Chonnam Med J.  2010 Apr;46(1):1-6. 10.4068/cmj.2010.46.1.1.

The Clinical Impact of the Dendritic Cell-based Cancer Vaccine: the Role in the Inflammatory Tumor Micro-environment

Affiliations
  • 1Clinical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. andyjosh@skku.edu

Abstract

As a novel therapeutic module, the dendritic cell (DC) based cancer vaccine has been recognized with great hope in eliminating cancers, including minimal residual cells, without harming normal tissue. A key factor in initiating and operating the immune system against foreign bodies including tumor cells, the DC has been regarded as the next possible breakthrough in new cancer therapy. However, the results of more than 15 years of clinical studies with DC vaccine revealed the difficulties fulfilling this expectation. Evidence has disclosed that the DC activation required for proper tumor-specific effector CD4+ and CD8+ T cell stimulation is inhibited in the micro-environment of cancer. Studies have further reported that DC phenotypes in cancer tissue and draining lymph nodes are mostly immature, which results in regulatory immune responses. Also, the existence of myeloid derived suppressor cells (MDSCs) and tumor associated macrophages (TAMs) adversely affect both DC function and immune suppression in the cancer-environment. In this review, the impact of an inflammatory micro-environment induced by cancer on the effect of DC-based cancer immunotherapy and the possibility of a clinical efficacy improvement are discussed.

Keyword

Cancer immunotherapy; DC-based cancer vaccine; tumor micro-environment

MeSH Terms

Dendritic Cells
Foreign Bodies
Immune System
Immunotherapy
Lymph Nodes
Macrophages
Phenotype

Reference

1. Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996. 2:52–58.
Article
2. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998. 4:328–332.
Article
3. Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005. 5:296–306.
Article
4. Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell immunotherapy: mapping the way. Nat Med. 2004. 10:475–480.
Article
5. Gilboa E. The promise of cancer vaccines. Nat Rev Cancer. 2004. 4:401–411.
Article
6. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007. 449:419–426.
Article
7. Nestle FO, Farkas A, Conrad C. Dendritic-cell-based therapeutic vaccination against cancer. Curr Opin Immunol. 2005. 17:163–169.
Article
8. Pardoll DM. Cancer vaccines. Nat Med. 1998. 4:5 Suppl. 525–531.
Article
9. Parmiani G, Pilla L, Castelli C, Rivoltini L. Vaccination of patients with solid tumours. Ann Oncol. 2003. 14:817–824.
Article
10. Steinman RM. Dendritic cells and immune-based therapies. Exp Hematol. 1996. 24:859–862.
11. Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. Int J Cancer. 2001. 94:459–473.
Article
12. Palucka AK, Ueno H, Fay J, Banchereau J. Dendritic cells: a critical player in cancer therapy? J Immunother. 2008. 31:793–805.
Article
13. Melief C. Cancer immunotherapy by dendritic cells. Immunity. 2008. 29:372–383.
Article
14. Kalinski P, Urban J, Narang R, Berk E, Wieckowski E, Muthuswamy R. Dendritic cell-based therapeutic cancer vaccines: what we have and what we need. Future Oncol. 2009. 5:379–390.
Article
15. Kortylewski M, Yu H. Role of Stat3 in suppressing anti-tumor immunity. Curr Opin Immunol. 2008. 20:228–233.
Article
16. Koebel C, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007. 450:903–907.
Article
17. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006. 6:836–848.
Article
18. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature. 2008. 453:410–414.
Article
19. Hamzah J, Nelson D, et al. Vascular targeting of anti-CD40 antibodies and IL-2 into autochthonous tumors enhances immunotherapy in mice. J Clin Invest. 2008. 118:1691–1699.
Article
20. 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.
Article
21. Ballestrero A, Boy D, Moran E, Cirmena G, Brossart P, Nencioni A. Immunotherapy with dendritic cells for cancer. Adv Drug Deliv Rev. 2008. 60:173–183.
Article
22. Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007. 117:1195–1203.
Article
23. Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991. 9:271–296.
Article
24. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998. 392:245–252.
Article
25. Gabrilovich D. Mechanisms and functional significance of tumour induced dendritic-cell defects. Nat Rev Immunol. 2004. 4:941–952.
Article
26. Schuurhuis D, Fu N, Ossendorp F, Melief CJ. Ins and outs of dendritic cells. Int Arch Allergy Immunol. 2006. 140:53–72.
Article
27. Dhodapkar MV, Dhodapkar KM, Palucka AK. Interactions of tumor cells with dendritic cells: balancing immunity and tolerance. Cell Death Differ. 2008. 15:39–50.
Article
28. Boonman ZF, van Mierlo GJ, Fransen MF, de Keizer RJ, Jager MJ, Melief CJ, et al. Maintenance of immune tolerance depends on normal tissue homeostasis. J Immunol. 2005. 175:4247–4254.
Article
29. van Mierlo G, Boonman Z, Dumortier HM, den Boer AT, Fransen MF, Nouta J, et al. Activation of dendritic cells that cross-present tumor-derived antigen licenses CD8+ CTL to cause tumor eradication. J Immunol. 2004. 173:6753–6759.
Article
30. Yang L, Carbone DP. Tumor host immune interactions and dendritic cell dysfunction. Adv Cancer Res. 2004. 92:13–27.
31. Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005. 83:451–461.
Article
32. Uchida K, Schneider S, Yochim JM, Kuramochi H, Hayashi K, Takasaki K, et al. Intratumoral COX-2 gene expression is a predictive factor for colorectal cancer response to fluoropyrimidine-based chemotherapy. Clin Cancer Res. 2005. 11:3363–3368.
Article
33. Williams C, Shattuck-Brandt RL, DuBois RN. The role of COX-2 in intestinal cancer. Ann N Y Acad Sci. 1999. 889:72–83.
Article
34. Inaba T, Sano H, Kawahito Y, Hla T, Akita K, Toda M, et al. Induction of cyclooxygenase-2 in monocyte /macrophage by mucins secreted from colon cancer cells. Proc Natl Acad Sci USA. 2003. 100:2736–2741.
Article
35. Soumaoro LT, Uetake H, Higuchi T, Takagi Y, Enomoto M, Sugihara K. Cyclooxygenase-2 expression: a significant prognostic indicator for patients with colorectal cancer. Clin Cancer Res. 2004. 10:8465–8471.
36. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol. 2001. 166:678–689.
Article
37. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, et al. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res. 2000. 6:1755–1766.
38. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001. 166:5398–5306.
Article
39. Young MR, Wright MA, Lozano Y, Prechel MM, Benefield J, Leonetti JP, et al. Increased recurrence and metastasis in patients whose primary head and neck squamous cell carcinomas secreted granulocyte-macrophage colony stimulating factor and contained CD34+ natural suppressor cells. Int J Cancer. 1997. 74:69–74.
Article
40. Bell D, Chomarat P, Broyles D, Netto G, Harb GM, Lebecque S, et al. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med. 1999. 190:1417–1426.
Article
41. Toriyama K, Wen DR, Paul E, Cochran AJ. Variations in the distribution, frequency, and phenotype of Langerhans cells during the evolution of malignant melanoma of the skin. J Invest Dermatol. 1993. 100:S269–S273.
Article
42. Troy AJ, Summers KL, Davidson PJ, Atkinson CH, Hart DN. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin Cancer Res. 1998. 4:585–593.
Article
43. Zou W, Machelon V, Coulomb-L'Hermin A, Borvak J, Nome F, Isaeva T, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001. 7:1339–1346.
Article
44. Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, et al. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003. 89:1463–1472.
Article
45. Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, et al. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J Immunol. 2004. 172:464–474.
Article
46. Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004. 10:48–54.
Article
47. Kortylewski M, Kujawski M, Wang T, Wei S, Zhang S, Pilon-Thomas S, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005. 11:1314–1321.
Article
48. Ghiringhelli F, Puig PE, Roux S, Parcellier A, Schmitt E, Solary E, et al. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J Exp Med. 2005. 202:919–929.
Article
49. Blaskovich MA, Sun J, Cantor A, Turkson J, Jove R, Sebti SM. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003. 63:1270–1279.
50. van Kester MS, Out-Luiting JJ, von dem Borne PA, Willemze R, Tensen CP, Vermeer MH. Cucurbitacin I inhibits Stat3 and induces apoptosis in Sezary cells. J Invest Dermatol. 2008. 128:1691–1695.
51. Mantovani A, Pierotti MA. Cancer and inflammation: a complex relationship. Cancer Lett. 2008. 267:180–181.
Article
52. Sica A, Bronte V. Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest. 2007. 117:1155–1166.
Article
53. Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumorinduced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008. 222:162–179.
Article
54. Borrello MG, Degl'innocenti D, Pierotti M. Inflammation and cancer: the oncogene-driven connection. Cancer Lett. 2008. 267:262–270.
Article
55. Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008. 267:271–285.
Article
56. Ostrand-Rosenberg S, Sinha P. Myeoid-Derived suppressor cells: linking inflammation and cancer. J Immunol. 2009. 182:4499–4506.
Article
57. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002. 420:860–867.
Article
58. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008. 8:579–591.
Article
59. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008. 13:23–35.
Article
60. Marx J. Cancer immunology. Cancer's bulwark against immune attack: MDS cells. Science. 2008. 319:154–156.
61. Umemura N, Saio M, Suwa T, Kitoh Y, Bai J, Nonaka K, et al. Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J Leukocyte Biol. 2008. 83:1136–1144.
Article
62. Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest. 2006. 116:2777–2790.
Article
63. Rossner S, Voigtlander C, Wiethe C, Hänig J, Seifarth C, Lutz MB. Myeloid dendritic cell precursors generated from bone marrow suppress T cell responses via cell contact and nitric oxide production in vitro. Eur J Immunol. 2005. 35:3533–3544.
64. Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006. 66:1123–1131.
Article
65. Yang R, Cai Z, Zhang Y, Yutzy WH 4th, Roby KF, Roden RB. CD80 in immune suppression by mouse ovarian carcinoma-associated Gr-1+ CD11b+ myeloid cells. Cancer Res. 2006. 66:6807–6815.
Article
66. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumorbearing mice. J Immunol. 2008. 181:5791–5802.
Article
67. Sawanobori Y, Ueha S, Kurachi M, Shimaoka T, Talmadge JE, Abe J, et al. Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice. Blood. 2008. 111:5457–5466.
Article
68. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004. 172:989–999.
Article
69. Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, et al. IL-4-induced arginase 1 suppresses allo-reactive T cells in tumor-bearing mice. J Immunol. 2003. 170:270–278.
Article
70. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007. 67:4507–4513.
Article
71. Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 2003. 24:302–306.
Article
72. Kusmartsev S, Gabrilovich DI. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukocyte Biol. 2003. 74:186–196.
Article
73. Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol. 2007. 25:2546–2553.
Article
74. Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008. 135:234–243.
Article
75. Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, et al. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res. 2005. 65:3044–3048.
Article
76. Srivastava MK, Bosch JJ, Thompson JA, Ksander BR, Edelman MJ, Ostrand-Rosenberg S. Lung cancer patients' CD4(+) T cells are activated in vitro by MHC II cell-based vaccines despite the presence of myeloid-derived suppressor cells. Cancer Immunol Immunother. 2008. 57:1493–1504.
Article
77. Bronte V, Wang M, Overwijk WW, Surman DR, Pericle F, Rosenberg SA, et al. Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1 +/Gr-1+ cells. J Immunol. 1998. 161:5313–5320.
78. Bronte V, Apolloni E, Cabrelle A, Ronca R, Serafini P, Zamboni P, et al. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood. 2000. 96:3838–3846.
Article
79. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001. 166:5398–5406.
Article
80. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. 2002. 168:689–695.
Article
81. Terabe M, Swann J, Ambrosino E, Sinha P, Takaku S, Hayakawa Y, et al. A non-classical non-Valpha 14Jalpha 18 CD1d-restricted (type II) NKT cell is sufficient for down-regulation of tumor immunosurveillance. J Exp Med. 2005. 202:1627–1633.
Article
82. Baxevanis CN, Perez SA, Papamichail M. Combinatorial treatments including vaccines, chemotherapy and monoclonal antibodies for cancer therapy. Cancer Immunol Immunother. 2009. 58:317–324.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr