Skip to main content

The Human Tumor Microenvironment

  • Chapter
  • First Online:

Abstract

For a long time, cancer therapy has had as its sole objective the direct elimination of tumor cells. In case of nonmetastatic disease, this is accomplished by surgery, which removes the primary tumor. Radiotherapy and conventional chemotherapies also aimed at targeting tumor cells preferentially. The high capacity of tumor cells to divide as compared to the normal cells makes them more sensitive to agents that physically, in the case of radiotherapy, or chemically, in the case of chemotherapy, attack DNA and lead to cell death. Chemotherapies targeting mutations in tumor cells such as BRAF have been developed as well. However, these approaches also destroy the nonmalignant cells and have local and/or systemic consequences. To increase specificity toward the tumor cells, cytotoxic agents have been coupled to antibodies that bind to the tumor cells in order to allow their specific targeting to the tumor and not to the normal cells. However, the entry of such constructs into tumors still remains a major issue.

The progresses that have been accomplished in the field of tumor immunology in these last 20 years have led to a drastic change in the representation of primary tumors and metastases and to cancer treatments. Tumors are not anymore represented as a simple accumulation of cells that have undergone oncogenic processes but as a complex and dynamic structure made of tumor cells and inflamed tissue. Tumors are infiltrated with blood vessels that bring nutriments and all kinds of leukocytes inside the tumor and at its periphery, in the so-called tumor stroma that also contains matrix proteins such as collagen fibers.

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

References

  1. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137–48.

    Article  CAS  PubMed  Google Scholar 

  2. Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194(4260):23–8.

    Article  CAS  PubMed  Google Scholar 

  3. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339(6127):1546–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  PubMed  Google Scholar 

  5. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481(7381):306–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Branzei D, Foiani M. Regulation of DNA repair throughout the cell cycle. Nat Rev Mol Cell Biol. 2008;9(4):297–308.

    Article  CAS  PubMed  Google Scholar 

  7. Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13(11):759–71.

    Article  CAS  PubMed  Google Scholar 

  8. Giraldo NA, Becht E, Vano Y, Sautès-Fridman C, Fridman WH. The immune response in cancer: from immunology to pathology to immunotherapy. Virchows Arch Int J Pathol. 2015;467(2):127–35.

    Article  CAS  Google Scholar 

  9. Crusz SM, Balkwill FR. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015;12(10):584–96.

    Article  CAS  PubMed  Google Scholar 

  10. Moore PS, Chang Y. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer. 2010;10(12):878–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12(4):298–306.

    Article  CAS  PubMed  Google Scholar 

  12. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11(10):889–96.

    Article  CAS  PubMed  Google Scholar 

  13. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39(4):782–95.

    Article  CAS  PubMed  Google Scholar 

  14. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–10.

    Article  CAS  PubMed  Google Scholar 

  15. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12(4):265–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. 2016;37(12):855–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Giraldo NA, Becht E, Pagès F, Skliris G, Verkarre V, Vano Y, et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res. 2015;21(13):3031–40.

    Article  CAS  PubMed  Google Scholar 

  18. Sautès-Fridman C, Lawand M, Giraldo NA, Kaplon H, Germain C, Fridman WH, et al. Tertiary lymphoid structures in cancers: prognostic value, regulation, and manipulation for therapeutic intervention. Front Immunol. 2016;7:407.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Dieu-Nosjean M-C, Goc J, Giraldo NA, Sautès-Fridman C, Fridman WH. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 2014;35(11):571–80.

    Article  CAS  PubMed  Google Scholar 

  20. de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, et al. Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res. 2011;71(20):6391–9.

    Article  PubMed  Google Scholar 

  21. Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, et al. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 2014;189(7):832–44.

    Article  CAS  PubMed  Google Scholar 

  22. Becht E, Giraldo NA, Germain C, de Reyniès A, Laurent-Puig P, Zucman-Rossi J, et al. Immune contexture, immunoscore, and malignant cell molecular subgroups for prognostic and theranostic classifications of cancers. Adv Immunol. 2016;130:95–190.

    Article  PubMed  Google Scholar 

  23. Vignali DAA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8(7):523–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang N, Bevan MJ. CD8(+) T cells: foot soldiers of the immune system. Immunity. 2011;35(2):161–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Speiser DE, Ho P-C, Verdeil G. Regulatory circuits of T cell function in cancer. Nat Rev Immunol. 2016;16(10):599–611.

    Article  CAS  PubMed  Google Scholar 

  26. Balkwill F, Montfort A, Capasso M. B regulatory cells in cancer. Trends Immunol. 2013;34(4):169–73.

    Article  CAS  PubMed  Google Scholar 

  27. Dieu-Nosjean M-C, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008;26(27):4410–7.

    Article  CAS  PubMed  Google Scholar 

  28. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313(5795):1960–4.

