Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The immune contexture in cancer prognosis and treatment

Key Points

  • Most cancers have evaded immune control, or immunosurveillance, at the time of presentation; however, residual signs of an active anticancer immune response indicate a positive prognosis

  • Positive immune-related prognostic features include the presence of specific T-lymphocyte subsets, the absence of immunosuppressive elements, the localization of the immune infiltrate and specific features of its organization

  • Successful anticancer therapies, including cytotoxic chemotherapies and targeted agents, improve the local immune contexture and mediate at least part of their long-term effects by reinstating immunosurveillance

  • The presence of either a pre-existing or induced immune response indicates a more favourable prognosis than that of patients whose tumours lack either of these features

  • Immune-checkpoint inhibitors have a profound effect on the local immune infiltrate, and a variety of biomarkers have the potential to indicate a pre-existing or developing anticancer immune response

  • The discovery of immunological biomarkers in oncology has been facilitated by the advent of ever more sophisticated technologies, posing new challenges to both data integration and bioinformatic analysis

Abstract

Immunotherapy is currently the most rapidly advancing area of clinical oncology, and provides the unprecedented opportunity to effectively treat, and even cure, several previously untreatable malignancies. A growing awareness exists of the fact that the success of chemotherapy and radiotherapy, in which the patient's disease can be stabilized well beyond discontinuation of treatment (and occasionally is cured), also relies on the induction of a durable anticancer immune response. Indeed, the local immune infiltrate undergoes dynamic changes that accompany a shift from a pre-existing immune response to a therapy-induced immune response. As a result, the immune contexture, which is determined by the density, composition, functional state and organization of the leukocyte infiltrate of the tumour, can yield information that is relevant to prognosis, prediction of a treatment response and various other pharmacodynamic parameters. Several complementary technologies can be used to explore the immune contexture of tumours, and to derive biomarkers that could enable the adaptation of individual treatment approaches for each patient, as well as monitoring a response to anticancer therapies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Key elements of cancer-related inflammation.
Figure 2: The association between CD8+ T-cell density of the tumour infiltrate and overall survival among patients with primary, or metastatic solid tumours.
Figure 3: Effects of the immune infiltrate on the prognosis of patients with cancer.

Similar content being viewed by others

References

  1. Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205–214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Palucka, A. K. & Coussens, L. M. The basis of oncoimmunology. Cell 164, 1233–1247 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Peng, D. et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527, 249–253 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Slaney, C. Y., Rautela, J. & Parker, B. S. The emerging role of immunosurveillance in dictating metastatic spread in breast cancer. Cancer Res. 73, 5852–5857 (2013).

    CAS  PubMed  Google Scholar 

  6. Elliott, M. R. & Ravichandran, K. S. The dynamics of apoptotic cell clearance. Dev. Cell 38, 147–160 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Coussens, L. M., Zitvogel, L. & Palucka, A. K. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science 339, 286–291 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhong, Z., Sanchez-Lopez, E. & Karin, M. Autophagy, inflammation, and immunity: a troika governing cancer and its treatment. Cell 166, 288–298 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Josephs, D. H., Bax, H. J. & Karagiannis, S. N. Tumour-associated macrophage polarisation and re-education with immunotherapy. Front. Biosci. (Elite Ed.) 7, 293–308 (2015).

    Google Scholar 

  10. Ruffell, B. & Coussens, L. M. Macrophages and therapeutic resistance in cancer. Cancer Cell 27, 462–472 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Nauts, H. C., Swift, W. E. & Coley, B. L. The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, M. D., reviewed in the light of modern research. Cancer Res. 6, 205–216 (1946).

    CAS  PubMed  Google Scholar 

  12. Kamat, A. M. et al. Bladder cancer. Lancet 388, 2796–2810 (2016).

    PubMed  Google Scholar 

  13. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).

    CAS  PubMed  Google Scholar 

  14. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    PubMed  PubMed Central  Google Scholar 

  15. Mlecnik, B. et al. Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 44, 698–711 (2016).

    CAS  PubMed  Google Scholar 

  16. Dieu-Nosjean, M. C. et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J. Clin. Oncol. 26, 4410–4417 (2008).

    CAS  PubMed  Google Scholar 

  17. Dieu-Nosjean, M. C. et al. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol. Rev. 271, 260–275 (2016).

    CAS  PubMed  Google Scholar 

  18. Becht, E. et al. Immune contexture, immunoscore, and malignant cell molecular subgroups for prognostic and theranostic classifications of cancers. Adv. Immunol. 130, 95–190 (2016).

