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:

Neutrophil diversity and plasticity in tumour progression and therapy

Abstract

Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.

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

Access options

Buy this article

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

Fig. 1: Neutrophils in tumour promotion.
Fig. 2: Antitumour potential of neutrophils.
Fig. 3: Therapeutic targeting of neutrophils.

Similar content being viewed by others

References

  1. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 13, 159–175 (2013).

    CAS  PubMed  Google Scholar 

  2. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 33, 657–670 (2010).

    CAS  PubMed  Google Scholar 

  3. Mantovani, A., Cassatella, M. A., Costantini, C. & Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 11, 519–531 (2011).

    CAS  PubMed  Google Scholar 

  4. Jaillon, S. et al. Neutrophils in innate and adaptive immunity. Semin. Immunopathol. 35, 377–394 (2013).

    CAS  PubMed  Google Scholar 

  5. Scapini, P. & Cassatella, M. A. Social networking of human neutrophils within the immune system. Blood 124, 710–719 (2014).

    CAS  PubMed  Google Scholar 

  6. Fridman, W. H., Zitvogel, L., Sautes-Fridman, C. & Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 14, 717–734 (2017).

    CAS  PubMed  Google Scholar 

  7. Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. Cancer-related inflammation. Nature 454, 436–444 (2008).

    CAS  PubMed  Google Scholar 

  8. Bonavita, E. et al. PTX3 is an extrinsic oncosuppressor regulating complement-dependent inflammation in cancer. Cell 160, 700–714 (2015).

    CAS  PubMed  Google Scholar 

  9. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Coffelt, S. B., Wellenstein, M. D. & de Visser, K. E. Neutrophils in cancer: neutral no more. Nat. Rev. Cancer 16, 431–446 (2016).

    CAS  PubMed  Google Scholar 

  12. Shaul, M. E. & Fridlender, Z. G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 16, 601–620 (2019).

    PubMed  Google Scholar 

  13. Galdiero, M. R. et al. Occurrence and significance of tumor-associated neutrophils in patients with colorectal cancer. Int. J. Cancer 139, 446–456 (2016).

    CAS  PubMed  Google Scholar 

  14. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015). This paper shows that a neutrophil signature is associated with adverse prognosis in most solid tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Ponzetta, A. et al. Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell 178, 346–360.e24 (2019). This study demonstrates a tripartite interaction between neutrophils, macrophages and a subset of T cells, UTCαβ, essential for the establishment of effective antitumour immunity in select tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Kargl, J. et al. Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat. Commun. 8, 14381 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    CAS  PubMed  Google Scholar 

  18. Ng, L. G., Ostuni, R. & Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 19, 255–265 (2019).

    CAS  PubMed  Google Scholar 

  19. Nemeth, T., Sperandio, M. & Mocsai, A. Neutrophils as emerging therapeutic targets. Nat. Rev. Drug Discov. 19, 253–275 (2020).

    CAS  PubMed  Google Scholar 

  20. Eruslanov, E. B., Singhal, S. & Albelda, S. M. Mouse versus human neutrophils in cancer: a major knowledge gap. Trends Cancer 3, 149–160 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Mestas, J. & Hughes, C. C. Of mice and not men: differences between mouse and human immunology. J. Immunol. 172, 2731–2738 (2004).

    CAS  PubMed  Google Scholar 

  22. Lawrence, S. M., Corriden, R. & Nizet, V. The ontogeny of a neutrophil: mechanisms of granulopoiesis and homeostasis. Microbiol. Mol. Biol. Rev. 82, e00057-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Skokowa, J. et al. Interactions among HCLS1, HAX1 and LEF-1 proteins are essential for G-CSF-triggered granulopoiesis. Nat. Med. 18, 1550–1559 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Zeidler, C., Germeshausen, M., Klein, C. & Welte, K. Clinical implications of ELA2-, HAX1-, and G-CSF-receptor (CSF3R) mutations in severe congenital neutropenia. Br. J. Haematol. 144, 459–467 (2009).

    CAS  PubMed  Google Scholar 

  25. Liu, F., Wu, H. Y., Wesselschmidt, R., Kornaga, T. & Link, D. C. Impaired production and increased apoptosis of neutrophils in granulocyte colony-stimulating factor receptor-deficient mice. Immunity 5, 491–501 (1996).

    CAS  PubMed  Google Scholar 

  26. Lieschke, G. J. et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84, 1737–1746 (1994).

    CAS  PubMed  Google Scholar 

  27. Romani, L. et al. Impaired neutrophil response and CD4+ T helper cell 1 development in interleukin 6-deficient mice infected with Candida albicans. J. Exp. Med. 183, 1345–1355 (1996).

    CAS  PubMed  Google Scholar 

  28. Walker, F. et al. IL6/sIL6R complex contributes to emergency granulopoietic responses in G-CSF- and GM-CSF-deficient mice. Blood 111, 3978–3985 (2008).

    CAS  PubMed  Google Scholar 

  29. Eash, K. J., Greenbaum, A. M., Gopalan, P. K. & Link, D. C. CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow. J. Clin. Invest. 120, 2423–2431 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Casanova-Acebes, M. et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 153, 1025–1035 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, D. et al. Neutrophil ageing is regulated by the microbiome. Nature 525, 528–532 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Adrover, J. M. et al. A neutrophil timer coordinates immune defense and vascular protection. Immunity 50, 390–402.e10 (2019). Together with reference 31, this paper identifies extrinsic and intrinsic mechanisms involved in the process of neutrophil ageing.

    CAS  PubMed  Google Scholar 

  33. Stark, M. A. et al. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 22, 285–294 (2005).

    CAS  PubMed  Google Scholar 

  34. Casanova-Acebes, M. et al. Neutrophils instruct homeostatic and pathological states in naive tissues. J. Exp. Med. 215, 2778–2795 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Coffelt, S. B. et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 522, 345–348 (2015). This study demonstrates that neutrophils orchestrate systemic inflammation to promote metastasis.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Wellenstein, M. D. et al. Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis. Nature 572, 538–542 (2019). This paper demonstrates that a WNT–IL-1β axis leads to neutrophil expansion and the subsequent formation of breast cancer metastases.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Jin, C. et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell 176, 998–1013.e16 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Gabrilovich, D. I., Ostrand-Rosenberg, S. & Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 12, 253–268 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Mollica Poeta, V., Massara, M., Capucetti, A. & Bonecchi, R. Chemokines and chemokine receptors: new targets for cancer immunotherapy. Front. Immunol. 10, 379 (2019).

    PubMed  PubMed Central  Google Scholar 

  40. Jamieson, T. et al. Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J. Clin. Invest. 122, 3127–3144 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Charles, K. A. et al. The tumor-promoting actions of TNF-α involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J. Clin. Invest. 119, 3011–3023 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Colotta, F., Re, F., Polentarutti, N., Sozzani, S. & Mantovani, A. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood 80, 2012–2020 (1992).

    CAS  PubMed  Google Scholar 

  43. Raccosta, L. et al. The oxysterol–CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J. Exp. Med. 210, 1711–1728 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Roumenina, L. T., Daugan, M. V., Petitprez, F., Sautes-Fridman, C. & Fridman, W. H. Context-dependent roles of complement in cancer. Nat. Rev. Cancer 19, 698–715 (2019).

    CAS  PubMed  Google Scholar 

  45. Reis, E. S., Mastellos, D. C., Ricklin, D., Mantovani, A. & Lambris, J. D. Complement in cancer: untangling an intricate relationship. Nat. Rev. Immunol. 18, 5–18 (2018).

    CAS  PubMed  Google Scholar 

  46. Sody, S. et al. Distinct spatio-temporal dynamics of tumor-associated neutrophils in small tumor lesions. Front. Immunol. 10, 1419 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Patel, S. et al. Unique pattern of neutrophil migration and function during tumor progression. Nat. Immunol. 19, 1236–1247 (2018). This study highlights functional changes that neutrophils undergo during tumour progression and demonstrates that immunosuppressive activity is limited to neutrophils from the late stages of cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Granot, Z. et al. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 20, 300–314 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Massara, M. et al. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat. Commun. 9, 676 (2018).

    PubMed  PubMed Central  Google Scholar 

  50. Wculek, S. K. & Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 528, 413–417 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kowanetz, M. et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl Acad. Sci. USA 107, 21248–21255 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Acharyya, S. et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 150, 165–178 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Wu, F. Y. et al. Chemokine decoy receptor d6 plays a negative role in human breast cancer. Mol. Cancer Res. 6, 1276–1288 (2008).

    CAS  PubMed  Google Scholar 

  54. Adrover, J. M. et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 21, 135–144 (2020). This article demonstrates the homeostatic degranulation and ‘disarming’ of neutrophils driven by regulators of circadian cycles.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Xie, X. et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity and orchestrated maturation during homeostasis and bacterial infection. Preprint at bioRxiv https://doi.org/10.1101/792200v1 (2019).

  56. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334.e10 (2019). This study identifies populations of tumour-infiltrating myeloid cells in human and mouse lung tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Puga, I. et al. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat. Immunol. 13, 170–180 (2011).

    PubMed  PubMed Central  Google Scholar 

  58. Lok, L. S. C. et al. Phenotypically distinct neutrophils patrol uninfected human and mouse lymph nodes. Proc. Natl Acad. Sci. USA 116, 19083–19089 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Locati, M., Curtale, G. & Mantovani, A. Diversity, mechanisms, and significance of macrophage plasticity. Annu. Rev. Pathol. 15, 123–147 (2020).

    CAS  PubMed  Google Scholar 

  60. Alshetaiwi, H. et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci. Immunol. 5, eaay6017 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Casbon, A. J. et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc. Natl Acad. Sci. USA 112, E566–E575 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 19, 108–119 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhu, Y. P. et al. Identification of an early unipotent neutrophil progenitor with pro-tumoral activity in mouse and human bone marrow. Cell Rep. 24, 2329–2341.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Fridlender, Z. G. et al. Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 16, 183–194 (2009). This paper demonstrates the polarization of TANs induced by TGFβ signalling.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Shaul, M. E. et al. Tumor-associated neutrophils display a distinct N1 profile following TGFβ modulation: a transcriptomics analysis of pro- vs. antitumor TANs. Oncoimmunology 5, e1232221 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. Sagiv, J. Y. et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 10, 562–573 (2015).

    CAS  PubMed  Google Scholar 

  67. Andzinski, L. et al. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int. J. Cancer 138, 1982–1993 (2016).

    CAS  PubMed  Google Scholar 

  68. Singhal, S. et al. Origin and role of a subset of tumor-associated neutrophils with antigen-presenting cell features in early-stage human lung cancer. Cancer Cell 30, 120–135 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Eruslanov, E. B. et al. Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J. Clin. Invest. 124, 5466–5480 (2014).

    PubMed  PubMed Central  Google Scholar 

  70. Si, Y. et al. Multidimensional imaging provides evidence for down-regulation of T cell effector function by MDSC in human cancer tissue. Sci. Immunol. 4, eaaw9159 (2019).

    CAS  PubMed  Google Scholar 

  71. Granot, Z. & Fridlender, Z. G. Plasticity beyond cancer cells and the “immunosuppressive switch”. Cancer Res. 75, 4441–4445 (2015).

    CAS  PubMed  Google Scholar 

  72. Butin-Israeli, V. et al. Neutrophil-induced genomic instability impedes resolution of inflammation and wound healing. J. Clin. Invest. 129, 712–726 (2019).

    PubMed  PubMed Central  Google Scholar 

  73. Gungor, N. et al. Genotoxic effects of neutrophils and hypochlorous acid. Mutagenesis 25, 149–154 (2010).

    PubMed  Google Scholar 

  74. Wilson, C. L. et al. NFκB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat. Commun. 6, 6818 (2015).

    CAS  PubMed  Google Scholar 

  75. Granot, Z. & Jablonska, J. Distinct functions of neutrophil in cancer and its regulation. Mediators Inflamm. 2015, 701067 (2015).

    PubMed  PubMed Central  Google Scholar 

  76. Tecchio, C., Scapini, P., Pizzolo, G. & Cassatella, M. A. On the cytokines produced by human neutrophils in tumors. Semin. Cancer Biol. 23, 159–170 (2013).

    CAS  PubMed  Google Scholar 

  77. Houghton, A. M. et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat. Med. 16, 219–223 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Lerman, I. et al. Infiltrating myeloid cells exert protumorigenic actions via neutrophil elastase. Mol. Cancer Res. 15, 1138–1152 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Caruso, J. A., Akli, S., Pageon, L., Hunt, K. K. & Keyomarsi, K. The serine protease inhibitor elafin maintains normal growth control by opposing the mitogenic effects of neutrophil elastase. Oncogene 34, 3556–3567 (2015).

    CAS  PubMed  Google Scholar 

  80. Wada, Y. et al. Neutrophil elastase induces cell proliferation and migration by the release of TGF-α, PDGF and VEGF in esophageal cell lines. Oncol. Rep. 17, 161–167 (2007).

    CAS  PubMed  Google Scholar 

  81. Nozawa, H., Chiu, C. & Hanahan, D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc. Natl Acad. Sci. USA 103, 12493–12498 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Scapini, P. et al. CXCL1/macrophage inflammatory protein-2-induced angiogenesis in vivo is mediated by neutrophil-derived vascular endothelial growth factor-A. J. Immunol. 172, 5034–5040 (2004).

    CAS  PubMed  Google Scholar 

  83. Shojaei, F., Singh, M., Thompson, J. D. & Ferrara, N. Role of Bv8 in neutrophil-dependent angiogenesis in a transgenic model of cancer progression. Proc. Natl Acad. Sci. USA 105, 2640–2645 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Albini, A., Bruno, A., Noonan, D. M. & Mortara, L. Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: implications for immunotherapy. Front. Immunol. 9, 527 (2018).

    PubMed  PubMed Central  Google Scholar 

  85. Phan, V. T. et al. Oncogenic RAS pathway activation promotes resistance to anti-VEGF therapy through G-CSF-induced neutrophil recruitment. Proc. Natl Acad. Sci. USA 110, 6079–6084 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Chung, A. S. et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat. Med. 19, 1114–1123 (2013).

    CAS  PubMed  Google Scholar 

  87. Shojaei, F. et al. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc. Natl Acad. Sci. USA 106, 6742–6747 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Tohme, S. et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 76, 1367–1380 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Guglietta, S. et al. Coagulation induced by C3aR-dependent NETosis drives protumorigenic neutrophils during small intestinal tumorigenesis. Nat. Commun. 7, 11037 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Albrengues, J. et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 361, eaao4227 (2018). This study demonstrates that proteolytic cleavage of the extracellular matrix protein laminin by proteases contained in NETs reveals a new epitope that triggers proliferation of cancer cells.

    PubMed  PubMed Central  Google Scholar 

  91. Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 8, 361ra138 (2016).

    PubMed  PubMed Central  Google Scholar 

  92. van der Windt, D. J. et al. Neutrophil extracellular traps promote inflammation and development of hepatocellular carcinoma in nonalcoholic steatohepatitis. Hepatology 68, 1347–1360 (2018).

    PubMed  Google Scholar 

  93. Cools-Lartigue, J. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Invest. 123, 3446–3458 (2013).

    CAS  PubMed Central  Google Scholar 

  94. Rayes, R. F. et al. Primary tumors induce neutrophil extracellular traps with targetable metastasis promoting effects. JCI Insight 5, e128008 (2019).

    Google Scholar 

  95. Aldabbous, L. et al. Neutrophil extracellular traps promote angiogenesis: evidence from vascular pathology in pulmonary hypertension. Arterioscler. Thromb. Vasc. Biol. 36, 2078–2087 (2016).

    CAS  PubMed  Google Scholar 

  96. Sceneay, J. et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72, 3906–3911 (2012).

    CAS  PubMed  Google Scholar 

  97. Jackstadt, R. et al. Epithelial NOTCH signaling rewires the tumor microenvironment of colorectal cancer to drive poor-prognosis subtypes and metastasis. Cancer Cell 36, 319–336.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Chen, M. B. et al. Inflamed neutrophils sequestered at entrapped tumor cells via chemotactic confinement promote tumor cell extravasation. Proc. Natl Acad. Sci. USA 115, 7022–7027 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Huh, S. J., Liang, S., Sharma, A., Dong, C. & Robertson, G. P. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 70, 6071–6082 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Spicer, J. D. et al. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 72, 3919–3927 (2012).

    CAS  PubMed  Google Scholar 

  101. Szczerba, B. M. et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 566, 553–557 (2019). This paper highlights that CTCs associated with neutrophils acquire a proliferative advantage.

    CAS  PubMed  Google Scholar 

  102. Ghajar, C. M. et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Catena, R. et al. Bone marrow-derived Gr1+ cells can generate a metastasis-resistant microenvironment via induced secretion of thrombospondin-1. Cancer Discov. 3, 578–589 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. El Rayes, T. et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl Acad. Sci. USA 112, 16000–16005 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Mensurado, S. et al. Tumor-associated neutrophils suppress pro-tumoral IL-17+ γδ T cells through induction of oxidative stress. PLoS Biol. 16, e2004990 (2018).

    PubMed  PubMed Central  Google Scholar 

  106. Schmielau, J. & Finn, O. J. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of T-cell function in advanced cancer patients. Cancer Res. 61, 4756–4760 (2001).

    CAS  PubMed  Google Scholar 

  107. Rice, C. M. et al. Tumour-elicited neutrophils engage mitochondrial metabolism to circumvent nutrient limitations and maintain immune suppression. Nat. Commun. 9, 5099 (2018).

    PubMed  PubMed Central  Google Scholar 

  108. 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 

  109. Rodriguez, P. C. et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 69, 1553–1560 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Liu, C. Y. et al. Population alterations of l-arginase- and inducible nitric oxide synthase-expressed CD11b+/CD14/CD15+/CD33+ myeloid-derived suppressor cells and CD8+ T lymphocytes in patients with advanced-stage non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 136, 35–45 (2010).

    CAS  PubMed  Google Scholar 

  111. O’Neill, L. A. & Pearce, E. J. Immunometabolism governs dendritic cell and macrophage function. J. Exp. Med. 213, 15–23 (2016).

    PubMed  PubMed Central  Google Scholar 

  112. Cubillos-Ruiz, J. R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Al-Khami, A. A. et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology 6, e1344804 (2017).

    PubMed  PubMed Central  Google Scholar 

  114. Condamine, T. et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 1, aaf8943 (2016).

    PubMed  PubMed Central  Google Scholar 

  115. Veglia, F. et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 569, 73–78 (2019). This paper shows that overexpression of FATP2 in neutrophils promotes the synthesis of PGE2 and the subsequent immunosuppressive activity.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Condamine, T. et al. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J. Clin. Invest. 124, 2626–2639 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Noman, M. Z. et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 211, 781–790 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. de Kleijn, S. et al. IFN-γ-stimulated neutrophils suppress lymphocyte proliferation through expression of PD-L1. PLoS ONE 8, e72249 (2013).

    PubMed  PubMed Central  Google Scholar 

  119. Cheng, Y. et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6–STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 9, 422 (2018).

    PubMed  PubMed Central  Google Scholar 

  120. Wang, T. T. et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF–PD-L1 pathway. Gut 66, 1900–1911 (2017).

    CAS  PubMed  Google Scholar 

  121. He, G. et al. Peritumoural neutrophils negatively regulate adaptive immunity via the PD-L1/PD-1 signalling pathway in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 34, 141 (2015).

    PubMed  PubMed Central  Google Scholar 

  122. Xu, W. et al. Immune-checkpoint protein VISTA regulates antitumor immunity by controlling myeloid cell-mediated inflammation and immunosuppression. Cancer Immunol. Res. 7, 1497–1510 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Wang, L. et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 208, 577–592 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Molgora, M. et al. The yin–yang of the interaction between myelomonocytic cells and NK cells. Scand. J. Immunol. 88, e12705 (2018).

    PubMed  PubMed Central  Google Scholar 

  125. Benigni, G. et al. CXCR3/CXCL10 axis regulates neutrophil–NK cell cross-talk determining the severity of experimental osteoarthritis. J. Immunol. 198, 2115–2124 (2017).

    CAS  PubMed  Google Scholar 

  126. Spiegel, A. et al. Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 6, 630–649 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Teijeira, A. et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity 52, 856–871 e858 (2020). This paper shows that NETs can protect tumour cells from immune cytotoxicity.

    CAS  PubMed  Google Scholar 

  128. Tumino, N. et al. PMN-MDSC are a new target to rescue graft-versus-leukemia activity of NK cells in haplo-HSC transplantation. Leukemia 34, 932–937 (2019).

    PubMed  PubMed Central  Google Scholar 

  129. Ogura, K. et al. NK cells control tumor-promoting function of neutrophils in mice. Cancer Immunol. Res. 6, 348–357 (2018).

    CAS  PubMed  Google Scholar 

  130. Ueda, R. et al. Interaction of natural killer cells with neutrophils exerts a significant antitumor immunity in hematopoietic stem cell transplantation recipients. Cancer Med. 5, 49–60 (2016).

    CAS  PubMed  Google Scholar 

  131. Colombo, M. P. et al. Granulocyte colony-stimulating factor gene transfer suppresses tumorigenicity of a murine adenocarcinoma in vivo. J. Exp. Med. 173, 889–897 (1991).

    CAS  PubMed  Google Scholar 

  132. Blaisdell, A. et al. Neutrophils oppose uterine epithelial carcinogenesis via debridement of hypoxic tumor cells. Cancer Cell 28, 785–799 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Mahiddine, K. et al. Relief of tumor hypoxia unleashes the tumoricidal potential of neutrophils. J. Clin. Invest. 130, 389–403 (2020).

    CAS  PubMed  Google Scholar 

  134. Gershkovitz, M. et al. TRPM2 mediates neutrophil killing of disseminated tumor cells. Cancer Res. 78, 2680–2690 (2018). This study demonstrates the mechanism responsible for the cytotoxic activity of neutrophil-derived ROS towards cancer cells.

    CAS  PubMed  Google Scholar 

  135. Gershkovitz, M., Fainsod-Levi, T., Zelter, T., Sionov, R. V. & Granot, Z. TRPM2 modulates neutrophil attraction to murine tumor cells by regulating CXCL2 expression. Cancer Immunol. Immunother. 68, 33–43 (2019).

    PubMed  Google Scholar 

  136. Finisguerra, V. et al. MET is required for the recruitment of anti-tumoural neutrophils. Nature 522, 349–353 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Koga, Y., Matsuzaki, A., Suminoe, A., Hattori, H. & Hara, T. Neutrophil-derived TNF-related apoptosis-inducing ligand (TRAIL): a novel mechanism of antitumor effect by neutrophils. Cancer Res. 64, 1037–1043 (2004).

    CAS  PubMed  Google Scholar 

  138. Glodde, N. et al. Reactive neutrophil responses dependent on the receptor tyrosine kinase c-MET limit cancer immunotherapy. Immunity 47, 789–802.e9 (2017).

    CAS  PubMed  Google Scholar 

  139. Hagerling, C. et al. Immune effector monocyte–neutrophil cooperation induced by the primary tumor prevents metastatic progression of breast cancer. Proc. Natl Acad. Sci. USA 116, 21704–21714 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Fridlender, Z. G. et al. Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils. PLoS ONE 7, e31524 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Governa, V. et al. The interplay between neutrophils and CD8+ T cells improves survival in human colorectal cancer. Clin. Cancer Res. 23, 3847–3858 (2017).

    CAS  PubMed  Google Scholar 

  142. Grivennikov, S. I. et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 491, 254–258 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Brennan, C. A. & Garrett, W. S. Gut microbiota, inflammation, and colorectal cancer. Annu. Rev. Microbiol. 70, 395–411 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Dmitrieva-Posocco, O. et al. Cell-type-specific responses to interleukin-1 control microbial invasion and tumor-elicited inflammation in colorectal cancer. Immunity 50, 166–180.e7 (2019). This study shows that IL-1 signalling in neutrophils has tumour-suppressive activity through the control of microbiota-induced inflammation.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Triner, D. et al. Neutrophils restrict tumor-associated microbiota to reduce growth and invasion of colon tumors in mice. Gastroenterology 156, 1467–1482 (2019).

    PubMed  Google Scholar 

  146. Colombo, M. P. et al. Granulocyte colony-stimulating factor (G-CSF) gene transduction in murine adenocarcinoma drives neutrophil-mediated tumor inhibition in vivo. Neutrophils discriminate between G-CSF-producing and G-CSF-nonproducing tumor cells. J. Immunol. 149, 113–119 (1992).

    CAS  PubMed  Google Scholar 

  147. Liu, Y. et al. CD11b+Ly6G+ cells inhibit tumor growth by suppressing IL-17 production at early stages of tumorigenesis. Oncoimmunology 5, e1061175 (2016).

    PubMed  Google Scholar 

  148. Mishalian, I. et al. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol. Immunother. 62, 1745–1756 (2013).

    CAS  PubMed  Google Scholar 

  149. Zhu, Y. P. et al. CyTOF reveals phenotypically-distinct human blood neutrophil populations differentially correlated with melanoma stage. Preprint at bioRxiv https://doi.org/10.1101/826644v2 (2019).

  150. Marini, O. et al. Mature CD10+ and immature CD10 neutrophils present in G-CSF-treated donors display opposite effects on T cells. Blood 129, 1343–1356 (2017).

    CAS  PubMed  Google Scholar 

  151. Templeton, A. J. et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J. Natl Cancer Inst. 106, dju124 (2014).

    PubMed  Google Scholar 

  152. Vano, Y. A. et al. Optimal cut-off for neutrophil-to-lymphocyte ratio: fact or fantasy? A prospective cohort study in metastatic cancer patients. PLoS ONE 13, e0195042 (2018).

    PubMed  PubMed Central  Google Scholar 

  153. Ivars Rubio, A. et al. Neutrophil–lymphocyte ratio in metastatic breast cancer is not an independent predictor of survival, but depends on other variables. Sci. Rep. 9, 16979 (2019).

    PubMed  PubMed Central  Google Scholar 

  154. Polidoro, M. A. et al. Impact of RAS mutations on the immune infiltrate of colorectal liver metastases: a preliminary study. J. Leukoc. Biol. https://doi.org/10.1002/JLB.5AB0220-608R (2020).

    Article  PubMed  Google Scholar 

  155. Kuang, D. M. et al. Peritumoral neutrophils link inflammatory response to disease progression by fostering angiogenesis in hepatocellular carcinoma. J. Hepatol. 54, 948–955 (2011).

    CAS  PubMed  Google Scholar 

  156. 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 

  157. Droeser, R. A. et al. High myeloperoxidase positive cell infiltration in colorectal cancer is an independent favorable prognostic factor. PLoS ONE 8, e64814 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhou, G. et al. CD177+ neutrophils suppress epithelial cell tumourigenesis in colitis-associated cancer and predict good prognosis in colorectal cancer. Carcinogenesis 39, 272–282 (2018).

    CAS  PubMed  Google Scholar 

  159. Zhang, H. et al. Tumor-infiltrating neutrophils is prognostic and predictive for postoperative adjuvant chemotherapy benefit in patients with gastric cancer. Ann. Surg. 267, 311–318 (2018).

    PubMed  Google Scholar 

  160. Posabella, A. et al. High density of CD66b in primary high-grade ovarian cancer independently predicts response to chemotherapy. J. Cancer Res. Clin. Oncol. 146, 127–136 (2020).

    CAS  PubMed  Google Scholar 

  161. Pylaeva, E. et al. NAMPT signaling is critical for the proangiogenic activity of tumor-associated neutrophils. Int. J. Cancer 144, 136–149 (2019).

    CAS  PubMed  Google Scholar 

  162. Shrestha, S. et al. Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonist attenuate tumor growth via polarization of neutrophils toward an antitumor phenotype. Oncoimmunology 5, e1067744 (2016).

    PubMed  Google Scholar 

  163. Yang, J. et al. Loss of CXCR4 in myeloid cells enhances antitumor immunity and reduces melanoma growth through NK cell and FASL mechanisms. Cancer Immunol. Res. 6, 1186–1198 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Matlung, H. L. et al. Neutrophils kill antibody-opsonized cancer cells by trogoptosis. Cell Rep. 23, 3946–3959.e6 (2018). This paper shows that blockade of the CD47–SIRPα checkpoint interaction increases the cytotoxic activity of neutrophils towards antibody-opsonized cancer cells.

    CAS  PubMed  Google Scholar 

  165. Chao, M. P. et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142, 699–713 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Ring, N. G. et al. Anti-SIRPα antibody immunotherapy enhances neutrophil and macrophage antitumor activity. Proc. Natl Acad. Sci. USA 114, E10578–E10585 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Chen, H. M. et al. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J. Clin. Invest. 128, 5647–5662 (2018).

    PubMed  PubMed Central  Google Scholar 

  168. Steele, C. W. et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell 29, 832–845 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Schalper, K. A. et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 26, 688–692 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Yuen, K. C. et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat. Med. 26, 693–698 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT03400332 (2018).

  172. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT03689699 (2018).

  173. Bilusic, M. et al. Phase I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J. Immunother. Cancer 7, 240 (2019).

    PubMed  PubMed Central  Google Scholar 

  174. Bertini, R. et al. Noncompetitive allosteric inhibitors of the inflammatory chemokine receptors CXCR1 and CXCR2: prevention of reperfusion injury. Proc. Natl Acad. Sci. USA 101, 11791–11796 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT03177187 (2017).

  176. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT01861054 (2013).

  177. Schott, A. F. et al. Phase Ib pilot study to evaluate reparixin in combination with weekly paclitaxel in patients with HER-2-negative metastatic breast cancer. Clin. Cancer Res. 23, 5358–5365 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT02001974 (2013).

  179. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02370238 (2015).

  180. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03161431 (2017).

  181. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03473925 (2018).

  182. Schall, T. J. & Proudfoot, A. E. Overcoming hurdles in developing successful drugs targeting chemokine receptors. Nat. Rev. Immunol. 11, 355–363 (2011).

    CAS  PubMed  Google Scholar 

  183. van Egmond, M. & Bakema, J. E. Neutrophils as effector cells for antibody-based immunotherapy of cancer. Semin. Cancer Biol. 23, 190–199 (2013).

    PubMed  Google Scholar 

  184. Brandsma, A. M. et al. Potent Fc receptor signaling by IgA leads to superior killing of cancer cells by neutrophils compared to IgG. Front. Immunol. 10, 704 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Pascal, V. et al. Anti-CD20 IgA can protect mice against lymphoma development: evaluation of the direct impact of IgA and cytotoxic effector recruitment on CD20 target cells. Haematologica 97, 1686–1694 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Treffers, L. W. et al. IgA-mediated killing of tumor cells by neutrophils is enhanced by CD47–SIRPα checkpoint inhibition. Cancer Immunol. Res. 8, 120–130 (2020).

    CAS  PubMed  Google Scholar 

  187. Otten, M. A. et al. Enhanced FcαRI-mediated neutrophil migration towards tumour colonies in the presence of endothelial cells. Eur. J. Immunol. 42, 1815–1821 (2012).

    CAS  PubMed  Google Scholar 

  188. Feng, M. et al. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 19, 568–586 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Casey, S. C. et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science 352, 227–231 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Advani, R. et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 379, 1711–1721 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Baudhuin, J. et al. Exocytosis acts as a modulator of the ILT4-mediated inhibition of neutrophil functions. Proc. Natl Acad. Sci. USA 110, 17957–17962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Mantovani, A. & Longo, D. L. Macrophage checkpoint blockade in cancer—back to the future. N. Engl. J. Med. 379, 1777–1779 (2018).

    PubMed  Google Scholar 

  193. McCracken, M. N., Cha, A. C. & Weissman, I. L. Molecular pathways: activating T cells after cancer cell phagocytosis from blockade of CD47 “don’t eat me” signals. Clin. Cancer Res. 21, 3597–3601 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Tseng, D. et al. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl Acad. Sci. USA 110, 11103–11108 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Liu, X. et al. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 21, 1209–1215 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Soto-Pantoja, D. R. et al. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res. 74, 6771–6783 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Georgoudaki, A. M. et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 15, 2000–2011 (2016).

    CAS  PubMed  Google Scholar 

  198. Viitala, M. et al. Immunotherapeutic blockade of macrophage clever-1 reactivates the CD8+ T-cell response against immunosuppressive tumors. Clin. Cancer Res. 25, 3289–3303 (2019).

    CAS  PubMed  Google Scholar 

  199. Nakamura, K. & Smyth, M. J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol. Immunol. 17, 1–12 (2020).

    CAS  PubMed  Google Scholar 

  200. Mantovani, A. Reflections on immunological nomenclature: in praise of imperfection. Nat. Immunol. 17, 215–216 (2016).

    CAS  PubMed  Google Scholar 

  201. Dinarello, C. et al. IL-1 family nomenclature. Nat. Immunol. 11, 973 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Murray, P. J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    CAS  PubMed  Google Scholar 

  204. Chua, W., Charles, K. A., Baracos, V. E. & Clarke, S. J. Neutrophil/lymphocyte ratio predicts chemotherapy outcomes in patients with advanced colorectal cancer. Br. J. Cancer 104, 1288–1295 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Cha, Y. J., Park, E. J., Baik, S. H., Lee, K. Y. & Kang, J. Clinical significance of tumor-infiltrating lymphocytes and neutrophil-to-lymphocyte ratio in patients with stage III colon cancer who underwent surgery followed by FOLFOX chemotherapy. Sci. Rep. 9, 11617 (2019).

    PubMed  PubMed Central  Google Scholar 

  206. Dell’Aquila, E. et al. Prognostic and predictive role of neutrophil/lymphocytes ratio in metastatic colorectal cancer: a retrospective analysis of the TRIBE study by GONO. Ann. Oncol. 29, 924–930 (2018).

    PubMed  Google Scholar 

  207. Chen, Y. et al. Pretreatment neutrophil-to-lymphocyte ratio is correlated with response to neoadjuvant chemotherapy as an independent prognostic indicator in breast cancer patients: a retrospective study. BMC Cancer 16, 320 (2016).

    PubMed  PubMed Central  Google Scholar 

  208. Xu, J. et al. Association of neutrophil/lymphocyte ratio and platelet/lymphocyte ratio with ER and PR in breast cancer patients and their changes after neoadjuvant chemotherapy. Clin. Transl. Oncol. 19, 989–996 (2017).

    CAS  PubMed  Google Scholar 

  209. Chae, S. et al. Neutrophil–lymphocyte ratio predicts response to chemotherapy in triple-negative breast cancer. Curr. Oncol. 25, e113–e119 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Cassidy, M. R. et al. Neutrophil to lymphocyte ratio is associated with outcome during ipilimumab treatment. EBioMedicine 18, 56–61 (2017).

    PubMed  PubMed Central  Google Scholar 

  211. Capone, M. et al. Baseline neutrophil-to-lymphocyte ratio (NLR) and derived NLR could predict overall survival in patients with advanced melanoma treated with nivolumab. J. Immunother. Cancer 6, 74 (2018).

    PubMed  PubMed Central  Google Scholar 

  212. Ferrucci, P. F. et al. Baseline neutrophil-to-lymphocyte ratio is associated with outcome of ipilimumab-treated metastatic melanoma patients. Br. J. Cancer 112, 1904–1910 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Schmidt, H. et al. Pretreatment levels of peripheral neutrophils and leukocytes as independent predictors of overall survival in patients with American Joint Committee on Cancer stage IV melanoma: results of the EORTC 18951 biochemotherapy trial. J. Clin. Oncol. 25, 1562–1569 (2007).

    CAS  PubMed  Google Scholar 

  214. Zaragoza, J. et al. High neutrophil to lymphocyte ratio measured before starting ipilimumab treatment is associated with reduced overall survival in patients with melanoma. Br. J. Dermatol. 174, 146–151 (2016).

    CAS  PubMed  Google Scholar 

  215. Khoja, L. et al. The full blood count as a biomarker of outcome and toxicity in ipilimumab-treated cutaneous metastatic melanoma. Cancer Med. 5, 2792–2799 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Jung, M. et al. Ipilimumab real-world efficacy and safety in Korean melanoma patients from the Korean named-patient program cohort. Cancer Res. Treat. 49, 44–53 (2017).

    CAS  PubMed  Google Scholar 

  217. Rosner, S. et al. Peripheral blood clinical laboratory variables associated with outcomes following combination nivolumab and ipilimumab immunotherapy in melanoma. Cancer Med. 7, 690–697 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Cho, H. et al. Pre-treatment neutrophil to lymphocyte ratio is elevated in epithelial ovarian cancer and predicts survival after treatment. Cancer Immunol. Immunother. 58, 15–23 (2009).

    CAS  PubMed  Google Scholar 

  219. Wisdom, A. J. et al. Neutrophils promote tumor resistance to radiation therapy. Proc. Natl Acad. Sci. USA 116, 18584–18589 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Amato, R. J., Xiong, Y., Peng, H. & Mohlere, V. Clinical outcomes model in renal cell cancer patients treated with modified vaccinia Ankara plus tumor-associated antigen 5T4. Int. J. Biol. Markers 30, e111–e121 (2015).

    CAS  PubMed  Google Scholar 

  221. Bilen, M. A. et al. Association between pretreatment neutrophil-to-lymphocyte ratio and outcome of patients with metastatic renal-cell carcinoma treated with nivolumab. Clin. Genitourin. Cancer 16, e563–e575 (2018).

    PubMed  PubMed Central  Google Scholar 

  222. Sharaiha, R. Z. et al. Elevated preoperative neutrophil:lymphocyte ratio as a predictor of postoperative disease recurrence in esophageal cancer. Ann. Surg. Oncol. 18, 3362–3369 (2011).

    PubMed  PubMed Central  Google Scholar 

  223. Fukui, T. et al. Activity of nivolumab and utility of neutrophil-to-lymphocyte ratio as a predictive biomarker for advanced non-small-cell lung cancer: a prospective observational study. Clin. Lung Cancer 20, 208–214.e2 (2019).

    CAS  PubMed  Google Scholar 

  224. Diem, S. et al. Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) as prognostic markers in patients with non-small cell lung cancer (NSCLC) treated with nivolumab. Lung Cancer 111, 176–181 (2017).

    PubMed  Google Scholar 

  225. Bagley, S. J. et al. Pretreatment neutrophil-to-lymphocyte ratio as a marker of outcomes in nivolumab-treated patients with advanced non-small-cell lung cancer. Lung Cancer 106, 1–7 (2017).

    PubMed  Google Scholar 

  226. Shiroyama, T. et al. Pretreatment advanced lung cancer inflammation index (ALI) for predicting early progression in nivolumab-treated patients with advanced non-small cell lung cancer. Cancer Med. 7, 13–20 (2018).

    CAS  PubMed  Google Scholar 

  227. Nakaya, A. et al. Neutrophil-to-lymphocyte ratio as an early marker of outcomes in patients with advanced non-small-cell lung cancer treated with nivolumab. Int. J. Clin. Oncol. 23, 634–640 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Russo, A. et al. Baseline neutrophilia, derived neutrophil-to-lymphocyte ratio (dNLR), platelet-to-lymphocyte ratio (PLR), and outcome in non small cell lung cancer (NSCLC) treated with nivolumab or docetaxel. J. Cell Physiol. 233, 6337–6343 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Tanizaki, J. et al. Peripheral blood biomarkers associated with clinical outcome in non-small cell lung cancer patients treated with nivolumab. J. Thorac. Oncol. 13, 97–105 (2018).

    CAS  PubMed  Google Scholar 

  230. Koinis, F. et al. Effect of first-line treatment on myeloid-derived suppressor cells’ subpopulations in the peripheral blood of patients with non-small cell lung cancer. J. Thorac. Oncol. 11, 1263–1272 (2016).

    PubMed  Google Scholar 

  231. Liu, Z. L. et al. Neutrophil–lymphocyte ratio as a prognostic marker for chemotherapy in advanced lung cancer. Int. J. Biol. Markers 31, e395–e401 (2016).

    CAS  PubMed  Google Scholar 

  232. Schernberg, A., Blanchard, P., Chargari, C. & Deutsch, E. Neutrophils, a candidate biomarker and target for radiation therapy? Acta Oncol. 56, 1522–1530 (2017).

    CAS  PubMed  Google Scholar 

  233. Xie, X. et al. Prognostic value of baseline neutrophil-to-lymphocyte ratio in outcome of immune checkpoint inhibitors. Cancer Invest. 37, 265–274 (2019).

    CAS  PubMed  Google Scholar 

  234. Sacdalan, D. B., Lucero, J. A. & Sacdalan, D. L. Prognostic utility of baseline neutrophil-to-lymphocyte ratio in patients receiving immune checkpoint inhibitors: a review and meta-analysis. Onco Targets Ther. 11, 955–965 (2018).

    PubMed  PubMed Central  Google Scholar 

  235. Schiffmann, L. M. et al. Tumour-infiltrating neutrophils counteract anti-VEGF therapy in metastatic colorectal cancer. Br. J. Cancer 120, 69–78 (2019).

    CAS  PubMed  Google Scholar 

  236. Wang, J. et al. Tumor-infiltrating neutrophils predict prognosis and adjuvant chemotherapeutic benefit in patients with biliary cancer. Cancer Sci. 109, 2266–2274 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Kargl, J. et al. Neutrophil content predicts lymphocyte depletion and anti-PD1 treatment failure in NSCLC. JCI Insight 4, e130850 (2019).

    PubMed Central  Google Scholar 

  238. Manfroi, B. et al. Tumor-associated neutrophils correlate with poor prognosis in diffuse large B-cell lymphoma patients. Blood Cancer J. 8, 66 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Zhou, S. L. et al. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology 150, 1646–1658.e17 (2016).

    CAS  PubMed  Google Scholar 

  240. Matsumoto, Y. et al. The significance of tumor-associated neutrophil density in uterine cervical cancer treated with definitive radiotherapy. Gynecol. Oncol. 145, 469–475 (2017).

    PubMed  Google Scholar 

  241. Wang, J., Shiratori, I., Uehori, J., Ikawa, M. & Arase, H. Neutrophil infiltration during inflammation is regulated by PILRα via modulation of integrin activation. Nat. Immunol. 14, 34–40 (2013).

    CAS  PubMed  Google Scholar 

  242. Strauss, L. et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci. Immunol. 5, eaay1863 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Jenmalm, M. C., Cherwinski, H., Bowman, E. P., Phillips, J. H. & Sedgwick, J. D. Regulation of myeloid cell function through the CD200 receptor. J. Immunol. 176, 191–199 (2006).

    CAS  PubMed  Google Scholar 

  244. Casulli, J. et al. CD200R deletion promotes a neutrophil niche for Francisella tularensis and increases infectious burden and mortality. Nat. Commun. 10, 2121 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Boxio, R., Bossenmeyer-Pourie, C., Steinckwich, N., Dournon, C. & Nusse, O. Mouse bone marrow contains large numbers of functionally competent neutrophils. J. Leukoc. Biol. 75, 604–611 (2004).

    CAS  PubMed  Google Scholar 

  246. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).

    CAS  PubMed  Google Scholar 

  247. Grassi, L. et al. Dynamics of transcription regulation in human bone marrow myeloid differentiation to mature blood neutrophils. Cell Rep. 24, 2784–2794 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Giladi, A. et al. Single-cell characterization of haematopoietic progenitors and their trajectories in homeostasis and perturbed haematopoiesis. Nat. Cell Biol. 20, 836–846 (2018).

    CAS  PubMed  Google Scholar 

  249. Evrard, M. et al. Developmental analysis of bone marrow neutrophils reveals populations specialized in expansion, trafficking, and effector functions. Immunity 48, 364–379.e8 (2018). This study identifies populations of NI in the bone marrow using mass cytometry.

    CAS  PubMed  Google Scholar 

  250. Shaul, M. E. et al. Circulating neutrophil subsets in advanced lung cancer patients exhibit unique immune signature and relate to prognosis. FASEB J. 34, 4204–4218 (2020).

    CAS  PubMed  Google Scholar 

  251. Engblom, C. et al. Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neutrophils. Science 358, eaal5081 (2017).

    PubMed  PubMed Central  Google Scholar 

  252. Lee, P. Y., Wang, J. X., Parisini, E., Dascher, C. C. & Nigrovic, P. A. Ly6 family proteins in neutrophil biology. J. Leukoc. Biol. 94, 585–594 (2013).

    CAS  PubMed  Google Scholar 

  253. Daley, J. M., Thomay, A. A., Connolly, M. D., Reichner, J. S. & Albina, J. E. Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J. Leukoc. Biol. 83, 64–70 (2008).

    CAS  PubMed  Google Scholar 

  254. Faget, J. et al. Efficient and specific Ly6G+ cell depletion: a change in the current practices toward more relevant functional analyses of neutrophils. Preprint at bioRxiv https://doi.org/10.1101/498881v1 (2018).

  255. Moses, K. et al. Survival of residual neutrophils and accelerated myelopoiesis limit the efficacy of antibody-mediated depletion of Ly-6G+ cells in tumor-bearing mice. J. Leukoc. Biol. 99, 811–823 (2016).

    CAS  PubMed  Google Scholar 

  256. Bruhn, K. W., Dekitani, K., Nielsen, T. B., Pantapalangkoor, P. & Spellberg, B. Ly6G-mediated depletion of neutrophils is dependent on macrophages. Results Immunol. 6, 5–7 (2016).

    PubMed  Google Scholar 

  257. Gael Boivin, G. et al. Durable and controlled depletion of neutrophils in mice. Nat. Commun. 11, 2762 (2020).

    PubMed  PubMed Central  Google Scholar 

  258. Csepregi, J. Z. et al. Myeloid-specific deletion of Mcl-1 yields severely neutropenic mice that survive and breed in homozygous form. J. Immunol. 201, 3793–3803 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Stackowicz, J., Jonsson, F. & Reber, L. L. Mouse models and tools for the in vivo study of neutrophils. Front. Immunol. 10, 3130 (2019).

    CAS  PubMed  Google Scholar 

  260. Dzhagalov, I., St John, A. & He, Y. W. The antiapoptotic protein Mcl-1 is essential for the survival of neutrophils but not macrophages. Blood 109, 1620–1626 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Hasenberg, A. et al. Catchup: a mouse model for imaging-based tracking and modulation of neutrophil granulocytes. Nat. Methods 12, 445–452 (2015).

    CAS  PubMed  Google Scholar 

  262. Romero-Moreno, R. et al. The CXCL5/CXCR2 axis is sufficient to promote breast cancer colonization during bone metastasis. Nat. Commun. 10, 4404 (2019).

    PubMed  PubMed Central  Google Scholar 

  263. Wislez, M. et al. High expression of ligands for chemokine receptor CXCR2 in alveolar epithelial neoplasia induced by oncogenic kras. Cancer Res. 66, 4198–4207 (2006).

    CAS  PubMed  Google Scholar 

  264. Purohit, A. et al. CXCR2 signaling regulates KRAS(G12D)-induced autocrine growth of pancreatic cancer. Oncotarget 7, 7280–7296 (2016).

    PubMed  PubMed Central  Google Scholar 

  265. Saintigny, P. et al. CXCR2 expression in tumor cells is a poor prognostic factor and promotes invasion and metastasis in lung adenocarcinoma. Cancer Res. 73, 571–582 (2013).

    CAS  PubMed  Google Scholar 

  266. Di Mitri, D. et al. Re-education of tumor-associated macrophages by CXCR2 blockade drives senescence and tumor inhibition in advanced prostate cancer. Cell Rep. 28, 2156–2168.e5 (2019).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Ministero dell’Istruzione, dell’Università e della Ricerca (MIUR) (PRIN 2015YYKPNN to A. M., PRIN 2017K7FSYB to S. J. and PRIN 20177J4E75 to R. B.), Ministero della Salute (GR-2016-02361263 to S. J. and GR-2016-02363531 to D. D. M.) and Fondazione AIRC per la ricerca sul cancro (AIRC IG-19014 to A. M., AIRC IG-22815 to S. J., AIRC IG-20269 to R. B. and AIRC Start-Up grant-19141 to D. D. M.) are acknowledged. A. P. is the recipient of a fellowship from Fondazione Umberto Veronesi (FUV).

Author information

Authors and Affiliations

Authors

Contributions

S. J., A. P., D. D. M., A. S., R. B. and A. M. all researched the data for the article. S. J., R. B. and A. M. provided substantial contributions to discussions of the content. S. J., A. P., D. D. M., A. S., R. B. and A. M. all contributed equally to writing the article. S. J., R. B. and A. M. reviewed and/or edited of the manuscript before submission.

Corresponding authors

Correspondence to Sebastien Jaillon or Alberto Mantovani.

Ethics declarations

Competing Interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks A. Hidalgo, Z. Granot and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Neutrophilia

An increase in the absolute number of peripheral blood neutrophils compared with the control value.

Oxysterols

Cholesterol metabolites involved in cholesterol homeostasis and immune and inflammatory responses. Oxysterols interact with the transcription factor liver X receptor (LXR) and the G protein-coupled receptors CXC-chemokine receptor 2 (CXCR2) and Epstein–Barr virus-induced G protein-coupled receptor 2 (EBI2).

Anaphylatoxin

A protein fragment (C3a and C5a) produced by cleavage of the complement components C3 and C5. Anaphylatoxins bind G protein-coupled receptors expressed on myeloid cells and are potent chemotactic agents.

Atypical chemokine receptor 2

(ACKR2). A seven-transmembrane receptor belonging to the chemokine receptor family that lacks a DRY motif and does not mediate chemotaxis (directional cell migration) but regulates chemokine bioavailability by scavenging, transcytosis or presentation of the ligand.

Pattern recognition receptors

Germline-encoded host sensors that recognize essential molecules expressed by microorganisms. Signalling through these receptors drives initiation of the inflammation, innate and adaptive responses.

Inflammasome

A multiprotein intracellular complex activated by pathogenic microorganisms and sterile stressors, important for the production of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18.

Neutrophil extracellular traps

(NETs). Networks of extracellular fibres composed of DNA and proteins, such as neutrophil elastase and histones, released from neutrophils upon activation.

Zeitgeber time

(ZT). A standardized unit of time based on a 12-h–12-h light–dark cycle. ZT0 corresponds to the beginning of the light phase and ZT12 to the beginning of the dark phase.

Reactive oxygen species

(ROS). Chemically reactive species containing oxygen that are produced through activation of the NADPH oxidase enzymatic system. ROS have important antimicrobial activity and induce genetic instability.

M1–M2

The extreme ends of a continuum of macrophage polarization states. Classically activated M1 macrophages in response to signals such as interferon-γ (IFNγ) mediate resistance to tumour formation, whereas in response to IL-4 and IL-13 an alternative form of macrophage activation (M2) that mediates tumour promotion is elicited.

N1 and N2

Antitumour neutrophils (N1) and pro-tumour neutrophils (N2). Interferon-β (IFNβ) and transforming growth factor-β (TGFβ) signalling pathways can trigger the formation of N1 and N2 neutrophils, respectively. N1 neutrophils are characterized by a normal density, a hypersegmented nucleus and a cytotoxic activity towards cancer cells, whereas N2 neutrophils have immunosuppressive activity. This classification may represent an oversimplification of neutrophil polarization, activation or maturation states.

Homologous recombination

Genetic recombination in which nucleotides are exchanged between two similar or identical molecules of DNA.

Neutrophil elastase

(NE). A serine protease stored in the neutrophil intracellular granules.

Circulating tumour cells

(CTCs). Cancer cells that have detached from a primary tumour and are found in the bloodstream.

Pre-metastatic niche

An environment in a secondary organ that provides favourable conditions for the seeding of metastatic cells.

Fc receptors

Surface receptors expressed by innate immune cells that recognize the Fc fragment of immunoglobulins. Human and mouse neutrophils express the IgG Fcγ receptors (FcγRs) and human neutrophils express the IgA Fcα receptor I (FcαRI).

Innate lymphoid cells

(ILCs). A group of cells of the innate immune response that belong to the lymphoid lineage and are characterized by a lack of antigen-specific receptors.

Unconventional T cells

(UTCs). A group of T lymphocytes that express the T cell receptor (TCR) αβ or γδ chains and are characterized by a lack of recognition of classical peptide antigens. UTCs include the mucosal-associated invariant T (MAIT) cells and invariant natural killer T (iNKT) cells.

Mass cytometry by time-of-flight

(CyTOF). A flow cytometry platform that utilizes elemental mass spectrometry to detect metal-conjugated antibodies that are bound intracellularly or extracellularly to antigens of interest in single cells.

Undifferentiated pleomorphic sarcoma

(UPS). An aggressive sarcoma of soft tissues or bone that occurs in any part of the body. UPS is characterized by the presence of pleomorphic spindle cells and large atypical cells exhibiting numerous irregular mitoses.

Antibody-dependent cellular cytotoxicity

(ADCC). Lysis of an antibody-coated target cell triggered via the interaction of target-bound antibodies with Fc receptors (FcRs) expressed by effector cells. ADCC can occur through the release of cytotoxic molecules, the expression of cell death-inducing molecules and trogoptosis.

Trogoptosis

A mechanism of cytotoxicity related to trogocytosis mediated by an active process of plasma membrane transfer between interacting cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jaillon, S., Ponzetta, A., Di Mitri, D. et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 20, 485–503 (2020). https://doi.org/10.1038/s41568-020-0281-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-0281-y

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