    Article  CAS  PubMed  Google Scholar 

  29. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. N Engl J Med. 2003;348(3):203–13.

    Google Scholar 

  30. Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353(25):2654–66.

    Article  PubMed  Google Scholar 

  31. Remark R, Alifano M, Cremer I, Lupo A, Dieu-Nosjean M-C, Riquet M, et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin. Clin Cancer Res. 2013;19(15):4079–91.

    Article  CAS  PubMed  Google Scholar 

  32. Tian C, Lu S, Fan Q, Zhang W, Jiao S, Zhao X, et al. Prognostic significance of tumor-infiltrating CD8+ or CD3+ T lymphocytes and interleukin-2 expression in radically resected non-small cell lung cancer. Chin Med J. 2015;128(1):105–10.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Che Y-Q, Feng L, Rong W-Q, Shen D, Wang Q, Yang L, et al. Correlation analysis of peripheral blood T cell subgroups, immunoglobulin and prognosis of early hepatocellular carcinoma after hepatectomy. Int J Clin Exp Med. 2014;7(11):4282–90.

    PubMed  PubMed Central  Google Scholar 

  34. Ness N, Andersen S, Valkov A, Nordby Y, Donnem T, Al-Saad S, et al. Infiltration of CD8+ lymphocytes is an independent prognostic factor of biochemical failure-free survival in prostate cancer. Prostate. 2014;74(14):1452–61.

    Article  CAS  PubMed  Google Scholar 

  35. Petitprez F, Fossati N, Vano Y, Freschi M, Becht E, Lucianò R, et al. European Urology Focus, 2017, ISSN 2405-4569, http://dx.doi.org/10.1016/j.euf.2017.05.013. (http://www.sciencedirect.com/science/article/pii/S2405456917301517)

  36. Mlecnik B, Bindea G, Angell HK, Maby P, Angelova M, Tougeron D, et al. Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity. 2016;44(3):698–711.

    Article  CAS  PubMed  Google Scholar 

  37. Galon J, Mlecnik B, Marliot F, Ou F-S, Bifulco CB, Lugli A, et al. Validation of the Immunoscore (IM) as a prognostic marker in stage I/II/III colon cancer: Results of a worldwide consortium-based analysis of 1,336 patients. J Clin Oncol. 2016 34:15_suppl, 3500–3500.

    Google Scholar 

  38. Kirilovsky A, Marliot F, El Sissy C, Haicheur N, Galon J, Pagès F. Rational bases for the use of the immunoscore in routine clinical settings as a prognostic and predictive biomarker in cancer patients. Int Immunol. 2016;28(8):373–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dieu-Nosjean M-C, Giraldo NA, Kaplon H, Germain C, Fridman WH, Sautès-Fridman C. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev. 2016;271(1):260–75.

    Article  CAS  PubMed  Google Scholar 

  40. Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013;123(7):2873–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Boissière-Michot F, Lazennec G, Frugier H, Jarlier M, Roca L, Duffour J, et al. Characterization of an adaptive immune response in microsatellite-instable colorectal cancer. Oncoimmunology. 2014;3:e29256.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Fridman WH, Remark R, Goc J, Giraldo NA, Becht E, Hammond SA, et al. The immune microenvironment: a major player in human cancers. Int Arch Allergy Immunol. 2014;164(1):13–26.

    Article  CAS  PubMed  Google Scholar 

  43. Mahmoud SMA, Lee AHS, Paish EC, Macmillan RD, Ellis IO, Green AR. The prognostic significance of B lymphocytes in invasive carcinoma of the breast. Breast Cancer Res Treat. 2012;132(2):545–53.

    Article  CAS  PubMed  Google Scholar 

  44. van Herpen CML, van der Voort R, van der Laak JAWM, Klasen IS, de Graaf AO, van Kempen LCL, et al. Intratumoral rhIL-12 administration in head and neck squamous cell carcinoma patients induces B cell activation. Int J Cancer. 2008;123(10):2354–61.

    Article  PubMed  Google Scholar 

  45. Santoiemma PP, Reyes C, Wang L-P, McLane MW, Feldman MD, Tanyi JL, et al. Systematic evaluation of multiple immune markers reveals prognostic factors in ovarian cancer. Gynecol Oncol. 2016;143(1):120–7.

    Article  CAS  PubMed  Google Scholar 

  46. Berntsson J, Nodin B, Eberhard J, Micke P, Jirström K. Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer. Int J Cancer. 2016;139(5):1129–39.

    Article  CAS  PubMed  Google Scholar 

  47. Goeppert B, Frauenschuh L, Zucknick M, Stenzinger A, Andrulis M, Klauschen F, et al. Prognostic impact of tumour-infiltrating immune cells on biliary tract cancer. Br J Cancer. 2013;109(10):2665–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Garg K, Maurer M, Griss J, Brüggen M-C, Wolf IH, Wagner C, et al. Tumor-associated B cells in cutaneous primary melanoma and improved clinical outcome. Hum Pathol. 2016;54:157–64.

    Article  CAS  PubMed  Google Scholar 

  49. Yuseff M-I, Pierobon P, Reversat A, Lennon-Duménil A-M. How B cells capture, process and present antigens: a crucial role for cell polarity. Nat Rev Immunol. 2013;13(7):475–86.

    Article  CAS  PubMed  Google Scholar 

  50. Carmi Y, Spitzer MH, Linde IL, Burt BM, Prestwood TR, Perlman N, et al. Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature. 2015;521(7550):99–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Silva NSD, Klein U. Dynamics of B cells in germinal centres. Nat Rev Immunol. 2015;15(3):137.

    Article  PubMed  PubMed Central  Google Scholar 

  52. de Wit J, Souwer Y, Jorritsma T, Klaasse Bos H, ten Brinke A, Neefjes J, et al. Antigen-specific B cells reactivate an effective cytotoxic T cell response against phagocytosed Salmonella through cross-presentation. PLoS One. 2010;5(9):e13016.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Deola S, Panelli MC, Maric D, Selleri S, Dmitrieva NI, Voss CY, et al. Helper B cells promote cytotoxic T cell survival and proliferation independently of antigen presentation through CD27/CD70 interactions. J Immunol. 2008;180(3):1362–72.

    Article  CAS  PubMed  Google Scholar 

  54. Barbera-Guillem E, Nelson MB, Barr B, Nyhus JK, May KF, Feng L, et al. B lymphocyte pathology in human colorectal cancer. Experimental and clinical therapeutic effects of partial B cell depletion. Cancer Immunol Immunother (CII). 2000;48(10):541–9.

    Article  CAS  Google Scholar 

  55. DeNardo DG, Andreu P, Coussens LM. Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity. Cancer Metastasis Rev. 2010;29(2):309–16.

    Article  PubMed  PubMed Central  Google Scholar 

  56. de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell. 2005;7(5):411–23.

    Article  PubMed  Google Scholar 

  57. Barbera-Guillem E, May KF, Nyhus JK, Nelson MB. Promotion of tumor invasion by cooperation of granulocytes and macrophages activated by anti-tumor antibodies. Neoplasia. 1999;1(5):453–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Olkhanud PB, Damdinsuren B, Bodogai M, Gress RE, Sen R, Wejksza K, et al. Tumor-evoked regulatory B cells promote breast cancer metastasis by converting resting CD4+ T cells to T-regulatory cells. Cancer Res. 2011;71(10):3505–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Meshcheryakova A, Tamandl D, Bajna E, Stift J, Mittlboeck M, Svoboda M, et al. B cells and ectopic follicular structures: novel players in anti-tumor programming with prognostic power for patients with metastatic colorectal cancer. PLoS One. 2014;9(6):e99008.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Wirsing AM, Rikardsen OG, Steigen SE, Uhlin-Hansen L, Hadler-Olsen E. Characterisation and prognostic value of tertiary lymphoid structures in oral squamous cell carcinoma. BMC Clin Pathol. 2014;14:38.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Algars A, Irjala H, Vaittinen S, Huhtinen H, Sundström J, Salmi M, et al. Type and location of tumor-infiltrating macrophages and lymphatic vessels predict survival of colorectal cancer patients. Int J Cancer. 2012;131(4):864–73.

    Article  PubMed  Google Scholar 

  62. Shu Q-H, Ge Y-S, Ma H-X, Gao X-Q, Pan J-J, Liu D, et al. Prognostic value of polarized macrophages in patients with hepatocellular carcinoma after curative resection. J Cell Mol Med. 2016;20(6):1024–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shimura S, Yang G, Ebara S, Wheeler TM, Frolov A, Thompson TC. Reduced infiltration of tumor-associated macrophages in human prostate cancer: association with cancer progression. Cancer Res. 2000;60(20):5857–61.

    CAS  PubMed  Google Scholar 

  64. Heller DS, Hameed M, Cracchiolo B, Wiederkehr M, Scott D, Skurnick J, et al. Int J Gynecol Cancer. 2003;13(1):67–70.

    Google Scholar 

  65. Čermáková P, Melichar B, Tomšová M, Zoul Z, Kalábová H, Spaček J, et al. Prognostic significance of CD3+ tumor-infiltrating lymphocytes in patients with endometrial carcinoma. Anticancer Res. 2014;34(10):5555–61.

    PubMed  Google Scholar 

  66. Wang XL, Jiang JT, Wu CP. Prognostic significance of tumor-associated macrophage infiltration in gastric cancer: a meta-analysis. Genet Mol Res (GMR). 2016;15(4) doi:10.4238/gmr15049040.

  67. Sjödahl G, Lövgren K, Lauss M, Chebil G, Patschan O, Gudjonsson S, et al. Infiltration of CD3+ and CD68+ cells in bladder cancer is subtype specific and affects the outcome of patients with muscle-invasive tumors. Urol Oncol. 2014;32(6):791–7.

    Article  PubMed  Google Scholar 

  68. Dong P, Ma L, Liu L, Zhao G, Zhang S, Dong L, et al. CD86+/CD206+, diametrically polarized tumor-associated macrophages, predict hepatocellular carcinoma patient prognosis. Int J Mol Sci. 2016;17(3):320.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Jensen TO, Schmidt H, Møller HJ, Høyer M, Maniecki MB, Sjoegren P, et al. Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma. J Clin Oncol. 2009;27(20):3330–7.

    Article  PubMed  Google Scholar 

  70. Medrek C, Pontén F, Jirström K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Colvin EK. Tumor-associated macrophages contribute to tumor progression in ovarian cancer. Front Oncol. 2014;4:137.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Mei J, Xiao Z, Guo C, Pu Q, Ma L, Liu C, et al. Prognostic impact of tumor-associated macrophage infiltration in non-small cell lung cancer: a systemic review and meta-analysis. Oncotarget. 2016;7(23):34217–28.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17(1):218.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Martin-Liberal J, de Olza MO, Hierro C, Gros A, Rodon J, Tabernero J. The expanding role of immunotherapy. Cancer Treat Rev. 2017;54:74–86. doi:10.1016/j.ctrv.2017.01.008. Epub 2017 Feb 11. Review.

  75. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJM, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Chen P-L, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6(8):827–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Daud AI, Loo K, Pauli ML, Sanchez-Rodriguez R, Sandoval PM, Taravati K, et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. J Clin Invest. 2016;126(9):3447–52.

    Article  PubMed  PubMed Central  Google Scholar 

  79. McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, et al. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase IA study. J Clin Oncol. 2016;34(8):833–42.

    Article  CAS  PubMed  Google Scholar 

  80. Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Liu X-D, Hoang A, Zhou L, Kalra S, Yetil A, Sun M, et al. Resistance to antiangiogenic therapy is associated with an immunosuppressive tumor microenvironment in metastatic renal cell carcinoma. Cancer Immunol Res. 2015;3(9):1017–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet A-L, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212(2):139–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Beuselinck B, Job S, Becht E, Karadimou A, Verkarre V, Couchy G, et al. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin Cancer Res. 2015;21(6):1329–39.

    Article  CAS  PubMed  Google Scholar 

  84. Becht E, Giraldo NA, Beuselinck B, Job S, Marisa L, Vano Y, et al. Prognostic and theranostic impact of molecular subtypes and immune classifications in renal cell cancer (RCC) and colorectal cancer (CRC). Oncoimmunology. 2015;4(12):e1049804.

    Article  PubMed  PubMed Central  Google Scholar 

  85. A BIOmarker driven trial with Nivolumab and Ipilimumab or VEGFR tKi in Naïve Metastatic Kidney Cancer - full text view - ClinicalTrials.gov [Internet]. [cited 2017 Feb 28]. Available from: https://clinicaltrials.gov/ct2/show/NCT02960906

  86. Fridman WH, Zitvogel L, Sautès-Fridman C, et al. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017 Jul 25. doi:10.1038/nrclinonc.2017.101. [Epub ahead of print] Review.

Download references

Acknowledgments

The authors thank all colleagues from the Cremer/Teillaud team, pathologists, clinicians, and patients who participated in the studies on the human tumor microenvironment described in this chapter.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Catherine Sautès-Fridman .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Vano, Y., Giraldo, N.A., Fridman, W.H., Sautès-Fridman, C. (2018). The Human Tumor Microenvironment. In: Zitvogel, L., Kroemer, G. (eds) Oncoimmunology. Springer, Cham. https://doi.org/10.1007/978-3-319-62431-0_2

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-62431-0_2

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-62430-3

  • Online ISBN: 978-3-319-62431-0

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics