Skip to main content

Advertisement

Log in

Overcoming immunosuppression in the melanoma microenvironment induced by chronic inflammation

  • Focussed Research Review
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Malignant melanoma is known by its rapid progression and poor response to currently applied treatments. Despite the well-documented melanoma immunogenicity, the results of immunotherapeutic clinical trials are not satisfactory. This poor antitumor reactivity is due to the development of chronic inflammation in the tumor microenvironment characterized by infiltrating leukocytes and soluble mediators, which lead to an immunosuppression associated with cancer progression. Using the ret transgenic mouse melanoma model that closely resembles human melanoma, we demonstrated increased levels of chronic inflammatory factors in skin tumors and metastatic lymph nodes, which correlated with tumor progression. Furthermore, Gr1+CD11b+ myeloid-derived suppressor cells (MDSC), known to block tumor-reactive T cells, were enriched in melanoma lesions and showed an enhanced immunosuppressive capacity. This MDSC accumulation was associated with a strong TCR ζ-chain downregulation in T cells suggesting that the tumor inflammatory microenvironment supports MDSC recruitment and immunosuppressive activity. Indeed, upon administration of phosphodiesterase-5 inhibitor sildenafil or paclitaxel in non-cytotoxic doses, we observed reduced levels of chronic inflammatory mediators in association with decreased MDSC amounts and immunosuppressive function. This led to a partial restoration of ζ-chain expression in T cells and to a significantly increased survival of tumor-bearing mice. CD8 T-cell depletion resulted in an abrogation of beneficial outcome of both drugs, suggesting the involvement of MDSC and CD8 T cells in the observed therapeutic effects. Our data imply that inhibition of chronic inflammation in the tumor microenvironment should be applied in conjunction with melanoma immunotherapies to increase their efficacy.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. MacKie RM, Hauschild A, Eggermont AM (2009) Epidemiology of invasive cutaneous melanoma. Ann Oncol 20(Suppl 6):vi1–vi7

    Article  PubMed  Google Scholar 

  2. Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, Grob JJ, Malvehy J, Newton-Bishop J, Stratigos A, Pehamberger H, Eggermont A (2010) Diagnosis and treatment of melanoma: European consensus-based interdisciplinary guideline. Eur J Cancer 46:270–283

    Article  PubMed  Google Scholar 

  3. Callahan MK, Wolchok JD, Allison JP (2010) Anti-CTLA-4 antibody therapy: immune monitoring during clinical development of a novel immunotherapy. Semin Oncol 37:473–484

    Article  PubMed  CAS  Google Scholar 

  4. Pandolfi F, Cianci R, Lolli S, Dunn IS, Newton EE, Haggerty TJ, Boyle LA, Kurnick JT (2008) Strategies to overcome obstacles to successful immunotherapy of melanoma. Int J Immunopathol Pharmacol 21:493–500

    PubMed  CAS  Google Scholar 

  5. Parmiani G, Castelli C, Santinami M, Rivoltini L (2007) Melanoma immunology: past, present and future. Curr Opin Oncol 19:121–127

    Article  PubMed  Google Scholar 

  6. Fujii S, Shimizu K, Hemmi H, Steinman RM (2007) Innate Valpha14(+) natural killer T cells mature dendritic cells, leading to strong adaptive immunity. Immunol Rev 220:183–198

    Article  PubMed  CAS  Google Scholar 

  7. Dissemond J, Kothen T, Mörs J, Weimann TK, Lindeke A, Goos M, Wagner SN (2003) Downregulation of tapasin expression in progressive human malignant melanoma. Arch Dermatol Res 295:43–49

    Article  PubMed  CAS  Google Scholar 

  8. Ferrone S, Marincola FM (1995) Loss of HLA class I antigens by melanoma cells: molecular mechanisms, functional significance and clinical relevance. Immunol Today 16:487–494

    Article  PubMed  CAS  Google Scholar 

  9. Burke S, Lakshmikanth T, Colucci F, Carbone E (2010) New views on natural killer cell-based immunotherapy for melanoma treatment. Trends Immunol 31:339–345

