis factor α. J Exp Med. 1995;182:1111–9.PubMedCrossRef Young JW, Szabolcs P, Moore MAS. Identification of dendritic cell colony-forming units among normal human CD34+ bone marrow progenitors that are expanded by c-kit-ligand and yield pure dendritic cell colonies in the presence of granulocyte/macrophage colony-stimulating factor and tumor necrosis factor α. J Exp Med. 1995;182:1111–9.PubMedCrossRef
35.
Zurück zum Zitat Siena S, Di Nicola M, Bregni M, et al. Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy. Exp Hematol. 1995;23:1463–71.PubMed Siena S, Di Nicola M, Bregni M, et al. Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy. Exp Hematol. 1995;23:1463–71.PubMed
36.
Zurück zum Zitat Markowicz S, Walewski J, Kawecki A. Isolation and characteristic of dendritic cell progenitors from the bone marrow of the Hodgkin’s disease patients. In: Banchereau J, Schmitt D, editors. Dendritic cells in fundamental and clinical immunology, vol. 2. New York: Plenum Press; 1995. p. 553–5.CrossRef Markowicz S, Walewski J, Kawecki A. Isolation and characteristic of dendritic cell progenitors from the bone marrow of the Hodgkin’s disease patients. In: Banchereau J, Schmitt D, editors. Dendritic cells in fundamental and clinical immunology, vol. 2. New York: Plenum Press; 1995. p. 553–5.CrossRef
37.
Zurück zum Zitat Bernhard H, Disks ML, Heimfeld S, Hand S, Gralow JR, Cheever MA. Generation of immunostimulatory dendritic cells from human CD34+ hematopoietic progenitor cells of the bone marrow and peripheral blood. Cancer Res. 1995;55:1099–104.PubMed Bernhard H, Disks ML, Heimfeld S, Hand S, Gralow JR, Cheever MA. Generation of immunostimulatory dendritic cells from human CD34+ hematopoietic progenitor cells of the bone marrow and peripheral blood. Cancer Res. 1995;55:1099–104.PubMed
38.
Zurück zum Zitat Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179:1109–18.PubMedCrossRef Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179:1109–18.PubMedCrossRef
39.
Zurück zum Zitat Banerjee DK, Dhodapkar MV, Matayeva E, Steinman RM, Dhodapkar KM. Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients. Blood. 2006;108:2655–61.PubMedCrossRef Banerjee DK, Dhodapkar MV, Matayeva E, Steinman RM, Dhodapkar KM. Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients. Blood. 2006;108:2655–61.PubMedCrossRef
40.
Zurück zum Zitat Hayashi Y, Hoon DS, Park MS, Terasaki PI, Foshag LJ, Morton DL. Induction of CD4+ cytotoxic T cells by sensitization with allogeneic melanomas bearing shared or cross-reactive HLA-A. Cell Immunol. 1992;139:411–25.PubMedCrossRef Hayashi Y, Hoon DS, Park MS, Terasaki PI, Foshag LJ, Morton DL. Induction of CD4+ cytotoxic T cells by sensitization with allogeneic melanomas bearing shared or cross-reactive HLA-A. Cell Immunol. 1992;139:411–25.PubMedCrossRef
41.
Zurück zum Zitat Robbins PF, el-Gamil M, Li YF, et al. Cloning of a new gene encoding an antigen recognized by melanoma-specific HLA-A24-restricted tumor-infiltrating lymphocytes. J Immunol. 1995;154:5944–50.PubMed Robbins PF, el-Gamil M, Li YF, et al. Cloning of a new gene encoding an antigen recognized by melanoma-specific HLA-A24-restricted tumor-infiltrating lymphocytes. J Immunol. 1995;154:5944–50.PubMed
42.
Zurück zum Zitat Darrow TL, Abdel-Wahab Z, Quinn-Allen MA, Seigler HF. Recognition and lysis of human melanoma by a CD3+, CD4+, CD8− T-cell clone restricted by HLA-A2. Cell Immunol. 1996;172:52–9.PubMedCrossRef Darrow TL, Abdel-Wahab Z, Quinn-Allen MA, Seigler HF. Recognition and lysis of human melanoma by a CD3+, CD4+, CD8 T-cell clone restricted by HLA-A2. Cell Immunol. 1996;172:52–9.PubMedCrossRef
43.
Zurück zum Zitat Nishimura MI, Avichezer D, Custer MC, et al. MHC class I-restricted recognition of a melanoma antigen by a human CD4+ tumor infiltrating lymphocyte. Cancer Res. 1999;59:6230–8.PubMed Nishimura MI, Avichezer D, Custer MC, et al. MHC class I-restricted recognition of a melanoma antigen by a human CD4+ tumor infiltrating lymphocyte. Cancer Res. 1999;59:6230–8.PubMed
44.
Zurück zum Zitat Voelkl S, Moore TV, Rehli M, Nishimura MI, Mackensen A, Fischer K. Characterization of MHC class-I restricted TCRalphabeta+ CD4− CD8− double negative T cells recognizing the gp100 antigen from a melanoma patient after gp100 vaccination. Cancer Immunol Immunother. 2009;58:709–18.PubMedCrossRef Voelkl S, Moore TV, Rehli M, Nishimura MI, Mackensen A, Fischer K. Characterization of MHC class-I restricted TCRalphabeta+ CD4 CD8 double negative T cells recognizing the gp100 antigen from a melanoma patient after gp100 vaccination. Cancer Immunol Immunother. 2009;58:709–18.PubMedCrossRef
45.
Zurück zum Zitat Sosman JA, Unger JM, Liu PY, et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol. 2002;8:2067–75.CrossRef Sosman JA, Unger JM, Liu PY, et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol. 2002;8:2067–75.CrossRef
46.
Zurück zum Zitat Wolchok JD, Weber JS, Hamid O, et al. Ipilimumab efficacy and safety in patients with advanced melanoma: a retrospective analysis of HLA subtype from four trials. Cancer Immun. 2010;10:9–14.PubMed Wolchok JD, Weber JS, Hamid O, et al. Ipilimumab efficacy and safety in patients with advanced melanoma: a retrospective analysis of HLA subtype from four trials. Cancer Immun. 2010;10:9–14.PubMed
47.
Zurück zum Zitat Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedCrossRef Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedCrossRef
Metadaten
Titel
Adjuvant vaccination with melanoma antigen-pulsed dendritic cells in stage III melanoma patients
verfasst von
Sergiusz Markowicz
Zbigniew I. Nowecki
Piotr Rutkowski
Andrzej W. Lipkowski
Marzena Biernacka
Anna Jakubowska-Mucka
Tomasz Switaj
Aleksandra Misicka
Henryk Skurzak
Hanna Polowniak-Pracka
Jan Walewski
Publikationsdatum
01.12.2012
Verlag
Springer US
Erschienen in
Medical Oncology / Ausgabe 4/2012
Print ISSN: 1357-0560
Elektronische ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-012-0168-1

Weitere Artikel der Ausgabe 4/2012

Medical Oncology 4/2012 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.