115
Views
36
CrossRef citations to date
0
Altmetric
Review

Vaccine immunotherapy in breast cancer treatment: promising, but still early

, , , , &
Pages 1225-1241 | Published online: 10 Jan 2014

References

  • Schaefer NG, Pestalozzi BC, Knuth A, Renner C. Potential use of humanized antibodies in the treatment of breast cancer. Expert Rev. Anticancer Ther.6(7), 1065–1074 (2006).
  • Acres B, Limacher JM, Bonnefoy J. Discovery and development of therapeutic cancer vaccines. Curr. Opin. Drug Discov. Devel.10(2), 185–192 (2007).
  • Nencioni A, Gruenback F, Patrone F, Brossart P. Anticancer vaccination strategies. Ann. Oncol.15 (Suppl. 4), 153–160 (2004).
  • Lyerly HK. Quantitating cellular immune response to cancer vaccines. Ann. Oncol.30 (3 Suppl. 8), 9–16 (2003).
  • Emens LA, Reilly RT, Jaffee EM. Augmenting the potency of breast cancer vaccines: combined modality immunotherapy. Breast Dis.20, 13–24 (2004).
  • Ko BK, Kawano K, Murray JL et al. Clinical studies of vaccines targeting breast cancer. Clin. Cancer Res.9(9), 3222–3234 (2003).
  • Curigliano G, Spitaleri G, Pietri E et al. Breast cancer vaccines: a clinical reality or fairy tale? Ann. Oncol.17(5), 750–762 (2006).
  • Pegram M, Hsu S, Lewis G et al. Inhibitory effects of combinations of HER2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers. Oncogene18(13), 2241–2251 (1999).
  • Ioannides CG, Ioannides MG, O’Brian CA. T-cell recognition of oncogene products: a new strategy for immunotherapy. Mol. Carcinog.6(2), 77–82 (1992).
  • Disis ML, Pupa S, Gralow JR, Dittadi R, Menard S, Cheever MA. High-titre HER2/neu protein-specific antibody can be detected in patients with early-stage breast cancer. J. Clin. Oncol.15(11), 3363–3367 (1997).
  • Baxevanis CN, Sotiropoulou PA, Sotiriadou NN, Papamichail M. Immunobiology of HER2/neu oncoprotein and its potential application in cancer immunotherapy. Cancer Immunol. Immunother.53(3), 166–175 (2004).
  • Nagata Y, Furugen R, Hiasa A et al. Peptides derived from a wild-type murine proto-oncogene c-erbB-2/HER2/neu can induce CTL and tumor suppression in syngeneic hosts. J. Immunol.159(3), 1336–1343 (1997).
  • Disis ML, Shiota FM, Cheever MA. Human HER 2/neu protein immunization circumvents tolerance to rat neu: a vaccine strategy for ‘self’ tumour antigens. J. Immunol.93(2), 192–199 (1998).
  • Disis ML, Grabstein KH, Sleath PR et al. HER2/neu peptide vaccines elicit T cell immunity to the HER2/neu protein in patients with breast or ovarian cancer. Proceedings of the 34th American Society Clinical Oncology. CA, USA, 16–19 May, 1998 (Abstract 97a).
  • Peoples GE, Goedegeburre PS, Smith R et al. Breast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu-derived peptide. Proc. Natl Acad. Sci. USA, 192, 432–436 (1995).
  • Fisk B, Blevins TL, Wharton JT et al. Identification of an immunodominant peptide of the HER2/neu proto-oncogene recognized by ovarian tumor-specific CTL lines. J. Exp. Med.181(6), 2109–2117 (1995).
  • Peoples GE, Gurney JM, Hueman MT, et al. Clinical trial results of a HER2/neu (E75) vaccine to prevent recurrence in high-risk breast cancer patients. J. Clin. Oncol.23(30), 7536–7545 (2005).
  • Zaks TZ, Rosenberg SA. Immunization with a peptide epitope (p369–377) from HER2/neu leads to peptide specific cytotoxic T lymphocytes that fail to recognize HER2/neu+ tumors. Cancer Res.58 (21), 369–377 (1998).
  • Knutson KL, Schiffman K, Cheever MA, Disis ML. Immunization of cancer patients with a HER2/neu, HLA-A2 peptide, p369–377, results in short-lived peptide-specific immunity. Clin. Cancer Res.8(5), 1014–1018 (2002).
  • Murray JL, Gillogly ME, Przepiorka D et al. Toxicity, immunogenicity, and induction of E75-specific tumor-lytic CTLs by HER2 peptide E75 (369–377) combined with granulocyte macrophage colony-stimulating factor in HLA-A2+ patients with metastatic breast and ovarian cancer. Clin. Cancer Res.8(11), 3407–3418 (2002).
  • Mittendorf EA, Storrer CE, Foley RJ et al. Evaluation of the HER2/neu-derived peptide GP2 for use in a peptide-based breast cancer vaccine trial. Cancer106(11), 2309–2317 (2006).
  • Disis ML, Cheever MA. HER2/neu oncogenic protein: issues in vaccine development. Crit. Rev. Immunol.18(1–2), 37–45 (1998).
  • Disis ML, Gooley TA, Rinn K et al. Generation of T-cell immunity to the HER2/neu protein after active immunization with HER2/neu peptide-based vaccines. J. Clin. Oncol.20(11), 2624–2632 (2002).
  • Disis ML, Goodell V, Schiffman K, Knutson KL. Humoral epitope-spreading following immunization with a HER2/neu peptide based vaccine in cancer patients. J. Clin. Immunol.24(5), 571–578 (2004).
  • Salazar LG, Fikes J, Southwood S et al. Immunization of cancer patients with HER2/neu-derived peptides demonstrating high-affinity binding to multiple class II alleles. Clin. Cancer Res.9(15), 5559–5565 (2003).
  • Disis ML, Schiffman K, Guthrie K et al. Effect of dose on immune response in patients vaccinated with an HER2/neu intracellular domain protein-based vaccine. J. Clin. Oncol.22(10), 1916–1925 (2004).
  • Knutson KL, Schiffman K, Disis ML. Immunization with a HER2/neu helper peptide vaccine generates HER2/neu CD8 T-cell immunity in cancer patients. J. Clin. Invest.107(4), 477–484 (2001).
  • Limentani S, Dorval T, White S et al. Phase I dose-escalation trial of a recombinant HER2 vaccine in patients with stage II/III HER2+ breast cancer. Proceedings of the 41th American Society Clinical Oncology. FL, USA, 13–17 May, 2005 (Abstract 2520).
  • Salazar LG, Murray JL, Disis ML, Cheever M. A Phase I vaccine trial of a HER2/neu peptide incorporated into PLG microspheres in patients with advanced stage HER2/expressing cancers. Proceedings of the 42nd American Society Clinical Oncology. MA, USA, 2–6 June, 2006 (Abstract 2572a).
  • Webster DJ, Waisman J, Macleod B et al. A Phase I/II study of a HER2/neu peptide vaccine plus concurrent trastuzumab. Proceedings of the 42nd American Society Clinical Oncology. MA, USA, 2–6 June, 2006 (Abstract 2528a).
  • Mittendorf EA, Khoo S, Storrer CE et al. Early results of a Phase I clinical trial of an Ii-Key/Her2/neu MHC class II peptide-based vaccine in breast cancer patients. Proceedings of the 42th American Society Clinical Oncology. MA, USA, 2–6 June, 2006 (Abstract 2532a).
  • McGuckin MA, Walsh MD, Hohn BG, et al. Prognostic significance of MUC-1 epithelial mucin expression in breast cancer. Hum. Pathol.26(4), 432–439 (1995).
  • Apostolopoulos V, Loveland BE, Pietersz GA, McKenzie IF. CTL in mice immunized with human mucin 1 are MHC-restricted. J. Immunol.155(11), 5089–5094 (1995).
  • Heukamp LC, van der Burg SH, Drijfhout JW et al. Identification of three non-VNTR MUC-1 derived HLA A*0201-restricted T-cell epitopes that induce protective anti-tumor immunity in HLA-A2/K(b) transgenic mice. Int. J. Cancer91(3), 385–90 (2001).
  • Tsang KY, Palena C, Gulley J et al. A human cytotoxic t-lymphocyte epitope and its agonist epitope from the nonvariable number of tandem repeat sequence of MUC-1. Clin. Cancer Res.10(6), 2139–2149 (2004).
  • Chen D, Xia J, Tanaka Y et al. Immunotherapy of spontaneous mammary carcinoma with fusions of dendritic cells and mucin 1-positive carcinoma cells. Immunology109(2), 300–307 (2003).
  • Kohlgraf KG, Gawron AJ, Higashi M. Tumor-specific immunity in MUC1. Tg mice induced by immunization with peptide vaccines from the cytoplasmatic tail of CD227 (MUC1). Cancer Immunol. Immunother.53(12), 1068–1084 (2004).
  • Brossart P, Wirths S, Stuhler G et al. Induction of cytotoxic T lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood96(9), 3102–3108 (2000).
  • Apostolopoulos V, Pietersz GA, McKenzia IFC. Oxidative/reductive conjugation of mannan to antigen selects for T1 or T2 immune response. Proc. Natl Acad. Sci. USA92, 10128–10132 (1995).
  • Apostolopoulos V, Xing PX, McKenzie IF. Murine immune response to cells transfected with human MUC1: immunization with cellular and synthetic antigens. Cancer Res.54(19), 5186–5193 (1994).
  • Zhang S, Graeber LA, Helling F et al. Augmenting the immunogenicity of synthetic MUC1 peptide vaccines in mice. Cancer Res.56(14), 3315–3319 (1996).
  • Acres RB, Hareuveni M, Balloul JM, Kieny MP. Vaccinia virus MUC1 immunization of mice: immune response and protection against the growth of murine tumors bearing the MUC1 antigen. J. Immunother.14(2), 136–143 (1993).
  • Goydos JS, Elder E, Whiteside TL et al. A Phase I trial of a synthetic mucin peptide vaccine. Induction of specific immune reactivity in patients with adenocarcinoma. J. Surg. Res.63(1), 298–304 (1996).
  • Reddish MA, MacLean GD, Koganty RR et al. Anti-MUC1 class I restricted CTLs in metastatic breast cancer patients immunized with a synthetic MUC1 peptide. Int. J. Cancer76, 817–823 (1998).
  • Karakinas V, Hwang L-A, Pearson J et al. Antibody and T cell responses of patients with adenocarcinoma immunized with Mannan-MUC1 fusion protein. J. Clin. Invest.100(11), 2783–2792 (1997).
  • MacLean GD, Reddish MA, Koganly RR, Longenecker B. Antibodies against mucin-associated sialyl-Tn epitopes correlate with survival of metastatic adenocarcinoma patients undergoing active specific immunotherapy with synthetic STn vaccine. J. Immunother.19(4), 59–68 (1996).
  • Gilewski T, Adluri S, Ragupathi G et al. Vaccination of high risk breast cancer patients with MUC-1 keyhole limpet hemocyanin conjugate plus QS-21. Clin. Cancer Res.6(5), 1693–1701 (2000).
  • Musselli C, Ragupathi G, Gilewski T et al. Reevaluation of the cellular immune response in breast cancer patients vaccinated with MUC1. Int. J. Cancer97(5), 660–667 (2002).
  • Berinstein NL. Carcinoembryonic antigen as a target for therapeutic anticancer vaccines: a review. J. Clin. Oncol.20(8), 2197–2207 (2002).
  • Tsang K, Zaremba S, Nieroda CA, et al. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia–CEA vaccine. J. Natl Cancer Inst.87(13), 982–990 (1995).
  • Morse M, Deng Y, Coleman D et al. A Phase I study of active immunotherapy with carcinoembryonic antigen peptide (CAP-1)- pulsed, autologous human cultured dendritic cells in patients with metastatic malignancies expressing carcinoembryonic antigen. Clin. Cancer Res.5(6), 1331–1338 (1999).
  • Samanci A, Yi Q, Fagerberg J et al. Pharmacological administration of granulocyte/macrophage-colony-stimulation factor is of significant importance for the induction of a strong humoral and cellular response in patients immunized with recombinant carcinoembryonic antigen. Cancer Immunol.47(3), 131–142 (1998).
  • Hodge JW, McLaughlin JP, Kantor JA, Schlom J. Diversified prime and boost protocols using recombinant vaccinia virus and recombinant nonreplicating avian pox virus to enhance T-cell immunity and antitumor responses. Vaccine15(6–7), 759–768 (1997).
  • Marshall J, Hoyer RJ, Toomey MA et al. Phase I study in advanced cancer patients of a diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses. J. Clin. Oncol.18(23), 3964–3974 (2000).
  • Von Mehren M, Arlen P, Gulley J et al. The influence of granulocyte macrophage colony-stimulating factor and prior chemotherapy on the immunological response to a vaccine (ALVAC–CEA B7.1) in patients with metastatic carcinoma. Clin. Cancer Res.7(5), 1181–1191 (2001).
  • Aarts WM, Schlom J, Hodge JW. Vector-based vaccine/cytokine combination therapy to enhance induction of immune responses to a self-antigen and antitumor activity. Cancer Res.62(20), 5770–5777 (2002).
  • Marshall JL, Gulley JL, Arlen PM et al. Phase I study of sequential vaccinations with Fowlpox-CEA (6D)–TRICOM alone and sequentially with vaccinia–CEA (6D)-TRICOM, with and without granulocyte-macrophage colony-stimulating factor, in patients with carcinoembryonic antigen-expressing carcinomas. J. Clin. Oncol.23(4), 720–731 (2005).
  • Vonderheide RH, Hahn WC, Schulze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity10(6), 673–679 (1999).
  • Arai J, Yasukawa M, Ohminami H et al. Identification of human telomerase reverse transcriptase-derived peptides that induce HLA-A24 restricted antileukemia cytotoxic T lymphocytes. Blood97(9), 2903–2907 (2001).
  • Nair SK, Heiser A, Boczkowski D et al. Induction of cytotoxic T cells response and tumor immunity against unrelated tumors using telomerase reverse transcriptase RNA transfected dendritic cells. Nat. Med.6(9), 1011–1017 (2000).
  • Vonderheide RH, Domchek SM, Schultze JL et al. Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin. Cancer Res.10(3), 828–839 (2002).
  • Maclean GD, Miles DW, Rubens RD et al. Enhancing the effect of THERATOPE STn–KLH cancer vaccine in patients with metastatic breast cancer by pretreatment with low-dose intravenous cyclophosphamide. J. Immunother. Emphasis Tumor Immunol.19(4), 309–316 (1996).
  • Sandmaier BM, Oparin DV, Holmberg LA et al. Evidence of a cellular response against sialyl-Tn in breast and ovarian cancer patients after high-dose chemotherapy, stem cell rescue, and immunization with Theratpe STn–KLH cancer vaccine. J. Immunother.22(1), 54–66 (1999).
  • Holmberg LA, Oparin DV, Gooley T et al. Clinical outcome of breast and ovarian cancer patients treated with high-dose chemotherapy, autologous stem cell rescue and THERATOPE STn–KLH cancer vaccine. Bone Marrow Transplant.25(12), 1233–1241 (2000).
  • Miles D, Cameron D, Dodwell D et al. Pretreatment chemotherapy (CX) regimens and responses in 1028 metastatic breast cancer (MBC) patients from an international, randomized Phase III clinical trial of STn–KLH therapeutic vaccine. Proceedings of the 39th American Society Clinical Oncology. IL, USA, May 31–June 3, 2003 (22, 55 Abstract 221).
  • Mayordomo J, Tres A, Miles D et al. Long-term follow-up of patients concomitantly treated with hormone therapy in a prospective controlled randomized multicenter clinical study comparing STn–KLH vaccine with KLH control in stage IV breast cancer following first-line chemotherapy. Proceedings of the 40th American Society Clinical Oncology. LA, USA, 5–8 June 2004, (22 Abstract 2603).
  • Ibrahim NK, Murray J, Parker J et al. Humoral immune-response to naturally occurring STn in metastatic breast cancer patients (MBC pts) treated with STn–KLH vaccine. Proceedings of the 40th American Society Clinical Oncology. LA, USA, 5–8 June 2004 (22 Abstract 2547).
  • Pharoah PD, Day NE, Caldas C. Somatic mutations in the p53 gene and prognosis in breast cancer: a meta-analysis. Br. J. Cancer80(12), 1968–1973 (1999).
  • Crawford LV, Pim DC, Bulbrook RD. Detection of antibodies against the cellular protein p53 in sera from patients with breast cancer. Int. J. Cancer30(4), 403–408 (1982).
  • Schlichtholz B, Legros Y, Gillet D et al. The immune response to p53 in breast cancer patients is directed against immunodominant epitopes unrelated to the mutational hot spot. Cancer Res.52(22), 6380–6384 (1992).
  • Peyrat JP, Bonneterre J, Lubin R et al. Prognostic significance of circulating p53 antibodies in patients undergoing surgery for locoregional breast cancer. Lancet345(8950), 621–622 (1995).
  • Mudenda B, Green JA, Green B et al. The relationship between serum p53 autoantibodies and characteristics of human breast cancer. Br. J. Cancer69(6), 1115–1119 (1994).
  • Trivers GE, Cawley HL, DeBenedetti VMG et al. Anti-p53 antibodies in sera of workers occupationally exposed to vinyl chloride. J. Natl Cancer Inst.87(18), 1400–1407 (1995).
  • Hammel P, Leroy-Viard K, Chaumette MT et al. Correlations between p53-protein accumulation, serum antibodies and gene mutation in colorectal cancer. Int. J. Cancer81(5), 712–718 (1999).
  • von Brevern MC, Hollstein MC, Cawley HM et al. Circulating anti-p53 antibodies in esophageal cancer patients are found predominantly in individuals with p53 core domain mutations in their tumors. Cancer Res.56(21), 4917–4921 (1996).
  • Lenner P, Wiklund F, Emdin SO et al. Serum antibodies against p53 in relation to cancer risk and prognosis in breast cancer: a population-based epidemiological study. Br. J. Cancer79(5–6), 927–932 (1999).
  • Willsher PC, Pinder SE, Robertson L et al. The significance of p53 autoantibodies in the serum of patients with breast cancer. Anticancer Res.16(2), 927–930 (1996).
  • Porzolt FMS, Hoher D, Muche D et al. Biological relevance of auto-antibodies against p53 in patients with metastatic breast cancer. Onkologie17, 402–408 (1994).
  • Tilkin AF, Lubin R, Soussi T et al. Primary proliferative T cell response to wild-type p53 protein in patients with breast cancer. Eur. J. Immunol.25(6), 1765–1769 (1995).
  • Van der Burg SH, Menon AG, Redeker A et al. Induction of p53-specific immune response in colorectal cancer patients receiving a recombinant ALVAC–p53 candidate vaccine. Clin. Cancer Res.8(5), 1019–1027 (2002).
  • Kuball J, Schuler M, Ferreira EA et al. Generating p53-specific cytotoxic T lymphocytes by recombinant adenoviral vector-based vaccination in mice, but not man. Gene Ther.9, 833–843 (2002).
  • Svane IM, Pedersen AE, Johnsen HE et al. Vaccination with p53-peptide-pulsed dendritic cells, of patients with advanced breast cancer: report from a Phase I study. Cancer Immunol. Immunother.53(7), 633–641 (2004).
  • Jocham D, Richter A, Hoffmann L et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: Phase III, randomised controlled trial. Lancet363(9409), 594–599 (2004).
  • Freedman RS, Edwards CL, Bowen JM et al. Viral oncolysates in patients with advanced ovarian cancer. Gynecol. Oncol.29(3), 337–347 (1998).
  • Freedman RS, Bowen JM, Atkinson EN et al. Randomized comparison of viral oncolysate plus radiation and radiation alone in uterine cervix carcinoma. Am. J. Clin. Oncol.12(3), 244–250 (1989).
  • Ahlert T, Sauerbrei W, Bastert G et al. Tumor cell number and viability as quality and efficacy parameters of autologous vurus-modified cancer vaccines in patients with breast or ovarian cancer. J. Clin. Oncol.15(4), 1354–1366 (1997).
  • Elder EM, Lotze EM, Whiteside TL. Successful culture and selection of cytokine gene modified human dermal fibroblasts for the biologic therapy of patients with cancer. Hum. Gene Ther.7(4), 479–487 (1996).
  • Dols A, Smith JW II, Meijer SL. Vaccination of women with metastatic breast cancer, using a costimulatory gene (CD80)-modified, HLA-A2-matched, allogeneic, breast cancer cell line: clinical and immunological results. Hum. Gene Ther.14(11), 1117–1123 (2003).
  • Dols A, Meijer SL, Hu HM et al. Identification of tumor-specific antibodies in patients with breast cancer vaccinated with gene-modified allogeneic tumor cells. J. Immunother.26(2), 163–170 (2003).
  • Wiseman CL. Inflammatory breast cancer: ten-year follow-up of a trial of surgery, chemotherapy, and allogeneic tumor cell/ BCG immunotherapy. Cancer Invest.13(3), 267–271 (1995).
  • Wiseman CL, Kharazi A. Objective clinical regression of metastatic breast cancer in disparate sites after use of whole-cell vaccine genetically modified to release sargramostim. Breast J.12(5), 475–480 (2006).
  • Jiang XP, Yang DC, Elliot RL, Jonathan F. Vaccination with a mixed vaccine of autogeneous and allogenic breast cancer cells and tumor associated antigens CA 15.3, CEA, and CA 125 results in immune and clinical responses in breast cancer patients. Cancer Biother. Radiopharm.15(5), 495–505 (2000).
  • Hernando JJ, Park TW, Kuhn WC. Dendritic cell-based vaccines in breast and gynaecologic cancer. Anticancer Res.23(5b), 4293–4303 (2003).
  • Restifo NP. Building better vaccines: how apoptotic cell death can induce inflammation and activate innate and adaptive immunity. Curr. Opin. Immunol.12(5), 597–603 (2000).
  • Sauter B, Albert ML, Francisco L et al. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J. Exp. Med.191(3), 423–433 (2000).
  • Yu B, Kusmartsev S, Cheng F et al. Effective combination of chemotherapy and dendritic cell administration for the treatment of advanced-stage experimental breast cancer. Clin. Cancer Res.9(1), 285–294 (2003).
  • Candido KA, Shimizu K, McLaughlin JC et al. Local administration of dendritic cells inhibits established breast tumor growth: implication for apoptosis-inducing agents. Cancer Res.61(1), 228–236 (2001).
  • Tong Y, Song W, Crystal RG. Combined intratumoral injection of bone marrow-derived dendritic cells and systemic chemotherapy to treat pre-existed murine tumors. Cancer Res.61(20), 7530–7535 (2001).
  • Casati A, Zimmermann VS, Benigni F et al. The immunogenicity of dendritic cell-based vaccines is not hampered by doxorubicin and melphalan administration. J. Immunology174(6), 3317–3325 (2005).
  • Neidhardt-Berard EM, Berard F, Banchereau J et al. Dendritic cells loaded with killed breast cancer cells induce differentiation of tumor-specific cytotoxic T lymphocytes. Breast Cancer Res.6(4), R322–R328 (2004).
  • Xia D, Moyana T, Xiang J. Combinational adenovirus-mediated gene therapy and dendritic cell vaccine in combating well-established tumors. Cell Res.16(3), 241–259 (2006).
  • Sakai Y, Morrison BJ, Burke JD et al. Vaccination by genetically modified dendritic cells expressing a truncated neu oncogene prevents development of breast cancer in transgenic mice. Cancer Res.64(21), 8022–8028 (2004).
  • Chen Y, Emtage P, Zhu Q et al. Induction of ErbB-2/neu-specific protective and therapeutic antitumor immunity using genetically modified dendritic cells: enhanced efficacy by cotransduction of gene encoding IL-12. Gene Ther.8(4), 316–323 (2001).
  • Chen Z, Huang H, Chang T et al. Enhanced HER2/neu-specific antitumor immunity by cotransduction of mouse dendritic cells with two genes encoding HER2/neu and α tumor necrosis factor. Cancer Gene Ther.9(9), 778–786 (2002).
  • Brossart P, Heinrich KS, Stuhler G et al. Identification of HLA-2-restricted T-cell epitopes derived from the MUC-1 tumor antigen for broadly applicable vaccine therapies. Blood93(12), 4309–4317 (1999).
  • Pecher G, Haring A, Kaiser L, Thiel E. Mucin gene (MUC1) transfected dendritic cells as vaccine: results of a Phase I/II clinical trial. Cancer Immunol. Immunother.51(11–12), 669–673 (2002).
  • Ojima T, Iwahashi M, Nakamura M, et al. Successful cancer vaccine therapy for carcinoembryonic antigen (CEA)-expressing colon cancer using genetically modified dendritic cells that express CEA and T helper-type 1 cytokines in CEA transgenic mice. Int. J. Cancer120(3), 585–593 (2007).
  • Brossart P, Stuhler G, Flad T et al. HER2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes. Cancer Res.58(4), 732–736 (1998).
  • Brossart P, Wirths S, Stuhler G et al. Induction of cytotoxic T-lymphocyte response in vivo after vaccinations with peptide-pulsed dendritic cells. Blood96(9), 3102–3108 (2000).
  • Peoples GE, Goedegeburre PS, Smith R et al. Breast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu derived peptide. Proc. Natl Acad. Sci. USA92, 423–426 (1995).
  • Fong L, Hou Y, Rivas A et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc. Natl Acad. Sci. USA98, 8809–8814 (2001).
  • Chui SY, Clay TM, Hobeika AC et al. Dendritic cell vaccination following high dose chemotherapy with autologous stem cell support for breast cancer: long term follow up. Proceedings of the 39th American Society of Clinical Oncology. IL, USA, 31 May–3 June, 2003 (Abstract 682).
  • Melisko ME, Kenzer M, Jones L et al. Vaccination with APC8024 (autologous dendritic cells loaded with antigen BA7072): immunologic and clinical activity in HER2/overexpressing breast cancer. Proceedings of the 39th American Society Clinical Oncology. IL, USA, 31 May–3 June, 2003 (22, 166 Abstract 665).
  • Kylstra JW, Park J, Jones L, Melisko M. Autologous antigen presenting cells loaded with BA7072 (APC8024) in patients with metastatic breast cancer overexpressing HER2/Neu: a Phase 1/2 study. Proceedings of the 39th American Society Clinical Oncology. IL, USA, 31 May–3 June, 2003 (22, 168 Abstract 673).
  • Svane IM, Pedersen AE, Johnsen HE et al. Vaccination with wild-type p53 peptide pulsed-dendritic cells in patients with advanced breast cancer. Results from a Phase I study. Proceedings of the 39th American Society Clinical Oncology. IL, USA, 31 May–3 June, 2003 (22, 17 Abstract 66).
  • Peethambaram P, Alberts S, Rinn K, Jones LA. Antigen presenting cell based immunotherapy targeting HER2/Neu positive solid tumors: results of a Phase 1 study of APC8024. Proceedings of the 40th American Society Clinical Oncology. New Orleans, LA, USA, 5–8 June, 2004 (Abstract 2528).
  • Kontani K, Taguchi O, Ozaki Y et al. Dendritic cell vaccine immunotherapy of cancer targeting MUC1 mucin. Int. J. Mol. Med.12(4), 493–502 (2003).
  • Czerniecki BJ, Koski GK, Koldovsky U et al. Targeting HER2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res.67(4), 1842–1852 (2007).
  • Svane IM, Pedersen AE, Johansen JS et al. Vaccination with p53 peptide-pulsed dendritic cells is associated with disease stabilization in patients with p53 expressing advanced breast cancer; monitoring of serum YKL-40 and IL-6 as response biomarkers. Cancer Immunol. Immunother.56(9), 1485–1499 (2007).
  • Gilewski T, Ragupathi G, Bhuta S et al. Immunization of metastatic breast cancer patients with a fully synthetic globo H conjugate: a Phase I trial. Proc. Natl Acad. Sci. USA98, 3270–3275 (2001).
  • Armstrong JL, Ragupathi, Powell S et al. Preliminary data of vaccination of high risk breast cancer (BC) patients with a heptavalent antigen: keyhole limpet hemocyanin (KLH) conjugate plus the immunologic adjuvant QS-21. Proceedings of the 39th American Society Clinical Oncology. IL, USA, 31 May–3 June 2003 (22, 168 Abstract 675).
  • Bitton RJ, Guthmann M, Koliren L et al. A Phase I/II safety and immunogenicity trial of hybridoma-derived anti-idiotypic monoclonal antibody vaccine (1E10), which mimics the neu-glycolyl-GM3 antigen, in patients with high risk breast cancer. Proceedings of the 40th American Society Clinical Oncology. New Orleans, LA, USA, 5–8 June, 2004 (22, 14S Abstract 737).
  • Cheever MA, Chen W. Therapy with cultured T cells: principles revisited. Immunol. Rev.157, 177–194 (1997).
  • Koga K, Matsumoto K, Akiyoshi T et al. Purification, characterization and biological significance of tumor-derived exosomes. Anticancer Res.25(6A), 3703–3707 (2005).
  • Yamano T, Kaneda Y, Hiramatsu SH et al. Immunity against breast cancer by TERT DNA vaccine primed with chemokine CCL21. Cancer Gene Ther.14(5), 451–459 (2007).
  • Monzavi-Karbassi B, Hennings LJ, Artaud C et al. Preclinical studies of carbohydrate mimetic peptide vaccines for breast cancer and melanoma. Vaccine25(16), 3022–3031 (2007).
  • Luo Y, Zhou H, Krueger J et al. Targeting tumor-associated macrophages as a novel strategy against breast cancer. J. Clin. Invest.116(8), 2132–2141 (2006).
  • Kim TS, Jung MY, Cho D, Cohen EP. Prolongation of the survival of breast cancer-bearing mice immunized with GM-CSF-secreting syngeneic/allogeneic fibroblasts transfected with a cDNA expression library from breast cancer cells. Vaccine24(42–43), 6564–6573 (2006).
  • Holmgren L, Ambrosino E, Birot O et al. A DNA vaccine targeting angiomotin inhibits angiogenesis and suppresses tumor growth. Proc. Natl Acad. Sci. USA103(24), 9208–9213 (2006).
  • Chan T, Sami A, El-Gayed A et al. HER-2/neu-gene engineered dendritic cell vaccine stimulates stronger HER2/neu-specific immune responses compared to DNA vaccination. Gene Ther.13(19), 1391–402 (2006).
  • Koido S, Tanaka Y, Tajiri H, Gong J. Generation and functional assessment of antigen-specific T cells stimulated by fusions of dendritic cells and allogeneic breast cancer cells. Vaccine25(14), 2610–2619. (2007).
  • Zhang Y, Ma B, Zhou Y et al. Dendritic cells fused with allogeneic breast cancer cell line induce tumor antigen-specific CTL responses against autologous breast cancer cells. Breast Cancer Res Treat. (2006) (Epub ahead of print).
  • Shi L, Sings HL, Bryan JT et al. GARDASIL: prophylactic human papillomavirus vaccine development – from bench top to bed-side. Clin. Pharmacol. Ther.81(2), 259–264 (2007).
  • Chang MH, Chen TH, Hsu HM et al.; for the Taiwan Childhood HCC Study Group. Prevention of hepatocellular carcinoma by universal vaccination against hepatitis B virus: the effect and problems. Clin. Cancer Res.11(21), 7953–7957 (2005).
  • Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat. Med.10(9), 909–915 (2004).
  • Hoos A, Parmiani G, Hege K et al. Cancer vaccine clinical trial working group. A clinical development paradigm for cancer vaccines and related biologics. J. Immunother. (1997)30(1), 1–15 (2007).
  • Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer26, 239–257 (1972).
  • Nowak AK, Lake RA, Marzo AL et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells, J. Immunol.170, 4905–4913 (2003).
  • Rovere P, Sabbadini MG, Vallinoto C et al.Delayed clearance of apoptotic lymphoma cells allows cross-presentation of intracellular antigens by mature dendritic cells. J. Leukoc. Biol.66, 345–349 (1999).
  • Turk JL, Parker D. Effect of cyclophosphamide on immunological control mechanisms. Immunol. Rev.65, 99–113 (1982).
  • Bass KK, Mastrangelo MJ. Immunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer. Cancer Immunol. Immunother.47, 1–12 (1998).
  • Berd D, Mastrangelo MJ, Engstrom PF, Paul A, Maguire H. Augmentation of the human immune response by cyclophosphamide. Cancer Res.42, 4862–4866 (1982).
  • Correale P, Cusi MG, Tsang KY et al. Chemo–immunotherapy of metastatic colorectal carcinoma with gemcitabine plus FOLFOX 4 followed by subcutaneous granulocyte macrophage colony-stimulating factor and interleukin-2 induces strong immunologic and antitumor activity in metastatic colon cancer patients, J. Clin. Oncol.23, 8950–8958 (2005).
  • Foon KA, John WJ, Chakraborty M, Das R. Clinical and immune responses in resected colon cancer patients treated with anti-idiotype monoclonal antibody vaccine that mimics the carcinoembryonic antigen, J. Clin. Oncol.17, 2889–2895 (1999).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.