    Article  PubMed  CAS  Google Scholar 

  10. Ostrand-Rosenberg S (2008) Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev 18:11–18

    Article  PubMed  CAS  Google Scholar 

  11. Lázár-Molnár E, Hegyesi H, Tóth S, Falus A (2000) Autocrine and paracrine regulation by cytokines and growth factors in melanoma. Cytokine 12:547–554

    Article  PubMed  Google Scholar 

  12. Antony PA, Restifo NP (2005) CD4+CD25+ T regulatory cells, immunotherapy of cancer, and interleukin-2. J Immunother 28:120–128

    Article  PubMed  CAS  Google Scholar 

  13. Zou W (2005) Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5:263–274

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  15. Ostrand-Rosenberg S (2010) Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother 59:1593–1600

    Article  PubMed  Google Scholar 

  16. Shurin MR, Naiditch H, Zhong H, Shurin GV (2011) Regulatory dendritic cells: new targets for cancer immunotherapy. Cancer Biol Ther 11:988–992

    Article  PubMed  Google Scholar 

  17. Ben-Neriah Y, Karin M (2011) Inflammation meets cancer, with NF-κB as the matchmaker. Nat Immunol 12:715–723

    Article  PubMed  CAS  Google Scholar 

  18. Rook GA, Dalgleish A (2011) Infection, immunoregulation, and cancer. Immunol Rev 240:141–159

    Article  PubMed  CAS  Google Scholar 

  19. Cramer DW, Finn OJ (2011) Epidemiologic perspective on immune-surveillance in cancer. Curr Opin Immunol 23:265–271

    Article  PubMed  CAS  Google Scholar 

  20. Mantovani A (2010) Molecular pathways linking inflammation and cancer. Curr Mol Med 10:369–373

    Article  PubMed  CAS  Google Scholar 

  21. Allavena P, Germano G, Marchesi F, Mantovani A (2011) Chemokines in cancer related inflammation. Exp Cell Res 317:664–673

    Article  PubMed  CAS  Google Scholar 

  22. Baniyash M (2006) Chronic inflammation, immunosuppression and cancer: new insights and outlook. Semin Cancer Biol 16:80–88

    Article  PubMed  CAS  Google Scholar 

  23. Tan TT, Coussens LM (2007) Humoral immunity, inflammation and cancer. Curr Opin Immunol 19:209–216

    Article  PubMed  CAS  Google Scholar 

  24. Sparmann A, Bar-Sagi D (2004) Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 6:447–458

    Article  PubMed  CAS  Google Scholar 

  25. Sumimoto H, Imabayashi F, Iwata T, Kawakami Y (2006) The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med 203:1651–1656

    Article  PubMed  CAS  Google Scholar 

  26. Haluska F, Pemberton T, Ibrahim N, Kalinsky K (2007) The RTK/RAS/BRAF/PI3K pathways in melanoma: biology, small molecule inhibitors, and potential applications. Semin Oncol 34:546–554

    Article  PubMed  CAS  Google Scholar 

  27. Lomas J, Martin-Duque P, Pons M, Quintanilla M (2008) The genetics of malignant melanoma. Front Biosci 13:5071–5093

    Article  PubMed  CAS  Google Scholar 

  28. Kato M, Takahashi M, Akhand AA, Liu W, Dai Y, Shimizu S, Iwamoto T, Suzuki H, Nakashima I (1999) Transgenic mouse model for skin malignant melanoma. Oncogene 17:1885–1888

    Article  Google Scholar 

  29. Eng C (1999) RET proto-oncogene in the development of human cancer. J Clin Oncol 17:380–383

    PubMed  CAS  Google Scholar 

  30. Umansky V, Abschuetz O, Osen W, Ramacher M, Zhao F, Kato M, Schadendorf D (2008) Melanoma-specific memory T cells are functionally active in ret transgenic mice without macroscopic tumors. Cancer Res 68:9451–9458