    CAS  PubMed  Google Scholar 

  19. Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 638–652 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Galon, J. et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313, 1960–1964 (2006).

    CAS  PubMed  Google Scholar 

  21. Fridman, W. H. et al. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res. 71, 5601–5605 (2011).

    CAS  PubMed  Google Scholar 

  22. Fridman, W. H., Pages, F., Sautes-Fridman, C. & Galon, J. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer 12, 298–306 (2012).

    CAS  PubMed  Google Scholar 

  23. Zhang, L. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 348, 203–213 (2003).

    CAS  PubMed  Google Scholar 

  24. Bindea, G. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013).

    CAS  PubMed  Google Scholar 

  25. Kirilovsky, A. et al. Rational bases for the use of the Immunoscore in routine clinical settings as a prognostic and predictive biomarker in cancer patients. Int. Immunol. 28, 373–382 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Giraldo, N. A. et al. Tumor-Infiltrating and peripheral blood T-cell immunophenotypes predict early relapse in localized clear cell renal cell carcinoma. Clin. Cancer Res. http://dx.doi.org/10.1158/1078-0432.CCR-16-2848 (2017).

  27. Becht, E. et al. Prognostic and theranostic impact of molecular subtypes and immune classifications in renal cell cancer (RCC) and colorectal cancer (CRC). Oncoimmunology 4, e1049804 (2015).

    PubMed  PubMed Central  Google Scholar 

  28. Petitprez, F. et al. PD-L1 Expression and CD8+ T cell infiltrate are associated with clinical progression in patients with node positive prostate cancer. Eur. Urol. Focus http://dx.doi.org/10.1016/j.euf.2017.05.013 (2017).

  29. Muris, J. J. et al. Prognostic significance of activated cytotoxic T-lymphocytes in primary nodal diffuse large B-cell lymphomas. Leukemia 18, 589–596 (2004).

    CAS  PubMed  Google Scholar 

  30. Scott, D. W. et al. Gene expression-based model using formalin-fixed paraffin-embedded biopsies predicts overall survival in advanced-stage classical Hodgkin lymphoma. J. Clin. Oncol. 31, 692–700 (2013).

    PubMed  Google Scholar 

  31. Giraldo, N. A. et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin. Cancer Res. 21, 3031–3040 (2015).

    CAS  PubMed  Google Scholar 

  32. Meng, Y., Harlin, H., O'Keefe, J. P. & Gajewski, T. F. Induction of cytotoxic granules in human memory CD8+ T cell subsets requires cell cycle progression. J. Immunol. 177, 1981–1987 (2006).

    CAS  PubMed  Google Scholar 

  33. Bronte, V. et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 7, 12150 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Di Caro, G. et al. Tertiary lymphoid tissue in the tumor microenvironment: from its occurrence to immunotherapeutic implications. Int. Rev. Immunol. 34, 123–133 (2015).

    CAS  PubMed  Google Scholar 

  35. Sautes-Fridman, C. et al. Tertiary lymphoid structures in cancers: prognostic value, regulation, and manipulation for therapeutic intervention. Front. Immunol. 7, 407 (2016).

    PubMed  PubMed Central  Google Scholar 

  36. Goc, J. et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 74, 705–715 (2014).

    CAS  PubMed  Google Scholar 

  37. Di Caro, G. et al. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin. Cancer Res. 20, 2147–2158 (2014).

    CAS  PubMed  Google Scholar 

  38. Kroeger, D. R., Milne, K. & Nelson, B. H. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin. Cancer Res. 22, 3005–3015 (2016).

    CAS  PubMed  Google Scholar 

  39. Finkin, S. et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 16, 1235–1244 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Shalapour, S. et al. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature 521, 94–98 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Sautes-Fridman, C. & Fridman, W. H. TLS in tumors: what lies within. Trends Immunol. 37, 1–2 (2016).

    CAS  PubMed  Google Scholar 

  42. Xu, B. et al. Prognostic value of tumor infiltrating NK cells and macrophages in stage II+III esophageal cancer patients. Oncotarget 7, 74904–74916 (2016).

    PubMed  PubMed Central  Google Scholar 

  43. Thommen, D. S. et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol. Res. 3, 1344–1355 (2015).

    CAS  PubMed  Google Scholar 

  44. Halama, N. et al. Localization and density of immune cells in the invasive margin of human colorectal cancer liver metastases are prognostic for response to chemotherapy. Cancer Res. 71, 5670–5677 (2011).

    CAS  PubMed  Google Scholar 

  45. Remark, R. 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. 19, 4079–4091 (2013).

    CAS  PubMed  Google Scholar 

  46. Naxerova, K. & Jain, R. K. Using tumour phylogenetics to identify the roots of metastasis in humans. Nat. Rev. Clin. Oncol. 12, 258–272 (2015).

    CAS  PubMed  Google Scholar 

  47. Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).

    CAS  PubMed  Google Scholar 

  48. Bruggen, M. C. et al. In situ mapping of innate lymphoid cells in human skin: evidence for remarkable differences between normal and inflamed skin. J. Invest. Dermatol. 136, 2396–2405 (2016).

    PubMed  Google Scholar 

  49. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Becht, E. et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17, 218 (2016).

    PubMed  PubMed Central  Google Scholar 

  51. Stoll, G. et al. Immune-related gene signatures predict the outcome of neoadjuvant chemotherapy. Oncoimmunology 3, e27884 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Stoll, G. et al. Meta-analysis of organ-specific differences in the structure of the immune infiltrate in major malignancies. Oncotarget 6, 11894–11909 (2015).

    PubMed  PubMed Central  Google Scholar 

  53. Kroemer, G., Senovilla, L., Galluzzi, L., Andre, F. & Zitvogel, L. Natural and therapy-induced immunosurveillance in breast cancer. Nat. Med. 21, 1128–1138 (2015).

    CAS  PubMed  Google Scholar 

  54. Ali, H. R., Chlon, L., Pharoah, P. D., Markowetz, F. & Caldas, C. Patterns of immune infiltration in breast cancer and their clinical implications: a gene-expression-based retrospective study. PLoS Med. 13, e1002194 (2016).

    PubMed  PubMed Central  Google Scholar 

  55. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).

    CAS  PubMed  Google Scholar 

  57. Spranger, S. & Gajewski, T. F. Tumor-intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology 5, e1086862 (2016).

    PubMed  Google Scholar 

  58. Corrales, L., McWhirter, S. M., Dubensky, T. W. Jr. & Gajewski, T. F. The host STING pathway at the interface of cancer and immunity. J. Clin. Invest. 126, 2404–2411 (2016).

    PubMed  PubMed Central  Google Scholar 

  59. Wang, T. et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat. Med. 10, 48–54 (2004).

    PubMed  Google Scholar 

  60. Salerno, E. P. et al. Human melanomas and ovarian cancers overexpressing mechanical barrier molecule genes lack immune signatures and have increased patient mortality risk. Oncoimmunology 5, e1240857 (2016).

    PubMed  PubMed Central  Google Scholar 

  61. Guinney, J. et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350–1356 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Dudley, J. C., Lin, M. T., Le, D. T. & Eshleman, J. R. Microsatellite instability as a biomarker for PD-1 blockade. Clin. Cancer Res. 22, 813–820 (2016).

    CAS  PubMed  Google Scholar 

  63. Becht, E. et al. Immune and stromal classification of colorectal cancer is associated with molecular subtypes and relevant for precision immunotherapy. Clin. Cancer Res. 22, 4057–4066 (2016).

    CAS  PubMed  Google Scholar 

  64. Beuselinck, B. et al. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin. Cancer Res. 21, 1329–1339 (2015).

    CAS  PubMed  Google Scholar 

  65. Cirenajwis, H. et al. Molecular stratification of metastatic melanoma using gene expression profiling: prediction of survival outcome and benefit from molecular targeted therapy. Oncotarget 6, 12297–12309 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Burstein, M. D. et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin. Cancer Res. 21, 1688–1698 (2015).

    CAS  PubMed  Google Scholar 

  67. Lehmann, B. D. et al. Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS ONE 11, e0157368 (2016).

    PubMed  PubMed Central  Google Scholar 

  68. Bonsang-Kitzis, H. et al. Biological network-driven gene selection identifies a stromal immune module as a key determinant of triple-negative breast carcinoma prognosis. Oncoimmunology 5, e1061176 (2016).

    CAS  PubMed  Google Scholar 

  69. Bense, R. D. et al. Relevance of tumor-infiltrating immune cell composition and functionality for disease outcome in breast cancer. J. Natl Cancer Inst. 109, djw192 (2017).

    Google Scholar 

  70. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Zitvogel, L., Kepp, O. & Kroemer, G. Decoding cell death signals in inflammation and immunity. Cell 140, 798–804 (2010).

    CAS  PubMed  Google Scholar 

  72. Brea, E. J. et al. Kinase regulation of human MHC class I molecule expression on cancer cells. Cancer Immunol. Res. 4, 936–947 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Reits, E. A. et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 203, 1259–1271 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Krysko, D. V. et al. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 12, 860–875 (2012).

    CAS  PubMed  Google Scholar 

  76. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 (2013).

    CAS  PubMed  Google Scholar 

  77. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    CAS  PubMed  Google Scholar 

  78. Michaud, M. et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 334, 1573–1577 (2011).

    CAS  PubMed  Google Scholar 

  79. Martins, I. et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 21, 79–91 (2014).

    CAS  PubMed  Google Scholar 

  80. Obeid, M. et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 13, 54–61 (2007).

    CAS  PubMed  Google Scholar 

  81. Apetoh, L. et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13, 1050–1059 (2007).

    CAS  PubMed  Google Scholar 

  82. Sistigu, A. et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 20, 1301–1309 (2014).

    CAS  PubMed  Google Scholar 

  83. Vacchelli, E. et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science 350, 972–978 (2015).

    CAS  PubMed  Google Scholar 

  84. Ruffell, B. et al. Leukocyte composition of human breast cancer. Proc. Natl Acad. Sci. USA 109, 2796–2801 (2012).

    CAS  PubMed  Google Scholar 

  85. Senovilla, L. et al. An immunosurveillance mechanism controls cancer cell ploidy. Science 337, 1678–1684 (2012).

    CAS  PubMed  Google Scholar 

  86. Park, J. H. et al. Clonal expansion of antitumor T cells in breast cancer correlates with response to neoadjuvant chemotherapy. Int. J. Oncol. 49, 471–478 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Nardone, V. et al. Tumor infiltrating T lymphocytes expressing FoxP3, CCR7 or PD-1 predict the outcome of prostate cancer patients subjected to salvage radiotherapy after biochemical relapse. Cancer Biol. Ther. 17, 1213–1220 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Tavare, R. et al. An effective immuno-PET imaging method to monitor CD8-dependent responses to immunotherapy. Cancer Res. 76, 73–82 (2016).

    CAS  PubMed  Google Scholar 

  89. Ladoire, S. et al. The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy 12, 864–875 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Ladoire, S. et al. Combined evaluation of LC3B puncta and HMGB1 expression predicts residual risk of relapse after adjuvant chemotherapy in breast cancer. Autophagy 11, 1878–1890 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Allard, B., Beavis, P. A., Darcy, P. K. & Stagg, J. Immunosuppressive activities of adenosine in cancer. Curr. Opin. Pharmacol. 29, 7–16 (2016).

    CAS  PubMed  Google Scholar 

  92. Wemeau, M. et al. Calreticulin exposure on malignant blasts predicts a cellular anticancer immune response in patients with acute myeloid leukemia. Cell Death Dis. 1, e104 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Fucikova, J. et al. Calreticulin expression in human non-small cell lung cancers correlates with increased accumulation of antitumor immune cells and favorable prognosis. Cancer Res. 76, 1746–1756 (2016).

    CAS  PubMed  Google Scholar 

  94. Fucikova, J. et al. Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients. Blood 128, 3113–3124 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Stoll, G. et al. Calreticulin expression: interaction with the immune infiltrate and impact on survival in patients with ovarian and non-small cell lung cancer. Oncoimmunology 5, e1177692 (2016).

    PubMed  PubMed Central  Google Scholar 

  96. Tesniere, A. et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 29, 482–491 (2010).

    CAS  PubMed  Google Scholar 

  97. Vacchelli, E., Enot, D. P., Pietrocola, F., Zitvogel, L. & Kroemer, G. Impact of pattern recognition receptors on the prognosis of breast cancer patients undergoing adjuvant chemotherapy. Cancer Res. 76, 3122–3126 (2016).

    CAS  PubMed  Google Scholar 

  98. Yang, H. et al. Contribution of RIP3 and MLKL to immunogenic cell death signaling in cancer chemotherapy. Oncoimmunology 5, e1149673 (2016).

    PubMed  PubMed Central  Google Scholar 

  99. Stoll, G. et al. Pro-necrotic molecules impact local immunosurveillance in human breast cancer. Oncoimmunology 6, e1299302 (2017).

    PubMed  PubMed Central  Google Scholar 

  100. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 28, 690–714 (2015).

    CAS  PubMed  Google Scholar 

  101. Vincent, J. et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 70, 3052–3061 (2010).

    CAS  PubMed  Google Scholar 

  102. Ghiringhelli, F. et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 56, 641–648 (2007).

    CAS  PubMed  Google Scholar 

  103. Viaud, S. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 342, 971–976 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Wang, W. et al. Effector T cells abrogate stroma-mediated chemoresistance in ovarian cancer. Cell 165, 1092–1105 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Galluzzi, L., Senovilla, L., Zitvogel, L. & Kroemer, G. The secret ally: immunostimulation by anticancer drugs. Nat. Rev. Drug Discov. 11, 215–233 (2012).

    CAS  PubMed  Google Scholar 

  106. Zitvogel, L., Rusakiewicz, S., Routy, B., Ayyoub, M. & Kroemer, G. Immunological off-target effects of imatinib. Nat. Rev. Clin. Oncol. 13, 431–446 (2016).

    CAS  PubMed  Google Scholar 

  107. Rusakiewicz, S. et al. Immune infiltrates are prognostic factors in localized gastrointestinal stromal tumors. Cancer Res. 73, 3499–3510 (2013).

    CAS  PubMed  Google Scholar 

  108. Delahaye, N. F. et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat. Med. 17, 700–707 (2011).

    CAS  PubMed  Google Scholar 

  109. Mizoguchi, I. et al. Sustained upregulation of effector natural killer cells in chronic myeloid leukemia after discontinuation of imatinib. Cancer Sci. 104, 1146–1153 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Riva, G. et al. Long-term molecular remission with persistence of BCR–ABL1-specific cytotoxic T cells following imatinib withdrawal in an elderly patient with Philadelphia-positive ALL. Br. J. Haematol. 164, 299–302 (2014).

    PubMed  Google Scholar 

  111. Hugo, W. et al. Non-genomic and immune evolution of melanoma acquiring MAPKi resistance. Cell 162, 1271–1285 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Frederick, D. T. et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin. Cancer Res. 19, 1225–1231 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Knight, D. A. et al. Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J. Clin. Invest. 123, 1371–1381 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Pozzi, C. et al. The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat. Med. 22, 624–631 (2016).

    CAS  PubMed  Google Scholar 

  116. Muller, P. et al. Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci. Transl Med. 7, 315ra188 (2015).

    PubMed  Google Scholar 

  117. Guislain, A. et al. Sunitinib pretreatment improves tumor-infiltrating lymphocyte expansion by reduction in intratumoral content of myeloid-derived suppressor cells in human renal cell carcinoma. Cancer Immunol. Immunother. 64, 1241–1250 (2015).

    CAS  PubMed  Google Scholar 

  118. Sliwkowski, M. X. & Mellman, I. Antibody therapeutics in cancer. Science 341, 1192–1198 (2013).

    CAS  PubMed  Google Scholar 

  119. Gibney, G. T., Weiner, L. M. & Atkins, M. B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 17, e542–e551 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Pitt, J. M. et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity 44, 1255–1269 (2016).

    CAS  PubMed  Google Scholar 

  121. Weide, B. et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin. Cancer Res. 22, 5487–5496 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Brand, A. et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK Cells. Cell Metab. 24, 657–671 (2016).

    CAS  PubMed  Google Scholar 

  123. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Huang, A. C. et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Bald, T. et al. Immune cell-poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Discov. 4, 674–687 (2014).

    CAS  PubMed  Google Scholar 

  126. Spranger, S. et al. Up-regulation of PD-L1, IDO, and Tregs in the melanoma tumor microenvironment is driven by CD8+ T cells. Sci. Transl Med. 5, 200ra116 (2013).

    PubMed  PubMed Central  Google Scholar 

  127. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. van Rooij, N. et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 31, e439–442 (2013).

    PubMed  Google Scholar 

  129. Rizvi, N. A. et al. Cancer immunology. mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Rosenberg, J. E. et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387, 1909–1920 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Ansell, S. M. et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N. Engl. J. Med. 372, 311–319 (2015).

    PubMed  Google Scholar 

  133. Vitale, I. et al. Illicit survival of cancer cells during polyploidization and depolyploidization. Cell Death Differ. 18, 1403–1413 (2011).

    CAS  PubMed  Google Scholar 

  134. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    PubMed  PubMed Central  Google Scholar 

  135. Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl Med. 9, eaah3560 (2017).

    PubMed  PubMed Central  Google Scholar 

  136. Li, B. et al. Landscape of tumor-infiltrating T cell repertoire of human cancers. Nat. Genet. 48, 725–732 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Inoue, H. et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. Oncoimmunology 5, e1204507 (2016).

    PubMed  PubMed Central  Google Scholar 

  138. O'Day, S. J. et al. Phase II multicenter trial of maintenance biotherapy after induction concurrent biochemotherapy for patients with metastatic melanoma. J. Clin. Oncol. 27, 6207–6212 (2009).

    PubMed  Google Scholar 

  139. Oh, D. Y. et al. Immune toxicities elicted by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res. 77, 1322–1330 (2017).

    CAS  PubMed  Google Scholar 

  140. Gros, A. et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 22, 433–438 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Paulsen, E. E. et al. Assessing PDL-1 and PD-1 in non-small cell lung cancer: a novel immunoscore approach. Clin. Lung Cancer 18, 220–233 e8 (2017).

    CAS  PubMed  Google Scholar 

  142. Darb-Esfahani, S. et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget 7, 1486–1499 (2016).

    PubMed  Google Scholar 

  143. Duchnowska, R. et al. Immune response in breast cancer brain metastases and their microenvironment: the role of the PD-1/PD-L axis. Breast Cancer Res. 18, 43 (2016).

    PubMed  PubMed Central  Google Scholar 

  144. Cimino-Mathews, A. et al. PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum. Pathol. 47, 52–63 (2016).

    CAS  PubMed  Google Scholar 

  145. Denkert, C. et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J. Clin. Oncol. 33, 983–991 (2015).

    CAS  PubMed  Google Scholar 

  146. Dunne, P. D. et al. Immune-derived PD-L1 gene expression defines a subgroup of stage II/III colorectal cancer patients with favorable prognosis who may be harmed by adjuvant chemotherapy. Cancer Immunol. Res. 4, 582–591 (2016).

    CAS  PubMed  Google Scholar 

  147. Taube, J. M. et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci. Transl Med. 4, 127ra37 (2012).

    PubMed  PubMed Central  Google Scholar 

  148. Blank, C. U., Haanen, J. B., Ribas, A. & Schumacher, T. N. CANCER IMMUNOLOGY. The “cancer immunogram”. Science 352, 658–660 (2016).

    CAS  PubMed  Google Scholar 

  149. Ock, C. Y. et al. Pan-cancer immunogenomic perspective on the tumor microenvironment based on PD-L1 and CD8 T-cell infiltration. Clin. Cancer Res. 22, 2261–2270 (2016).

    CAS  PubMed  Google Scholar 

  150. Koyama, S. et al. STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res. 76, 999–1008 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Shin, D. S. et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 7, 188–201 (2017).

    CAS  PubMed  Google Scholar 

  153. Benci, J. L. et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 167, 1540–1554.e12 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Koyama, S. et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 7, 10501 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Orvedahl, A. et al. HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe 1, 23–35 (2007).

    CAS  PubMed  Google Scholar 

  156. Li, Y. et al. ICP34.5 protein of herpes simplex virus facilitates the initiation of protein translation by bridging eukaryotic initiation factor 2α (eIF2α) and protein phosphatase 1. J. Biol. Chem. 286, 24785–24792 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Puzanov, I. et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J. Clin. Oncol. 34, 2619–2626 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Natarajan, A. et al. Novel radiotracer for immunoPET imaging of PD-1 checkpoint expression on tumor infiltrating lymphocytes. Bioconjug. Chem. 26, 2062–2069 (2015).

    CAS  PubMed  Google Scholar 

  159. Lavin, Y. et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765.e17 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Dadi, S. et al. Cancer immunosurveillance by tissue-resident innate lymphoid cells and innate-like T cells. Cell 164, 365–377 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Fan, X. & Rudensky, A. Y. Hallmarks of tissue-resident lymphocytes. Cell 164, 1198–1211 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Zitvogel, L., Pitt, J. M., Daillere, R., Smyth, M. J. & Kroemer, G. Mouse models in oncoimmunology. Nat. Rev. Cancer 16, 759–773 (2016).

    CAS  PubMed  Google Scholar 

  165. Moschella, F. et al. Cyclophosphamide induces a type I interferon-associated sterile inflammatory response signature in cancer patients' blood cells: implications for cancer chemoimmunotherapy. Clin. Cancer Res. 19, 4249–4261 (2013).

    CAS  PubMed  Google Scholar 

  166. Lo, C. S. et al. Neoadjuvant chemotherapy of ovarian cancer results in three patterns of tumor-infiltrating lymphocyte response with distinct implications for immunotherapy. Clin. Cancer Res. 23, 925–934 (2017).

    CAS  PubMed  Google Scholar 

  167. Limagne, E. et al. Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX–bevacizumab drug treatment regimen. Cancer Res. 76, 5241–5252 (2016).

    CAS  PubMed  Google Scholar 

  168. Teng, F. et al. Tumor-infiltrating lymphocytes, forkhead box P3, programmed death ligand-1, and cytotoxic T lymphocyte-associated antigen-4 expressions before and after neoadjuvant chemoradiation in rectal cancer. Transl Res. 166, 721–732.e1 (2015).

    CAS  PubMed  Google Scholar 

  169. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Mahoney, K. M. & Atkins, M. B. Prognostic and predictive markers for the new immunotherapies. Oncol. (Williston Park) 28 (Suppl. 3), 39–48 (2014).

    Google Scholar 

  171. Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Garon, E. B. et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372, 2018–2028 (2015).

    PubMed  Google Scholar 

  173. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    PubMed  PubMed Central  Google Scholar 

  174. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Johnson, D. B. et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat. Commun. 7, 10582 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Chiappinelli, K. B. et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 162, 974–986 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Cha, E. et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl Med. 6, 238ra70 (2014).

    PubMed  PubMed Central  Google Scholar 

  178. Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).

    CAS  PubMed  Google Scholar 

  179. Hamid, O. et al. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J. Transl Med. 9, 204 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Chen, P. L. 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. 6, 827–837 (2016).

    PubMed  PubMed Central  Google Scholar 

  181. Martens, A. et al. Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin. Cancer Res. 22, 2908–2918 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Simeone, E. et al. Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol. Immunother. 63, 675–683 (2014).

    CAS  PubMed  Google Scholar 

  183. Weber, J. S. et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J. Clin. Oncol. 31, 4311–4318 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Jacquelot, N. et al. Chemokine receptor patterns in lymphocytes mirror metastatic spreading in melanoma. J. Clin. Invest. 126, 921–937 (2016).

    PubMed  PubMed Central  Google Scholar 

  185. Hannani, D. et al. Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Res. 25, 208–224 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Ghiringhelli, F. et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β-dependent adaptive immunity against tumors. Nat. Med. 15, 1170–1178 (2009).

    CAS  PubMed  Google Scholar 

  187. Loi, S. et al. CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc. Natl Acad. Sci. USA 110, 11091–11096 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Ren, Z. H. et al. CD73 is associated with poor prognosis in HNSCC. Oncotarget 7, 61690–61702 (2016).

    PubMed  PubMed Central  Google Scholar 

  189. Turcotte, M. et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 75, 4494–4503 (2015).

    CAS  PubMed  Google Scholar 

  190. Wang, X., Du, Z., Li, L., Shi, M. & Yu, Y. Beclin 1 and p62 expression in non-small cell lung cancer: relation with malignant behaviors and clinical outcome. Int. J. Clin. Exp. Pathol. 8, 10644–10652 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Schlafli, A. M. et al. Prognostic value of the autophagy markers LC3 and p62/SQSTM1 in early-stage non-small cell lung cancer. Oncotarget 7, 39544–39555 (2016).

    PubMed  PubMed Central  Google Scholar 

  192. Baccelli, I. et al. Co-expression of MET and CD47 is a novel prognosticator for survival of luminal breast cancer patients. Oncotarget 5, 8147–8160 (2014).

    PubMed  PubMed Central  Google Scholar 

  193. Yoshida, K. et al. CD47 is an adverse prognostic factor and a therapeutic target in gastric cancer. Cancer Med. 4, 1322–1333 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Willingham, S. B. et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl Acad. Sci. USA 109, 6662–6667 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Chao, M. P. et al. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 71, 1374–1384 (2011).

    CAS  PubMed  Google Scholar 

  196. Bidwell, B. N. et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).

    CAS  PubMed  Google Scholar 

  197. Yamazaki, T. et al. Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 21, 69–78 (2014).

    CAS  PubMed  Google Scholar 

  198. Exner, R. et al. Prognostic value of HMGB1 in early breast cancer patients under neoadjuvant chemotherapy. Cancer Med. 5, 2350–2358 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank N. Giraldo and F. Petitprez for their help in finalizing the manuscript and the figures. The work of W.H.F. and C.S.F. is supported by the Institut National de la santé et de la Recherche Medicale (INSERM); University Paris-Descartes; University Pierre and Marie Curie; the Site de Recherche Integrée sur le Cancer (SIRIC) Cancer Research for Personalized Medicine (CARPEM) programme; the LabEx Immuno-Oncology; the Institut National Du Cancer (INCa); the Cancéropôle Ile-de-France; O. Lecomte; and the Association pour la recherche sur le cancer (ARC). The work of L.Z. and G.K. is supported by the INCa, the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; ARC; Cancéropôle Ile-de-France; INSERM (HTE); INCa; Institut Universitaire de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumour Immune Elimination (SOCRATE); CARPEM; and the Paris Alliance of Cancer Research Institutes (PACRI). The work of L.Z. is also supported by the Swiss Institute for Experimental Cancer Research (ISREC), by the Swiss Bridge Foundation, and by IMMUNTRAIN-H2020.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to researching the data for the article and to discussion of the content. All authors reviewed/edited the manuscript before submission.

Corresponding authors

Correspondence to Wolf H. Fridman or Guido Kroemer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

Influence of the density of tumour-infiltrating immune cell types on the prognosis of patients with cancer (Figs 2 and 3) (PDF 263 kb)

PowerPoint slides

Glossary

Tumour microenvironment

The area immediately surrounding the tumour that is typically composed of nonmalignant lymphoid and/or myeloid cells as well as fibroblasts, vascular cells and lymphatic vessels. Specific characteristics of the tumour microenvironment can have either positive or negative implications for patient outcomes.

M1 macrophages

Macrophages that produce predominantly proinflammatory factors. M1 macrophages and are generally associated with a good prognosis when present at high densities in the tumour microenvironment.

Erysipelas

An acute bacterial infection, usually by Streptococcus pyogenes, characterized by the appearance of large, raised red patches on the skin.

M2 macrophages

Macrophages producing proangiogenic and immunosuppressive factors. M2 macrophages are generally associated with a poor prognosis when present at high densities in the tumour microenvironment.

Type 1 T helper cells

(TH1 cells). A subset of CD4+ T-helper cells that produce IFNγ and IL-2.

Tc1 cells

A subset of CD8+ cytotoxic T cells that produce IFNγ.

Immune contexture

A term describing the integration of knowledge of the density, functional orientation and spatial organization of the immune infiltrate.

Natural killer cells

(NK cells). Lymphocytes devoid of antigen receptors that are capable of cell-mediated cytotoxicity, including the destruction of tumour cells lacking MHC class I molecules.

Myeloid-derived suppressor cells

(MDSCs). A heterogeneous population composed of cells of a granulocytic or macrophagic origin that produce immunosuppressive factors. MDSCs are generally associated with a poor prognosis when present at high densities in the tumour microenvironment.

Regulatory T cells

(Treg cells). Regulatory T cells, which can be identified based on cell-surface antigen expression (CD4+/CD25+/FOXP3+). Such cells are generally immunosuppressive and associated with a poor prognosis.

CIBERSORT

(Cell-type identification by estimating relative subsets of RNA transcripts). A metagene-based analytical method of weighting the contribution of different leukocyte subpopulations to the overall immune infiltrate during the analysis of transcriptomic data by measuring the expression of genes associated with specific immune cell types relative to those expressed in all haematological cell types.

Microenvironment cell populations–counter

(MCP-Counter). A method based on metagenes highly expressed in one and only one cellular population present in the tumour microenvironment, which enables intersample quantification of infiltrating cells based on transcriptomic data.

Immunome

A method that curates metagenes preferentially expressed in immune cells, thus enabling the quantification of these cells based on their transcriptome.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fridman, W., Zitvogel, L., Sautès–Fridman, C. et al. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14, 717–734 (2017). https://doi.org/10.1038/nrclinonc.2017.101

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrclinonc.2017.101

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer