Skip to main content

The Use of Dendritic Cells for Peptide-Based Vaccination in Cancer Immunotherapy

  • Protocol
  • First Online:
Cancer Vaccines

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1139))

Abstract

Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.

Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Matzinger P (2002) The danger model: a renewed sense of self. Science 296(5566):301–305

    CAS  PubMed  Google Scholar 

  2. Finn OJ (2008) Cancer immunology. N Engl J Med 358(25):2704–2715

    CAS  PubMed  Google Scholar 

  3. McCarthy EF (2006) The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop J 26:154–158

    PubMed Central  PubMed  Google Scholar 

  4. Finn OJ (2008) Immunological weapons acquired early in life win battles with cancer late in life. J Immunol 181(3):1589–1592

    CAS  PubMed  Google Scholar 

  5. Pejawar-Gaddy S, Finn OJ (2008) Cancer vaccines: accomplishments and challenges. Crit Rev Oncol Hematol 67(2):93–102

    PubMed  Google Scholar 

  6. Salem ML et al (2007) Tumours: immunotherapy. Encyclopedia of life sciences, 2007, John Wiley & Sons, Ltd. www.els.net

  7. Yannelli JR, Wroblewski JM (2004) On the road to a tumor cell vaccine: 20 years of cellular immunotherapy. Vaccine 23(1):97–113

    CAS  PubMed  Google Scholar 

  8. Vermorken JB et al (1999) Active specific immunotherapy for stage II and stage III human colon cancer: a randomised trial. Lancet 353(9150):345–350

    CAS  PubMed  Google Scholar 

  9. Melief CJ (2008) Cancer immunotherapy by dendritic cells. Immunity 29(3):372–383

    CAS  PubMed  Google Scholar 

  10. Banchereau J et al (2003) Dendritic cells: controllers of the immune system and a new promise for immunotherapy. Novartis Found Symp 252:226–235, discussion 235–228, 257–267

    CAS  PubMed  Google Scholar 

  11. Stift A et al (2003) Dendritic cell-based vaccination in solid cancer. J Clin Oncol 21(1):135–142

    CAS  PubMed  Google Scholar 

  12. Brossart P (2002) Dendritic cells in vaccination therapies of malignant diseases. Transfus Apher Sci 27(2):183–186

    PubMed  Google Scholar 

  13. Nagorsen D, Thiel E (2006) Clinical and immunologic responses to active specific cancer vaccines in human colorectal cancer. Clin Cancer Res 12(10):3064–3069

    CAS  PubMed  Google Scholar 

  14. Steinman RM, Cohn ZA (1973) Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med 137(5):1142–1162

    CAS  PubMed Central  PubMed  Google Scholar 

  15. Rosenzwajg M et al (1996) Human dendritic cell differentiation pathway from CD34+ hematopoietic precursor cells. Blood 87(2):535–544

    CAS  PubMed  Google Scholar 

  16. Strobl H et al (1996) TGF-beta 1 promotes in vitro development of dendritic cells from CD34+ hemopoietic progenitors. J Immunol 157(4):1499–1507

    CAS  PubMed  Google Scholar 

  17. Yao V et al (2002) Dendritic cells. ANZ J Surg 72(7):501–506

    PubMed  Google Scholar 

  18. Reid CD (1997) The dendritic cell lineage in haemopoiesis. Br J Haematol 96(2):217–223

    CAS  PubMed  Google Scholar 

  19. Szabolcs P et al (1996) Dendritic cells and macrophages can mature independently from a human bone marrow-derived, post-colony-forming unit intermediate. Blood 87(11):4520–4530

    CAS  PubMed  Google Scholar 

  20. Rossi G et al (1992) Development of a Langerhans cell phenotype from peripheral blood monocytes. Immunol Lett 31(2):189–197

    CAS  PubMed  Google Scholar 

  21. Kasinrerk W et al (1993) CD1 molecule expression on human monocytes induced by granulocyte-macrophage colony-stimulating factor. J Immunol 150(2):579–584