    Article  PubMed  CAS  Google Scholar 

  31. Houghton A, Polsky D (2002) Focus on melanoma. Cancer Cell 2:275–278

    Article  PubMed  CAS  Google Scholar 

  32. Zhao F, Falk C, Osen W, Kato M, Schadendorf D, Umansky V (2009) Activation of p38 mitogen-activated protein kinase drives dendritic cells to become tolerogenic in ret transgenic mice spontaneously developing melanoma. Clin Cancer Res 15:4382–4390

    Article  PubMed  CAS  Google Scholar 

  33. Meyer C, Sevko A, Ramacher M, Bazhin AV, Falk CS, Osen W, Borrello I, Kato M, Schadendorf D, Baniyash M, Umansky V (2011) Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proc Natl Acad Sci USA 108:17111–17116

    Article  PubMed  CAS  Google Scholar 

  34. Kimpfler S, Sevko A, Ring S, Falk C, Osen W, Frank K, Kato M, Mahnke K, Schadendorf D, Umansky V (2009) Skin melanoma development in ret transgenic mice despite the depletion of CD25+Foxp3+ regulatory T cells in lymphoid organs. J Immunol 183:6330–6337

    Article  PubMed  CAS  Google Scholar 

  35. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, Ugel S, Sonda N, Bicciato S, Falisi E, Calabrese F, Basso G, Zanovello P, Cozzi E, Mandruzzato S, Bronte V (2010) Tumor-induced tolerance and immune suppression depend on the C/EBPb transcription factor. Immunity 32:790–802

    Article  PubMed  CAS  Google Scholar 

  36. Bronstein-Sitton N, Vaknin I, Ezernitchi AV, Leshem B, Halabi A, Houri-Hadad Y, Greenbaum E, Zakay-Rones Z, Shapira L, Baniyash M (2003) Sustained exposure to bacterial antigen induces interferon gamma-dependent T cell receptor zeta down-regulation and impaired T cell function. Nat Immunol 4:957–964

    Article  PubMed  CAS  Google Scholar 

  37. Rössner S, Voigtländer C, Wiethe C, Hänig J, Seifarth C, Lutz MB (2005) Myeloid dendritic cell precursors generated from bone marrow suppress T cell responses via cell contact and nitric oxide production in vitro. Eur J Immunol 35:3533–3544

    Article  PubMed  Google Scholar 

  38. Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, Kelly-Scumpia KM, O’Malley KA, Wynn JL, Antonenko S, Al-Quran SZ, Swan R, Chung CS, Atkinson MA, Ramphal R, Gabrilovich DI, Reeves WH, Ayala A, Phillips J, Laface D, Heyworth PG, Clare-Salzler M, Moldawer LL (2007) MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med 204:1463–1474

    Article  PubMed  CAS  Google Scholar 

  39. Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, Mandruzzato S, Bronte V (2010) Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol 22:238–244

    Article  PubMed  CAS  Google Scholar 

  40. Bronte V, Zanovello P (2005) Regulation of immune responses by l-arginine metabolism. Nat Rev Immunol 5:641–654

    Article  PubMed  CAS  Google Scholar 

  41. Rodríguez PC, Ochoa AC (2006) T cell dysfunction in cancer: role of myeloid cells and tumor cells regulating amino acid availability and oxidative stress. Semin Cancer Biol 16:66–72

    Article  PubMed  Google Scholar 

  42. Umansky V, Schirrmacher V (2001) Nitric oxide-induced apoptosis in tumor cells. Adv Cancer Res 82:107–131

    Article  PubMed  CAS  Google Scholar 

  43. Bogdan C (2001) Nitric oxide and the immune response. Nat Immunol 2:907–916

    Article  PubMed  CAS  Google Scholar 

  44. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI (2010) Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol 184:3106–3116

    Article  PubMed  CAS  Google Scholar 

  45. Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D, De Palma A, Mauri P, Monegal A, Rescigno M, Savino B, Colombo P, Jonjic N, Pecanic S, Lazzarato L, Fruttero R, Gasco A, Bronte V, Viola A (2011) Chemokine nitrosylation prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 208:1949–1962

    Article  PubMed  CAS  Google Scholar 

  46. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S (2010) Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70:68–77

    Article  PubMed  CAS  Google Scholar 

  47. Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S (2009) Myeloid-derived suppressor cells down-regulate l-selectin expression on CD4+ and CD8+ T cells. J Immunol 183:937–944

    Article  PubMed  CAS  Google Scholar 

  48. Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, Castelli C, Mariani L, Parmiani G, Rivoltini L (2007) 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 25:2546–2553

    Article  PubMed  CAS  Google Scholar 

  49. Poschke I, Mougiakakos D, Hansson J, Masucci GV, Kiessling R (2010) Immature immunosuppressive CD14+HLA-DR-/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res 70:4335–4345

    Article  PubMed  CAS  Google Scholar 

  50. Ezernitchi AV, Vaknin I, Cohen-Daniel L, Levy O, Manaster E, Halabi A, Pikarsky E, Shapira L, Baniyash M (2006) TCR zeta down-regulation under chronic inflammation is mediated by myeloid suppressor cells differentially distributed between various lymphatic organs. J Immunol 177:4763–4772

    PubMed  CAS  Google Scholar 

  51. Rodríguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC (2002) Regulation of T cell receptor CD3zeta chain expression by l-arginine. J Biol Chem 277:21123–21129

    Article  PubMed  Google Scholar 

  52. Baniyash M (2004) TCR zeta-chain downregulation: curtailing an excessive inflammatory immune response. Nat Rev Immunol 4:675–687

    Article  PubMed  CAS  Google Scholar 

  53. Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Higashi H, Iwashige H, Aridome K, Hokita S, Aikou T (2002) CD3-zeta chain expression of intratumoral lymphocytes is closely related to survival in gastric carcinoma patients. Cancer 94:1437–1442

    Article  PubMed  CAS  Google Scholar 

  54. Whiteside TL (2004) Down-regulation of zeta-chain expression in T cells: a biomarker of prognosis in cancer? Cancer Immunol Immunother 53:865–878

    PubMed  CAS  Google Scholar 

  55. Ugel S, Delpozzo F, Desantis G, Papalini F, Simonato F, Sonda N, Zilio S, Bronte V (2009) Therapeutic targeting of myeloid-derived suppressor cells. Curr Opin Pharmacol 9:470–481

    Article  PubMed  CAS  Google Scholar 

  56. Mirza N, Fishman M, Fricke I, Dunn M, Neuger AM, Frost TJ, Lush RM, Antonia S, Gabrilovich DI (2006) All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res 66:9299–9307

    Article  PubMed  CAS  Google Scholar 

  57. Pan PY, Wang GX, Yin B, Ozao J, Ku T, Divino CM, Chen SH (2008) Reversion of immune tolerance in advanced malignancy: modulation of myeloid-derived suppressor cell development by blockade of stem-cell factor function. Blood 111:219–228

    Article  PubMed  CAS  Google Scholar 

  58. Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, Martin F, Apetoh L, Rébé C, Ghiringhelli F (2010) 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 70:3052–3061

    Article  PubMed  CAS  Google Scholar 

  59. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11:6713–6721

    Article  PubMed  CAS  Google Scholar 

  60. Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, Dolcetti L, Bronte V, Borrello I (2006) Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 203:2691–2702

    Article  PubMed  CAS  Google Scholar 

  61. De Santo C, Serafini P, Marigo I, Dolcetti L, Bolla M, Del Soldato P, Melani C, Guiducci C, Colombo MP, Iezzi M, Musiani P, Zanovello P, Bronte V (2005) Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc Natl Acad Sci USA 102:4185–4190

    Article  PubMed  Google Scholar 

  62. Sinha P, Clements VK, Ostrand-Rosenberg S (2005) Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol 174:636–645

    PubMed  CAS  Google Scholar 

  63. Ghofrani HA, Osterloh IH, Grimminger F (2006) Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat Rev Drug Discov 5:689–702

    Article  PubMed  CAS  Google Scholar 

  64. Serafini P, Mgebroff S, Noonan K, Borrello I (2008) Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res 68:5439–5449

    Article  PubMed  CAS  Google Scholar 

  65. Capuano G, Rigamonti N, Grioni M, Freschi M, Bellone M (2009) Modulators of arginine metabolism support cancer immunosurveillance. BMC Immunol 10:1

    Article  PubMed  Google Scholar 

  66. Cheng P, Corzo CA, Luetteke N, Yu B, Nagaraj S, Bui MM, Ortiz M, Nacken W, Sorg C, Vogl T, Roth J, Gabrilovich DI (2008) Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J Exp Med 205:2235–2249

    Article  PubMed  CAS  Google Scholar 

  67. Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-Rosenberg S, Srikrishna G (2008) Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J Immunol 181:4666–4675

    PubMed  CAS  Google Scholar 

  68. Tartour E, Pere H, Maillere B, Terme M, Merillon N, Taieb J, Sandoval F, Quintin-Colonna F, Lacerda K, Karadimou A, Badoual C, Tedgui A, Fridman WH, Oudard S (2011) Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 30:83–95

    Article  PubMed  CAS  Google Scholar 

  69. Laties A, Zrenner E (2002) Viagra (sildenafil citrate) and ophthalmology. Prog Retin Eye Res 21:485–506

    Article  PubMed  CAS  Google Scholar 

  70. Nowak AK, Lake RA, Robinson BW (2006) Combined chemoimmunotherapy of solid tumours: improving vaccines? Adv Drug Deliv Rev 58:975–990

    Article  PubMed  CAS  Google Scholar 

  71. Zitvogel L, Kepp O, Kroemer G (2011) Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol 8:151–160

    Article  PubMed  CAS  Google Scholar 

  72. Kaneno R, Shurin GV, Kaneno FM, Naiditch H, Luo J, Shurin MR (2011) Chemotherapeutic agents in low non-cytotoxic concentrations increase immunogenicity of human colon cancer cells. Cell Oncol (Dordr) 34:97–106

    CAS  Google Scholar 

  73. Kaneno R, Shurin GV, Tourkova IL, Shurin MR (2009) Chemomodulation of human dendritic cell function by anti-neoplastic agents in low non-cytotoxic concentrations. J Transl Med 7:58

    Article  PubMed  Google Scholar 

  74. Shurin GV, Tourkova IL, Kaneno R, Shurin MR (2009) Chemotherapeutic agents in non-cytotoxic concentrations increase antigen presentation by dendritic cells via an IL-12-dependent mechanism. J Immunol 183:137–144

    Article  PubMed  CAS  Google Scholar 

  75. Shurin GV, Tourkova IL, Shurin MR (2008) Low-dose chemotherapeutic agents regulate small rho GTPase activity in dendritic cells. J Immunother 31:491–499

    Article  PubMed  CAS  Google Scholar 

  76. Zhong H, Han B, Tourkova IL, Lokshin A, Rosenbloom A, Shurin MR, Shurin GV (2007) Low-dose paclitaxel prior to intratumoral dendritic cell vaccine modulates intratumoral cytokine network and lung cancer growth. Clin Cancer Res 13:5455–5462

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This project has been funded by the DKFZ-MOST Cooperation in Cancer Research (grant CA128, to Viktor Umansky), Dr. Mildred Scheel Foundation for Cancer Research (grant 108992, to Viktor Umansky), the Initiative and Networking Fund of the Helmholtz Association within the Helmholtz Alliance on Immunotherapy of Cancer (to Viktor Umansky).

Conflict of interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Viktor Umansky.

Additional information

This paper is a Focussed Research Review based on a presentation given at the Second International Conference on Cancer Immunotherapy and Immunomonitoring (CITIM 2011), held in Budapest, Hungary, 2nd–5th May 2011. It is part of a CII series of Focussed Research Reviews and meeting report.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Umansky, V., Sevko, A. Overcoming immunosuppression in the melanoma microenvironment induced by chronic inflammation. Cancer Immunol Immunother 61, 275–282 (2012). https://doi.org/10.1007/s00262-011-1164-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-011-1164-6

Keywords

Navigation