    CAS  PubMed  Google Scholar 

  22. Zhou LJ, Tedder TF (1996) CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells. Proc Natl Acad Sci USA 93(6):2588–2592

    CAS  PubMed Central  PubMed  Google Scholar 

  23. den Brok MH et al (2005) Dendritic cells: tools and targets for antitumor vaccination. Expert Rev Vaccines 4(5):699–710

    Google Scholar 

  24. Morisaki T et al (2003) Dendritic cell-based combined immunotherapy with autologous tumor-pulsed dendritic cell vaccine and activated T cells for cancer patients: rationale, current progress, and perspectives. Hum Cell 16(4):175–182

    PubMed  Google Scholar 

  25. Tacken PJ et al (2007) Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol 7(10):790–802

    CAS  PubMed  Google Scholar 

  26. Caux C et al (1992) GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells. Nature 360(6401):258–261

    CAS  PubMed  Google Scholar 

  27. Cella M et al (1996) Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med 184(2):747–752

    CAS  PubMed  Google Scholar 

  28. Whiteside TL, Odoux C (2004) Dendritic cell biology and cancer therapy. Cancer Immunol Immunother 53(3):240–248

    PubMed  Google Scholar 

  29. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature 392(6673):245–252

    CAS  PubMed  Google Scholar 

  30. Aarntzen EH et al (2008) Dendritic cell vaccination and immune monitoring. Cancer Immunol Immunother 57(10):1559–1568

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Nicola M et al (1999) The influence of interleukin (IL)-4 and IL-13 on human dendritic cell differentiation from CD34+ progenitor cells: the importance of the source of serum. Exp Hematol 27(2):386–387

    PubMed  Google Scholar 

  32. Esche C et al (1999) The use of dendritic cells for cancer vaccination. Curr Opin Mol Ther 1(1):72–81

    CAS  PubMed  Google Scholar 

  33. Zhang SN et al (2011) Optimizing DC vaccination by combination with oncolytic adenovirus coexpressing IL-12 and GM-CSF. [Research Support, Non-U.S. Gov’t]. Mol Ther 19(8):1558–1568

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Brinkman JA et al (2004) Peptide-based vaccines for cancer immunotherapy. Expert Opin Biol Ther 4(2):181–198

    CAS  PubMed  Google Scholar 

  35. Kavanagh B et al (2007) Vaccination of metastatic colorectal cancer patients with matured dendritic cells loaded with multiple major histocompatibility complex class I peptides. J Immunother 30(7):762–772

    CAS  PubMed  Google Scholar 

  36. Harley CB et al (1990) Telomeres shorten during ageing of human fibroblasts. Nature 345(6274):458–460

    CAS  PubMed  Google Scholar 

  37. Cong YS et al (2002) Human telomerase and its regulation. Microbiol Mol Biol Rev 66(3):407–425

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Vonderheide RH (2008) Prospects and challenges of building a cancer vaccine targeting telomerase. Biochimie 90(1):173–180

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Vonderheide RH et al (2004) Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 10(3):828–839

    CAS  PubMed  Google Scholar 

  40. Su Z et al (2005) Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer. J Immunol 174(6):3798–3807

    CAS  PubMed  Google Scholar 

  41. Kono K et al (2002) Dendritic cells pulsed with HER-2/neu-derived peptides can induce specific T-cell responses in patients with gastric cancer. Clin Cancer Res 8(11):3394–3400

    CAS  PubMed  Google Scholar 

  42. Aloysius M et al (2009) Generation in vivo of peptide-specific cytotoxic T cells and presence of regulatory T cells during vaccination with hTERT (class I and II) peptide-pulsed DCs. [Research Support, Non-U.S. Gov’t]. J Transl Med 7:18

    PubMed Central  PubMed  Google Scholar 

  43. Suso EM et al (2011) hTERT mRNA dendritic cell vaccination: complete response in a pancreatic cancer patient associated with response against several hTERT epitopes. Cancer Immunol Immunother 60(6):809–818

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Terada T et al (1996) Expression of MUC apomucins in normal pancreas and pancreatic tumours. J Pathol 180(2):160–165

    CAS  PubMed  Google Scholar 

  45. Taylor-Papadimitriou J et al (1999) MUC1 and cancer. Biochim Biophys Acta 1455(2–3):301–313

    CAS  PubMed  Google Scholar 

  46. Brossart P et al (2001) The epithelial tumor antigen MUC1 is expressed in hematological malignancies and is recognized by MUC1-specific cytotoxic T-lymphocytes. Cancer Res 61(18):6846–6850

    CAS  PubMed  Google Scholar 

  47. Jerome KR et al (1993) Tumor-specific cytotoxic T cell clones from patients with breast and pancreatic adenocarcinoma recognize EBV-immortalized B cells transfected with polymorphic epithelial mucin complementary DNA. J Immunol 151(3):1654–1662

    CAS  PubMed  Google Scholar 

  48. Yamamoto K et al (2005) MUC1 peptide vaccination in patients with advanced pancreas or biliary tract cancer. Anticancer Res 25(5):3575–3579

    CAS  PubMed  Google Scholar 

  49. Brossart P et al (2000) Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 96(9):3102–3108

    CAS  PubMed  Google Scholar 

  50. Brossart P, Bevan MJ (1997) Presentation of exogenous protein antigens on major histocompatibility complex class I molecules by dendritic cells: pathway of presentation and regulation by cytokines. Blood 90(4):1594–1599

    CAS  PubMed Central  PubMed  Google Scholar 

  51. 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):5910–5918

    CAS  PubMed  Google Scholar 

  52. Kim Y et al (2003) Gastrointestinal tract cancer screening using fecal carcinoembryonic antigen. Ann Clin Lab Sci 33(1):32–38

    PubMed  Google Scholar 

  53. Kaufman HL et al (2007) Poxvirus-based vaccine therapy for patients with advanced pancreatic cancer. J Transl Med 5:60

    PubMed Central  PubMed  Google Scholar 

  54. Tassi E et al (2008) Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. J Immunol 181(9):6595–6603

    CAS  PubMed  Google Scholar 

  55. Ueda Y et al (2004) Dendritic cell-based immunotherapy of cancer with carcinoembryonic antigen-derived, HLA-A24-restricted CTL epitope: clinical outcomes of 18 patients with metastatic gastrointestinal or lung adenocarcinomas. [Clinical trial]. Int J Oncol 24(4):909–917

    CAS  PubMed  Google Scholar 

  56. Liu KJ et al (2004) Generation of carcinoembryonic antigen (CEA)-specific T-cell responses in HLA-A*0201 and HLA-A*2402 late-stage colorectal cancer patients after vaccination with dendritic cells loaded with CEA peptides. Clin Cancer Res 10(8):2645–2651

    CAS  PubMed  Google Scholar 

  57. 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):268–276

    CAS  PubMed  Google Scholar 

  58. Lesterhuis WJ et al (2006) Vaccination of colorectal cancer patients with CEA-loaded dendritic cells: antigen-specific T cell responses in DTH skin tests. Ann Oncol 17(6):974–980

    CAS  PubMed  Google Scholar 

  59. Lesterhuis WJ et al (2008) Dendritic cell vaccines in melanoma: from promise to proof? Crit Rev Oncol Hematol 66(2):118–134

    CAS  PubMed  Google Scholar 

  60. Lesterhuis WJ et al (2010) Immunogenicity of dendritic cells pulsed with CEA peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res 30(12):5091–5097

    PubMed  Google Scholar 

  61. Ambrosini G et al (1997) A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3(8):917–921

    CAS  PubMed  Google Scholar 

  62. Lopes RB et al (2007) Expression of the IAP protein family is dysregulated in pancreatic cancer cells and is important for resistance to chemotherapy. Int J Cancer 120(11):2344–2352

    CAS  PubMed  Google Scholar 

  63. Casati C et al (2003) The apoptosis inhibitor protein survivin induces tumor-specific CD8+ and CD4+ T cells in colorectal cancer patients. Cancer Res 63(15):4507–4515

    CAS  PubMed  Google Scholar 

  64. Otto K et al (2005) Lack of toxicity of therapy-induced T cell responses against the universal tumour antigen survivin. Vaccine 23(7):884–889

    CAS  PubMed  Google Scholar 

  65. Tsuruma T et al (2004) Phase I clinical study of anti-apoptosis protein, survivin-derived peptide vaccine therapy for patients with advanced or recurrent colorectal cancer. J Transl Med 2(1):19

    PubMed Central  PubMed  Google Scholar 

  66. Tsuruma T et al (2008) Clinical and immunological evaluation of anti-apoptosis protein, survivin-derived peptide vaccine in phase I clinical study for patients with advanced or recurrent breast cancer. J Transl Med 6:24

    PubMed Central  PubMed  Google Scholar 

  67. Karanikas V et al (2008) Baseline levels of CD8+ T cells against survivin and survivin-2B in the blood of lung cancer patients and cancer-free individuals. Clin Immunol 129(2):230–240

    CAS  PubMed  Google Scholar 

  68. Wobser M et al (2006) Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunol Immunother 55(10):1294–1298

    CAS  PubMed  Google Scholar 

  69. Nagaraj S et al (2007) Dendritic cell-based full-length survivin vaccine in treatment of experimental tumors. J Immunother 30(2):169–179

    CAS  PubMed  Google Scholar 

  70. Eggert AA et al (1999) Biodistribution and vaccine efficiency of murine dendritic cells are dependent on the route of administration. Cancer Res 59(14):3340–3345

    CAS  PubMed  Google Scholar 

  71. Moyer JS et al (2008) Intratumoral dendritic cells and chemoradiation for the treatment of murine squamous cell carcinoma. J Immunother 31(9):885–895

    PubMed  Google Scholar 

  72. Rossowska J et al (2007) Tissue localization of tumor antigen-loaded mouse dendritic cells applied as an anti-tumor vaccine and their influence on immune response. Folia Histochem Cytobiol 45(4):349–355

    CAS  PubMed  Google Scholar 

  73. Cohen S et al (2009) Dendritic cell-based therapeutic vaccination against myeloma: vaccine formulation determines efficacy against light chain myeloma. J Immunol 182(3):1667–1673

    CAS  PubMed  Google Scholar 

  74. De Vries IJ et al (2003) Effective migration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients is determined by their maturation state. Cancer Res 63(1):12–17

    PubMed  Google Scholar 

  75. Trakatelli M et al (2006) A new dendritic cell vaccine generated with interleukin-3 and interferon-beta induces CD8+ T cell responses against NA17-A2 tumor peptide in melanoma patients. Cancer Immunol Immunother 55(4):469–474

    CAS  PubMed  Google Scholar 

  76. Zitvogel L, Tursz T (2005) In vivo veritas. Nat Biotechnol 23(11):1372–1374

    CAS  PubMed  Google Scholar 

  77. Bedrosian I et al (2003) Intranodal administration of peptide-pulsed mature dendritic cell vaccines results in superior CD8+ T-cell function in melanoma patients. J Clin Oncol 21(20):3826–3835

    CAS  PubMed  Google Scholar 

  78. Kyte JA et al (2006) Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor-mRNA. Cancer Gene Ther 13(10):905–918

    CAS  PubMed  Google Scholar 

  79. Fong L et al (2001) Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol 166(6):4254–4259

    CAS  PubMed  Google Scholar 

  80. Mullins DW et al (2003) Route of immunization with peptide-pulsed dendritic cells controls the distribution of memory and effector T cells in lymphoid tissues and determines the pattern of regional tumor control. J Exp Med 198(7):1023–1034

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Adema GJ et al (2005) Migration of dendritic cell based cancer vaccines: in vivo veritas? Curr Opin Immunol 17(2):170–174

    CAS  PubMed  Google Scholar 

  82. Liau LM et al (2005) Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res 11(15):5515–5525

    CAS  PubMed  Google Scholar 

  83. Palmer DH et al (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

    PubMed  Google Scholar 

  84. Trepiakas R et al (2010) Vaccination with autologous dendritic cells pulsed with multiple tumor antigens for treatment of patients with malignant melanoma: results from a phase I/II trial. Cytotherapy 12(6):721–734

    CAS  PubMed  Google Scholar 

  85. Waisman A, Yogev N (2009) B7-H1 and CD8+ Treg: the enigmatic role of B7-H1 in peripheral tolerance. Eur J Immunol 39(6):1448–1451

    CAS  PubMed  Google Scholar 

  86. Yoshimura A (2009) Regulation of cytokine signaling by the SOCS and Spred family proteins. Keio J Med 58(2):73–83

    CAS  PubMed  Google Scholar 

  87. Lob S, Konigsrainer A (2008) Is IDO a key enzyme bridging the gap between tumor escape and tolerance induction? Langenbecks Arch Surg 393(6):995–1003

    PubMed  Google Scholar 

  88. Mahnke K et al (2007) Tolerogenic dendritic cells and regulatory T cells: a two-way relationship. J Dermatol Sci 46(3):159–167

    CAS  PubMed  Google Scholar 

  89. Marigo I et al (2008) Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev 222:162–179

    CAS  PubMed  Google Scholar 

  90. Li H et al (2009) Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol 182(1):240–249

    CAS  PubMed  Google Scholar 

  91. Bronte V, Mocellin S (2009) Suppressive influences in the immune response to cancer. J Immunother 32(1):1–11

    PubMed  Google Scholar 

  92. Llopiz D et al (2009) Peptide inhibitors of transforming growth factor-beta enhance the efficacy of antitumor immunotherapy. Int J Cancer 125(11):2614–2623

    CAS  PubMed  Google Scholar 

  93. Pellegrini M et al (2009) Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies. Nat Med 15(5):528–536

    CAS  PubMed  Google Scholar 

  94. Vogt TK et al (2009) Novel function for interleukin-7 in dendritic cell development. Blood 113(17):3961–3968

    CAS  PubMed  Google Scholar 

  95. Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9(3):162–174

    CAS  PubMed Central  PubMed  Google Scholar 

  96. Lechner MG et al (2011) Functional characterization of human Cd33+ and Cd11b+ myeloid-derived suppressor cell subsets induced from peripheral blood mononuclear cells co-cultured with a diverse set of human tumor cell lines. J Transl Med 9:90

    CAS  PubMed Central  PubMed  Google Scholar 

  97. Chatila TA (2009) Regulatory T cells: key players in tolerance and autoimmunity. Endocrinol Metab Clin North Am 38(2):265–272

    CAS  PubMed Central  PubMed  Google Scholar 

  98. Mills KH (2004) Regulatory T cells: friend or foe in immunity to infection? Nat Rev Immunol 4(11):841–855

    CAS  PubMed  Google Scholar 

  99. Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6(4):345–352

    CAS  PubMed  Google Scholar 

  100. Zhou G et al (2006) Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines. Blood 107(2):628–636

    CAS  PubMed Central  PubMed  Google Scholar 

  101. Xu L et al (2011) In situ prior proliferation of CD4+ CCR6+ regulatory T cells facilitated by TGF-beta secreting DC is crucial for their enrichment and suppression in tumor immunity. PLoS One 6(5):1–10

    Google Scholar 

  102. Diaz-Montero CM et al (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

    CAS  PubMed Central  PubMed  Google Scholar 

  103. Stewart TJ, Smyth MJ (2011) Improving cancer immunotherapy by targeting tumor-induced immune suppression. Cancer Metastasis Rev 30(1):125–140

    CAS  PubMed  Google Scholar 

  104. Bak SP et al (2008) Murine ovarian cancer vascular leukocytes require arginase-1 activity for T cell suppression. Mol Immunol 46(2):258–268

    CAS  PubMed Central  PubMed  Google Scholar 

  105. Corzo CA et al (2009) Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 182(9):5693–5701

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Ray P et al (2011) Lung myeloid-derived suppressor cells and regulation of inflammation. Immunol Res 50(2–3):153–158

    CAS  PubMed  Google Scholar 

  107. Wang Z et al (2008) Short-term anti-CD25 monoclonal antibody treatment and neogenetic CD4(+)CD25(high) regulatory T cells in kidney transplantation. Transpl Immunol 19(1):69–73

    PubMed  Google Scholar 

  108. Yang L et al (2004) Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 6(4):409–421

    CAS  PubMed  Google Scholar 

  109. Finke J et al (2011) MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy. Int Immunopharmacol 11(7):856–861

    CAS  PubMed  Google Scholar 

  110. Ko JS et al (2009) Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res 15(6):2148–2157

    CAS  PubMed  Google Scholar 

  111. Kusmartsev S et al (2008) Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma. Clin Cancer Res 14(24):8270–8278

    CAS  PubMed  Google Scholar 

  112. Qian F et al (2009) Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2,3-dioxygenase-mediated arrest of T-cell proliferation in human epithelial ovarian cancer. Cancer Res 69(13):5498–5504

    CAS  PubMed  Google Scholar 

  113. Wang RF et al (2008) Toll-like receptors and immune regulation: implications for cancer therapy. Oncogene 27(2):181–189

    PubMed  Google Scholar 

  114. Platz KP et al (2005) IL-2 antagonists: the European perspective. Transplant Proc 37(4):1783–1784

    CAS  PubMed  Google Scholar 

  115. Duvic M, Talpur R (2008) Optimizing denileukin diftitox (Ontak) therapy. Future Oncol 4(4):457–469

    CAS  PubMed  Google Scholar 

  116. Ozao-Choy J et al (2009) The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res 69(6):2514–2522

    CAS  PubMed  Google Scholar 

  117. Chen YL et al (2008) Depletion of CD4(+)CD25(+) regulatory T cells can promote local immunity to suppress tumor growth in benzo[a]pyrene-induced forestomach carcinoma. World J Gastroenterol 14(38):5797–5809

    PubMed Central  PubMed  Google Scholar 

  118. Vonderheide RH, June CH (2003) A translational bridge to cancer immunotherapy: exploiting costimulation and target antigens for active and passive T cell immunotherapy. Immunol Res 27(2–3):341–356

    CAS  PubMed  Google Scholar 

  119. Kowalczyk DW (2002) Tumors and the danger model. Acta Biochim Pol 49(2):295–302

    CAS  PubMed  Google Scholar 

  120. Kawai T, Akira S (2007) TLR signaling. Semin Immunol 19(1):24–32

    CAS  PubMed  Google Scholar 

  121. Seya T et al (2006) Role of Toll-like receptors in adjuvant-augmented immune therapies. Evid Based Complement Alternat Med 3(1):31–38, discussion 133–137

    PubMed Central  PubMed  Google Scholar 

  122. Salem ML et al (2009) Recovery from cyclophosphamide-induced lymphopenia results in expansion of immature dendritic cells which can mediate enhanced prime-boost vaccination antitumor responses in vivo when stimulated with the TLR3 agonist poly(I:C). J Immunol 182(4):2030–2040

    CAS  PubMed Central  PubMed  Google Scholar 

  123. Salem ML et al (2009) The TLR3 agonist poly(I:C) targets CD8+ T cells and augments their antigen-specific responses upon their adoptive transfer into naive recipient mice. Vaccine 27(4):549–557

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Salem ML et al (2006) The adjuvant effects of the toll-like receptor 3 ligand polyinosinic-cytidylic acid poly(I:C) on antigen-specific CD8+ T cell responses are partially dependent on NK cells with the induction of a beneficial cytokine milieu. Vaccine 24(24):5119–5132

    CAS  PubMed  Google Scholar 

  125. Salem ML et al (2005) Defining the antigen-specific T-cell response to vaccination and poly(I:C)/TLR3 signaling: evidence of enhanced primary and memory CD8 T-cell responses and antitumor immunity. J Immunother 28(3):220–228

    CAS  PubMed  Google Scholar 

  126. Salem ML et al (2007) Defining the ability of cyclophosphamide preconditioning to enhance the antigen-specific CD8+ T-cell response to peptide vaccination: creation of a beneficial host microenvironment involving type I IFNs and myeloid cells. J Immunother 30(1):40–53

    CAS  PubMed  Google Scholar 

  127. Banchereau J, Palucka AK (2005) Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol 5(4):296–306

    CAS  PubMed  Google Scholar 

  128. Fajardo-Moser M et al (2008) Mechanisms of dendritic cell-based vaccination against infection. Int J Med Microbiol 298(1–2):11–20

    PubMed  Google Scholar 

  129. Datta SK et al (2003) A subset of Toll-like receptor ligands induces cross-presentation by bone marrow-derived dendritic cells. J Immunol 170(8):4102–4110

    CAS  PubMed  Google Scholar 

  130. Edwards AD et al (2003) Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8 alpha+ DC correlates with unresponsiveness to imidazoquinolines. Eur J Immunol 33(4):827–833

    CAS  PubMed  Google Scholar 

  131. Lore K et al (2003) Toll-like receptor ligands modulate dendritic cells to augment cytomegalovirus- and HIV-1-specific T cell responses. J Immunol 171(8):4320–4328

    CAS  PubMed  Google Scholar 

  132. Kadowaki N et al (2001) Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med 194(6):863–869

    CAS  PubMed Central  PubMed  Google Scholar 

  133. West MA et al (2004) Enhanced dendritic cell antigen capture via toll-like receptor-induced actin remodeling. Science 305(5687):1153–1157

    CAS  PubMed  Google Scholar 

  134. Meyer T, Stockfleth E (2008) Clinical investigations of Toll-like receptor agonists. Expert Opin Investig Drugs 17(7):1051–1065

    CAS  PubMed  Google Scholar 

  135. Boullart AC et al (2008) Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother 57(11):1589–1597

    CAS  PubMed Central  PubMed  Google Scholar 

  136. Jasani B et al (2009) Ampligen: a potential toll-like 3 receptor adjuvant for immunotherapy of cancer. Vaccine 27(25–26):3401–3404

    CAS  PubMed  Google Scholar 

  137. Renn CN et al (2006) TLR activation of Langerhans cell-like dendritic cells triggers an antiviral immune response. J Immunol 177(1):298–305

    CAS  PubMed  Google Scholar 

  138. Tirapu I et al (2009) PolyI:C-induced reduction in uptake of soluble antigen is independent of dendritic cell activation. Int Immunol 21(7):871–879

    CAS  PubMed Central  PubMed  Google Scholar 

  139. Walker J, Tough DF (2006) Modification of TLR-induced activation of human dendritic cells by type I IFN: synergistic interaction with TLR4 but not TLR3 agonists. Eur J Immunol 36(7):1827–1836

    CAS  PubMed  Google Scholar 

  140. Adams M et al (2003) Dendritic cell (DC) based therapy for cervical cancer: use of DC pulsed with tumour lysate and matured with a novel synthetic clinically non-toxic double stranded RNA analogue poly [I]:poly [C(12)U] (Ampligen R). Vaccine 21(7–8):787–790

    CAS  PubMed  Google Scholar 

  141. Mailliard RB et al (2004) Alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res 64(17):5934–5937

    CAS  PubMed  Google Scholar 

  142. Navabi H et al (2009) A clinical grade poly I:C-analogue (Ampligen) promotes optimal DC maturation and Th1-type T cell responses of healthy donors and cancer patients in vitro. Vaccine 27(1):107–115

    CAS  PubMed  Google Scholar 

  143. Zhu X et al (2007) Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J Transl Med 5:10

    PubMed Central  PubMed  Google Scholar 

  144. Austyn JM (1998) Dendritic cells. Curr Opin Hematol 5(1):3–15

    CAS  PubMed  Google Scholar 

  145. Satthaporn S, Eremin O (2001) Dendritic cells (I): biological functions. J R Coll Surg Edinb 46(1):9–19

    CAS  PubMed  Google Scholar 

  146. Satthaporn S, Eremin O (2001) Dendritic cells (II): role and therapeutic implications in cancer. J R Coll Surg Edinb 46(3):159–167

    CAS  PubMed  Google Scholar 

  147. Shortman K, Caux C (1997) Dendritic cell development: multiple pathways to nature’s adjuvants. Stem Cells 15(6):409–419

    CAS  PubMed  Google Scholar 

  148. Shortman K et al (1997) Dendritic cells and T lymphocytes: developmental and functional interactions. Ciba Found Symp 204:130–138, discussion 138–141

    CAS  PubMed  Google Scholar 

  149. Schmidt J et al (2007) Intratumoural injection of the toll-like receptor-2/6 agonist ‘macrophage-activating lipopeptide-2’ in patients with pancreatic carcinoma: a phase I/II trial. Br J Cancer 97(5):598–604

    CAS  PubMed Central  PubMed  Google Scholar 

  150. Schneider C et al (2004) Tumour suppression induced by the macrophage activating lipopeptide MALP-2 in an ultrasound guided pancreatic carcinoma mouse model. Gut 53(3):355–361

    CAS  PubMed Central  PubMed  Google Scholar 

  151. Schill T et al (2012) Stimulation of pulmonary immune responses by the TLR2/6 agonist MALP-2 and effect on melanoma metastasis to the lung. Exp Dermatol 21(2):91–98

    CAS  PubMed  Google Scholar 

  152. Pratesi G et al (2005) Therapeutic synergism of gemcitabine and CpG-oligodeoxynucleotides in an orthotopic human pancreatic carcinoma xenograft. Cancer Res 65(14):6388–6393

    CAS  PubMed  Google Scholar 

  153. Vanderlocht J et al (2010) Increased tumor-specific CD8+ T cell induction by dendritic cells matured with a clinical grade TLR-agonist in combination with IFN-gamma. Int J Immunopathol Pharmacol 23(1):35–50

    CAS  PubMed  Google Scholar 

  154. Okamoto M, Sato M (2003) Toll-like receptor signaling in anti-cancer immunity. J Med Invest 50(1–2):9–24

    PubMed  Google Scholar 

  155. Osada T et al (2006) Dendritic cell-based immunotherapy. Int Rev Immunol 25(5–6):377–413

    CAS  PubMed  Google Scholar 

  156. Cheever MA (2008) Twelve immunotherapy drugs that could cure cancers. Immunol Rev 222:357–368

    CAS  PubMed  Google Scholar 

  157. Cheever MA et al (2008) Translational Research Working Group developmental pathway for immune response modifiers. Clin Cancer Res 14(18):5692–5699

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this protocol

Cite this protocol

Salem, M.L. (2014). The Use of Dendritic Cells for Peptide-Based Vaccination in Cancer Immunotherapy. In: Lawman, M., Lawman, P. (eds) Cancer Vaccines. Methods in Molecular Biology, vol 1139. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-0345-0_37

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-0345-0_37

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-0344-3

  • Online ISBN: 978-1-4939-0345-0

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics