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 pro-tumorigenic host response to cancer therapies

Abstract

Resistance to cancer therapy remains a major challenge in clinical oncology. Although the initial treatment phase is often successful, eventual resistance, characterized by tumour relapse or spread, is discouraging. The majority of studies devoted to investigating the basis of resistance have focused on tumour-related changes that contribute to therapy resistance and tumour aggressiveness. However, over the last decade, the diverse roles of various host cells in promoting therapy resistance have become more appreciated. A growing body of evidence demonstrates that cancer therapy can induce host-mediated local and systemic responses, many of which shift the delicate balance within the tumour microenvironment, ultimately facilitating or supporting tumour progression. In this Review, recent advances in understanding how the host response to different cancer therapies may promote therapy resistance are discussed, with a focus on therapy-induced immunological, angiogenic and metastatic effects. Also summarized is the potential of evaluating the host response to cancer therapy in an era of precision medicine in oncology.

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

Fig. 1: Host-mediated immunological responses.
Fig. 2: Host-mediated angiogenic responses.
Fig. 3: Host-mediated metastatic responses.

Similar content being viewed by others

References

  1. Kerbel, R. S. Tumor angiogenesis. N. Engl. J. Med. 358, 2039–2049 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Yamaguchi, H. et al. Stromal fibroblasts mediate extracellular matrix remodeling and invasion of scirrhous gastric carcinoma cells. PLOS ONE 9, e85485 (2014).

    PubMed  PubMed Central  Google Scholar 

  3. Nadir, Y. & Brenner, B. Heparanase multiple effects in cancer. Thromb Res. 133, S90–S94 (2014).

    CAS  PubMed  Google Scholar 

  4. Shaked, Y. & Voest, E. E. Bone marrow derived cells in tumor angiogenesis and growth: are they the good, the bad or the evil? Biochim. Biophys. Acta 1796, 1–4 (2009).

    CAS  PubMed  Google Scholar 

  5. Harney, A. S. et al. Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov. 5, 932–943 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Powell, D. R. & Huttenlocher, A. Neutrophils in the tumor microenvironment. Trends Immunol. 37, 41–52 (2016).

    CAS  PubMed  Google Scholar 

  7. Maltby, S., Khazaie, K. & McNagny, K. M. Mast cells in tumor growth: angiogenesis, tissue remodelling and immune-modulation. Biochim. Biophys. Acta. 1796, 19–26 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Nieman, K. M., Romero, I. L., Van Houten, B. & Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim. Biophys. Acta 1831, 1533–1541 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Galluzzi, L., Chan, T. A., Kroemer, G., Wolchok, J. D. & Lopez-Soto, A. The hallmarks of successful anticancer immunotherapy. Sci. Transl Med. 10, eaat7807 (2018).

    PubMed  Google Scholar 

  10. Jayson, G. C., Kerbel, R., Ellis, L. M. & Harris, A. L. Antiangiogenic therapy in oncology: current status and future directions. Lancet 388, 518–529 (2016).

    CAS  PubMed  Google Scholar 

  11. Groenendijk, F. H. & Bernards, R. Drug resistance to targeted therapies: deja vu all over again. Mol. Oncol. 8, 1067–1083 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Mansoori, B., Mohammadi, A., Davudian, S., Shirjang, S. & Baradaran, B. The different mechanisms of cancer drug resistance: a brief review. Adv. Pharm. Bull. 7, 339–348 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    CAS  PubMed  Google Scholar 

  14. Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012). This major review focuses on the role of host cells within the TME, which contribute to the hallmarks of cancer.

    CAS  PubMed  Google Scholar 

  15. Shaked, Y. Balancing efficacy of and host immune responses to cancer therapy: the yin and yang effects. Nat. Rev. Clin. Oncol. 13, 611–626 (2016). This is perhaps the first extensive review on the pro-tumorigenic and anti-tumorigenic role of anticancer therapies and their impact on tumour regrowth and resistance to therapy, highlighting the balance between the action of and the reaction to anticancer drugs.

    CAS  PubMed  Google Scholar 

  16. Daenen, L. G. et al. Treatment-induced host-mediated mechanisms reducing the efficacy of antitumor therapies. Oncogene 33, 1341–1347 (2014).

    CAS  PubMed  Google Scholar 

  17. Kerbel, R. S. & Shaked, Y. Therapy-activated stromal cells can dictate tumor fate. J. Exp. Med. 213, 2831–2833 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Topalian, S. L., Drake, C. G. & Pardoll, D. M. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27, 450–461 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zagozdzon, R. & Golab, J. Immunomodulation by anticancer chemotherapy: more is not always better (review). Int. J. Oncol. 18, 417–424 (2001).

    CAS  PubMed  Google Scholar 

  20. Karagiannis, G. S., Condeelis, J. S. & Oktay, M. H. Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin. Exp. Metastasis 35, 269–284 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Blyth, B. J., Cole, A. J., MacManus, M. P. & Martin, O. A. Radiation therapy-induced metastasis: radiobiology and clinical implications. Clin. Exp. Metastasis 35, 223–236 (2018).

    CAS  PubMed  Google Scholar 

  22. Meeren, A. V., Bertho, J. M., Vandamme, M. & Gaugler, M. H. Ionizing radiation enhances IL-6 and IL-8 production by human endothelial cells. Mediators Inflamm. 6, 185–193 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Toste, P. A. et al. Chemotherapy-induced inflammatory gene signature and protumorigenic phenotype in pancreatic CAFs via stress-associated MAPK. Mol. Cancer Res. 14, 437–447 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Fisher, D. T., Appenheimer, M. M. & Evans, S. S. The two faces of IL-6 in the tumor microenvironment. Semin. Immunol. 26, 38–47 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. David, J. M., Dominguez, C., Hamilton, D. H. & Palena, C. The IL-8/IL-8R axis: a double agent in tumor immune resistance. Vaccines 4, E22 (2016).

    Google Scholar 

  26. Rialdi, A. et al. Topoisomerase 1 inhibition suppresses inflammatory genes and protects from death by inflammation. Science 352, aad7993 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Zhang, F. et al. Specific decrease in B-cell-derived extracellular vesicles enhances post-chemotherapeutic CD8+ T cell responses. Immunity 50, 738–750.e7 (2019).

    CAS  PubMed  Google Scholar 

  28. Ostrand-Rosenberg, S. & Sinha, P. Myeloid-derived suppressor cells: linking inflammation and cancer. J. Immunol. 182, 4499–4506 (2009).

    CAS  PubMed  Google Scholar 

  29. Bruchard, M. et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat. Med. 19, 57–64 (2013). This study describes the immunomodulatory effects of several chemotherapeutic drugs. For example, chemotherapy-activated MDSCs secrete immunomodulatory molecules that eventually contribute to tumour growth.

    CAS  PubMed  Google Scholar 

  30. Takeuchi, S. et al. Chemotherapy-derived inflammatory responses accelerate the formation of immunosuppressive myeloid cells in the tissue microenvironment of human pancreatic cancer. Cancer Res. 75, 2629–2640 (2015).

    CAS  PubMed  Google Scholar 

  31. Hasnis, E. et al. Anti-Bv8 antibody and metronomic gemcitabine improve pancreatic adenocarcinoma treatment outcome following weekly gemcitabine therapy. Neoplasia 16, 501–510 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kozin, S. V. et al. Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res. 70, 5679–5685 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Sugimura, K. et al. High infiltration of tumor-associated macrophages is associated with a poor response to chemotherapy and poor prognosis of patients undergoing neoadjuvant chemotherapy for esophageal cancer. J. Surg. Oncol. 111, 752–759 (2015).

    PubMed  Google Scholar 

  34. Cassetta, L. et al. Human tumor-associated macrophage and monocyte transcriptional landscapes reveal cancer-specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell 35, 588–602.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Mantovani, A. & Allavena, P. The interaction of anticancer therapies with tumor-associated macrophages. J. Exp. Med. 212, 435–445 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Sanchez, L. R. et al. The emerging roles of macrophages in cancer metastasis and response to chemotherapy. J. Leukoc. Biol. 106, 259–274 (2019).

    CAS  PubMed  Google Scholar 

  37. Mitchem, J. B. et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 73, 1128–1141 (2013).

    CAS  PubMed  Google Scholar 

  38. Ruffell, B. et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 26, 623–637 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. De Palma, M. & Lewis, C. E. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 23, 277–286 (2013).

    PubMed  Google Scholar 

  40. Gallin, E. K. & Green, S. W. Exposure to gamma-irradiation increases phorbol myristate acetate-induced H2O2 production in human macrophages. Blood 70, 694–701 (1987).

    CAS  PubMed  Google Scholar 

  41. Milas, L., Wike, J., Hunter, N., Volpe, J. & Basic, I. Macrophage content of murine sarcomas and carcinomas: associations with tumor growth parameters and tumor radiocurability. Cancer Res. 47, 1069–1075 (1987).

    CAS  PubMed  Google Scholar 

  42. Middleton, J. D., Stover, D. G. & Hai, T. Chemotherapy-exacerbated breast cancer metastasis: a paradox explainable by dysregulated adaptive-response. Int. J. Mol. Sci. 19, E3333 (2018).

    PubMed  Google Scholar 

  43. Gilbert, L. A. & Hemann, M. T. DNA damage-mediated induction of a chemoresistant niche. Cell 143, 355–366 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Oelmann, E. et al. Tissue inhibitor of metalloproteinases 1 is an autocrine and paracrine survival factor, with additional immune-regulatory functions, expressed by Hodgkin/Reed–Sternberg cells. Blood 99, 258–267 (2002).

    CAS  PubMed  Google Scholar 

  45. DeNardo, D. G. et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 1, 54–67 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Crohns, M. et al. Cytokines in bronchoalveolar lavage fluid and serum of lung cancer patients during radiotherapy—association of interleukin-8 and VEGF with survival. Cytokine 50, 30–36 (2010).

    CAS  PubMed  Google Scholar 

  47. Xu, J. et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 73, 2782–2794 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Nguyen, D. H. et al. Radiation acts on the microenvironment to affect breast carcinogenesis by distinct mechanisms that decrease cancer latency and affect tumor type. Cancer Cell 19, 640–651 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Wrzesinski, S. H., Wan, Y. Y. & Flavell, R. A. Transforming growth factor-β and the immune response: implications for anticancer therapy. Clin. Cancer Res. 13, 5262–5270 (2007).

    CAS  PubMed  Google Scholar 

  50. Mutsaers, A. J. et al. Dose-dependent increases in circulating TGF-α and other EGFR ligands act as pharmacodynamic markers for optimal biological dosing of cetuximab and are tumor independent. Clin. Cancer Res. 15, 2397–2405 (2009).

    CAS  PubMed  Google Scholar 

  51. Loupakis, F. et al. EGFR ligands as pharmacodynamic biomarkers in metastatic colorectal cancer patients treated with cetuximab and irinotecan. Target Oncol. 9, 205–214 (2014).

    PubMed  Google Scholar 

  52. Asimakopoulos, F. et al. Macrophages in multiple myeloma: emerging concepts and therapeutic implications. Leukemia Lymphoma 54, 2112–2121 (2013).

    PubMed  PubMed Central  Google Scholar 

  53. Beyar-Katz, O. et al. Bortezomib-induced pro-inflammatory macrophages as a potential factor limiting anti-tumour efficacy. J. Pathol. 239, 262–273 (2016).

    CAS  PubMed  Google Scholar 

  54. Zhang, W. et al. Depletion of tumor-associated macrophages enhances the effect of sorafenib in metastatic liver cancer models by antimetastatic and antiangiogenic effects. Clin. Cancer Res. 16, 3420–3430 (2010).

    CAS  PubMed  Google Scholar 

  55. Roberts, P. J. & Der, C. J. Targeting the Raf–MEK–ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 26, 3291–3310 (2007).

    CAS  PubMed  Google Scholar 

  56. Sharkey, M. S., Lizee, G., Gonzales, M. I., Patel, S. & Topalian, S. L. CD4+ T-cell recognition of mutated B-RAF in melanoma patients harboring the V599E mutation. Cancer Res. 64, 1595–1599 (2004).

    CAS  PubMed  Google Scholar 

  57. Wang, T. et al. BRAF inhibition stimulates melanoma-associated macrophages to drive tumor growth. Clin. Cancer Res. 21, 1652–1664 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Demicheli, R., Retsky, M. W., Hrushesky, W. J., Baum, M. & Gukas, I. D. The effects of surgery on tumor growth: a century of investigations. Ann. Oncol. 19, 1821–1828 (2008). This review is among several other studies and reviews summarizing the long history leading up to the concept of surgery-induced metastasis. The review focuses on tumour dormancy, circulating tumour cells and clinical studies that support this concept.

    CAS  PubMed  Google Scholar 

  59. Shakhar, G. & Ben-Eliyahu, S. Potential prophylactic measures against postoperative immunosuppression: could they reduce recurrence rates in oncological patients? Ann. Surg. Oncol. 10, 972–992 (2003).

    PubMed  Google Scholar 

  60. Crucitti, A. et al. Laparoscopic surgery for colorectal cancer is not associated with an increase in the circulating levels of several inflammation-related factors. Cancer Biol. Ther. 16, 671–677 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Alieva, M., van Rheenen, J. & Broekman, M. L. D. Potential impact of invasive surgical procedures on primary tumor growth and metastasis. Clin. Exp. Metastasis 35, 319–331 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Predina, J. et al. Changes in the local tumor microenvironment in recurrent cancers may explain the failure of vaccines after surgery. Proc. Natl Acad. Sci. USA 110, E415–E424 (2013).

    CAS  PubMed  Google Scholar 

  63. Sammour, T., Kahokehr, A., Chan, S., Booth, R. J. & Hill, A. G. The humoral response after laparoscopic versus open colorectal surgery: a meta-analysis. J. Surg. Res. 164, 28–37 (2010).

    PubMed  Google Scholar 

  64. Tartter, P. I., Steinberg, B., Barron, D. M. & Martinelli, G. The prognostic significance of natural killer cytotoxicity in patients with colorectal cancer. Arch. Surg. 122, 1264–1268 (1987).

    CAS  PubMed  Google Scholar 

  65. Fujisawa, T. & Yamaguchi, Y. Autologous tumor killing activity as a prognostic factor in primary resected nonsmall cell carcinoma of the lung. Cancer 79, 474–481 (1997).

    CAS  PubMed  Google Scholar 

  66. Shaashua, L. et al. Perioperative COX-2 and β-adrenergic blockade improves metastatic biomarkers in breast cancer patients in a phase-II randomized trial. Clin. Cancer Res. 23, 4651–4661 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Horowitz, M., Neeman, E., Sharon, E. & Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 12, 213–226 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  69. Ellis, L. M. & Hicklin, D. J. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat. Rev. Cancer 8, 579–591 (2008).

    CAS  PubMed  Google Scholar 

  70. Shaked, Y. et al. Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents. Cancer Cell 14, 263–273 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Alishekevitz, D. et al. Macrophage-induced lymphangiogenesis and metastasis following paclitaxel chemotherapy is regulated by VEGFR3. Cell Rep. 17, 1344–1356 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Shaked, Y. et al. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science 313, 1785–1787 (2006). This study is perhaps one of the first demonstrating how therapy induces a systemic angiogenic response that, in turn, contributes to tumour regrowth. In response to VDAs, endothelial precursor cells rapidly home to the treated tumour site, supporting angiogenesis and subsequent tumour regrowth.

    CAS  PubMed  Google Scholar 

  73. Natori, T. et al. G-CSF stimulates angiogenesis and promotes tumor growth: potential contribution of bone marrow-derived endothelial progenitor cells. Biochem. Biophys. Res. Commun. 297, 1058–1061 (2002).

    CAS  PubMed  Google Scholar 

  74. Furstenberger, G. et al. Circulating endothelial cells and angiogenic serum factors during neoadjuvant chemotherapy of primary breast cancer. Br. J. Cancer. 94, 524–531 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Okazaki, T. et al. Granulocyte colony-stimulating factor promotes tumor angiogenesis via increasing circulating endothelial progenitor cells and Gr1+CD11b+ cells in cancer animal models. Int. Immunol. 18, 1–9 (2006).

    CAS  PubMed  Google Scholar 

  76. Farace, F., Massard, C., Borghi, E., Bidart, J. M. & Soria, J. C. Vascular disrupting therapy-induced mobilization of circulating endothelial progenitor cells. Ann. Oncol. 18, 1421–1422 (2007).

    CAS  PubMed  Google Scholar 

  77. Taylor, M. et al. Reversing resistance to vascular-disrupting agents by blocking late mobilization of circulating endothelial progenitor cells. Cancer Discov. 2, 434–449 (2012).

    CAS  PubMed  Google Scholar 

  78. Fremder, E. et al. Tumor-derived microparticles induce bone marrow-derived cell mobilization and tumor homing: a process regulated by osteopontin. Int. J. Cancer 135, 270–281 (2014).

    CAS  PubMed  Google Scholar 

  79. Welford, A. F. et al. TIE2-expressing macrophages limit the therapeutic efficacy of the vascular-disrupting agent combretastatin A4 phosphate in mice. J. Clin. Invest. 121, 1969–1973 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Hughes, R. et al. Perivascular M2 macrophages stimulate tumor relapse after chemotherapy. Cancer Res. 75, 3479–3491 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Liu, T. et al. Tumor-associated macrophages in human breast cancer produce new monocyte attracting and pro-angiogenic factor YKL-39 indicative for increased metastasis after neoadjuvant chemotherapy. Oncoimmunology 7, e1436922 (2018).

    PubMed  PubMed Central  Google Scholar 

  82. Leibovich, S. J. et al. Macrophage-induced angiogenesis is mediated by tumour necrosis factor-α. Nature 329, 630–632 (1987).

    CAS  PubMed  Google Scholar 

  83. Sangaletti, S. et al. Oncogene-driven intrinsic inflammation induces leukocyte production of tumor necrosis factor that critically contributes to mammary carcinogenesis. Cancer Res. 70, 7764–7775 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Sprowl, J. A. et al. Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: a study in isogenic resistant tumor cells. Breast Cancer Res. 14, R2 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Kioi, M. et al. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Invest. 120, 694–705 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Heckmann, M., Douwes, K., Peter, R. & Degitz, K. Vascular activation of adhesion molecule mRNA and cell surface expression by ionizing radiation. Exp. Cell Res. 238, 148–154 (1998).

    CAS  PubMed  Google Scholar 

  87. Cox, T. R. et al. LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res. 73, 1721–1732 (2013). This paper is one of a few published studies that demonstrates the link between LOX and ECM remodelling leading to fibrosis and metastasis. LOX-induced lung fibrosis accounts for metastatic spread.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Oh, E. T. et al. Radiation-induced angiogenic signaling pathway in endothelial cells obtained from normal and cancer tissue of human breast. Oncogene 33, 1229–1238 (2014).

    CAS  PubMed  Google Scholar 

  89. Sofia Vala, I. et al. Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis. PLOS ONE 5, e11222 (2010).

    PubMed  PubMed Central  Google Scholar 

  90. Park, H. J., Griffin, R. J., Hui, S., Levitt, S. H. & Song, C. W. Radiation-induced vascular damage in tumors: implications of vascular damage in ablative hypofractionated radiotherapy (SBRT and SRS). Radiat. Res. 177, 311–327 (2012).

    CAS  PubMed  Google Scholar 

  91. Garcia-Barros, M. et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science 300, 1155–1159 (2003).

    CAS  PubMed  Google Scholar 

  92. Nikolinakos, P. G. et al. Plasma cytokine and angiogenic factor profiling identifies markers associated with tumor shrinkage in early-stage non-small cell lung cancer patients treated with pazopanib. Cancer Res. 70, 2171–2179 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Ebos, J. M., Lee, C. R., Christensen, J. G., Mutsaers, A. J. & Kerbel, R. S. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc. Natl Acad. Sci. USA 104, 17069–17074 (2007). This paper is one of the first to demonstrate a host systemic effect in response to the anti-angiogenic receptor tyrosine kinase inhibitor sunitinib. Circulating factors whose levels change in response to treatment are suggested as biomarkers for drug activity.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Mancuso, M. R. et al. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J. Clin. Invest. 116, 2610–2621 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Rigamonti, N. et al. Role of angiopoietin-2 in adaptive tumor resistance to VEGF signaling blockade. Cell Rep. 8, 696–706 (2014).

    CAS  PubMed  Google Scholar 

  96. Scholz, A. et al. Endothelial cell-derived angiopoietin-2 is a therapeutic target in treatment-naive and bevacizumab-resistant glioblastoma. EMBO Mol. Med. 8, 39–57 (2016).

    CAS  PubMed  Google Scholar 

  97. Shojaei, F. et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat. Biotechnol. 25, 911–920 (2007).

    CAS  PubMed  Google Scholar 

  98. Shojaei, F. et al. Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature 450, 825–831 (2007).

    CAS  PubMed  Google Scholar 

  99. Keklikoglou, I. et al. Periostin limits tumor response to VEGFA inhibition. Cell Rep. 22, 2530–2540 (2018).

    CAS  PubMed  Google Scholar 

  100. Cooke, V. G. et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 21, 66–81 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Crawford, Y. & Ferrara, N. Tumor and stromal pathways mediating refractoriness/resistance to anti-angiogenic therapies. Trends Pharmacol. Sci. 30, 624–630 (2009).

    CAS  PubMed  Google Scholar 

  102. Crawford, Y. et al. PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell 15, 21–34 (2009).

    CAS  PubMed  Google Scholar 

  103. Guan, H., Jia, S. F., Zhou, Z., Stewart, J. & Kleinerman, E. S. Herceptin down-regulates HER-2/neu and vascular endothelial growth factor expression and enhances taxol-induced cytotoxicity of human Ewing’s sarcoma cells in vitro and in vivo. Clin. Cancer Res. 11, 2008–2017 (2005).

    CAS  PubMed  Google Scholar 

  104. Izumi, Y., Xu, L., di Tomaso, E., Fukumura, D. & Jain, R. K. Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature 416, 279–280 (2002).

    CAS  PubMed  Google Scholar 

  105. Minder, P., Zajac, E., Quigley, J. P. & Deryugina, E. I. EGFR regulates the development and microarchitecture of intratumoral angiogenic vasculature capable of sustaining cancer cell intravasation. Neoplasia 17, 634–649 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Wang, W. M. et al. Epidermal growth factor receptor inhibition reduces angiogenesis via hypoxia-inducible factor-1α and Notch1 in head neck squamous cell carcinoma. PLOS ONE 10, e0119723 (2015).

    PubMed  PubMed Central  Google Scholar 

  107. Forget, P., Simonet, O. & De Kock, M. Cancer surgery induces inflammation, immunosuppression and neo-angiogenesis, but is it influenced by analgesics? F1000Res 2, 102 (2013).

    PubMed  PubMed Central  Google Scholar 

  108. Hofer, S. O. et al. The effect of surgical wounding on tumour development. Eur J. Surg. Oncol. 25, 231–243 (1999).

    CAS  PubMed  Google Scholar 

  109. Curigliano, G. et al. Systemic effects of surgery: quantitative analysis of circulating basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and transforming growth factor β (TGF-β) in patients with breast cancer who underwent limited or extended surgery. Breast Cancer Res. Treat 93, 35–40 (2005).

    CAS  PubMed  Google Scholar 

  110. Bono, A. et al. Angiogenic cells, macroparticles and RNA transcripts in laparoscopic vs open surgery for colorectal cancer. Cancer Biol. Ther. 10, 682–685 (2010).

    CAS  PubMed  Google Scholar 

  111. Langenberg, M. H. et al. Liver surgery induces an immediate mobilization of progenitor cells in liver cancer patients: a potential role for G-CSF. Cancer Biol. Ther. 9, 743–748 (2010).

    CAS  PubMed  Google Scholar 

  112. Rachman-Tzemah, C. et al. Blocking surgically induced lysyl oxidase activity reduces the risk of lung metastases. Cell Rep. 19, 774–784 (2017). This paper describes a novel mechanism by which surgery induces metastasis. The paper demonstrates in mice that lungs are more prone to metastatic seeding after an abdominal surgical incision, owing to ECM remodelling at sites distant from the surgical wound.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Folkman, J. Angiogenesis and apoptosis. Semin. Cancer Biol. 13, 159–167 (2003).

    CAS  PubMed  Google Scholar 

  114. Retsky, M. et al. Hypothesis: Induced angiogenesis after surgery in premenopausal node-positive breast cancer patients is a major underlying reason why adjuvant chemotherapy works particularly well for those patients. Breast Cancer Res. 6, R372–R374 (2004).

    PubMed  PubMed Central  Google Scholar 

  115. Voloshin, T., Gingis-Velitski, S. & Shaked, Y. The angiogenic profile of colorectal cancer patients following open or laparoscopic colectomy. Cancer Biol. Ther. 10, 686–688 (2010).

    PubMed  Google Scholar 

  116. Valastyan, S. & Weinberg, R. A. Tumor metastasis: molecular insights and evolving paradigms. Cell 147, 275–292 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Lambert, A. W., Pattabiraman, D. R. & Weinberg, R. A. Emerging biological principles of metastasis. Cell 168, 670–691 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Joyce, J. A. & Pollard, J. W. Microenvironmental regulation of metastasis. Nat. Rev. Cancer 9, 239–252 (2009).

    CAS  PubMed  Google Scholar 

  119. Gingis-Velitski, S. et al. Host response to short-term, single-agent chemotherapy induces matrix metalloproteinase-9 expression and accelerates metastasis in mice. Cancer Res. 71, 6986–6996 (2011).

    CAS  PubMed  Google Scholar 

  120. Daenen, L. G. et al. Chemotherapy enhances metastasis formation via VEGFR-1-expressing endothelial cells. Cancer Res. 71, 6976–6985 (2011). Along with reference 119, this paper is one of the first studies describing a true systemic host response phenomenon following treatment with chemotherapeutic drugs.

    CAS  PubMed  Google Scholar 

  121. Kuonen, F., Secondini, C. & Ruegg, C. Molecular pathways: emerging pathways mediating growth, invasion, and metastasis of tumors progressing in an irradiated microenvironment. Clin. Cancer Res. 18, 5196–5202 (2012).

    CAS  PubMed  Google Scholar 

  122. Vilalta, M., Rafat, M., Giaccia, A. J. & Graves, E. E. Recruitment of circulating breast cancer cells is stimulated by radiotherapy. Cell Rep. 8, 402–409 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Vilalta, M., Rafat, M. & Graves, E. E. Effects of radiation on metastasis and tumor cell migration. Cell Mol. Life Sci. 73, 2999–3007 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Shree, T. et al. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 25, 2465–2479 (2011). This important paper demonstrates how macrophages contribute to tumour resistance, showing that, in response to several cytotoxic agents, macrophages express various cathepsins, which in turn protect tumour cells from the cytotoxic effect of the drug.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Voloshin, T. et al. Blocking IL1β pathway following paclitaxel chemotherapy slightly inhibits primary tumor growth but promotes spontaneous metastasis. Mol. Cancer Ther. 14, 1385–1394 (2015).

    CAS  PubMed  Google Scholar 

  126. Arwert, E. N. et al. A unidirectional transition from migratory to perivascular macrophage is required for tumor cell intravasation. Cell Rep. 23, 1239–1248 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Nakasone, E. S. et al. Imaging tumor–stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 21, 488–503 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Gutter-Kapon, L. et al. Heparanase is required for activation and function of macrophages. Proc. Natl Acad. Sci. USA 113, E7808–E7817 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Rastogi, P. et al. Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J. Clin. Oncol. 26, 778–785 (2008).

    PubMed  Google Scholar 

  130. Symmans, W. F. et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J. Clin. Oncol. 25, 4414–4422 (2007).

    PubMed  Google Scholar 

  131. Rohan, T. E. et al. Tumor microenvironment of metastasis and risk of distant metastasis of breast cancer. J. Natl Cancer Inst. 106 (2014).

  132. Sparano, J. A. et al. A metastasis biomarker (MetaSite Breast Score) is associated with distant recurrence in hormone receptor-positive, HER2-negative early-stage breast cancer. NPJ Breast Cancer 3, 42 (2017).

    PubMed  PubMed Central  Google Scholar 

  133. Karagiannis, G. S. et al. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci. Transl Med. 9, eaan0026 (2017). This seminal paper provides both preclinical and clinical evidence for the host response to chemotherapy and its contribution to metastasis. Specifically, the study reports the formation of TMEM in patients with breast cancer who underwent neoadjuvant chemotherapy, which increases the potential for metastasis.

    PubMed  PubMed Central  Google Scholar 

  134. DeMichele, A., Yee, D. & Esserman, L. Mechanisms of resistance to neoadjuvant chemotherapy in breast cancer. N. Engl. J. Med. 377, 2287–2289 (2017).

    PubMed  Google Scholar 

  135. Timaner, M. et al. Dequalinium blocks macrophage-induced metastasis following local radiation. Oncotarget 6, 27537–27554 (2015).

    PubMed  PubMed Central  Google Scholar 

  136. Keklikoglou, I. et al. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat. Cell Biol. 21, 190–202 (2019). This publication demonstrates that tumour cells at the primary tumour site secrete exosomes in response to chemotherapy, thereby contributing to the formation of the pre-metastatic niche and tumour cell seeding.

    CAS  PubMed  Google Scholar 

  137. Chang, Y. S., Jalgaonkar, S. P., Middleton, J. D. & Hai, T. Stress-inducible gene Atf3 in the noncancer host cells contributes to chemotherapy-exacerbated breast cancer metastasis. Proc. Natl Acad. Sci. USA 114, E7159–E7168 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Erler, J. T. et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15, 35–44 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Ebos, J. M. et al. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239 (2009). This study is one of the first to demonstrate that anti-angiogenic drugs inhibit primary tumour growth but at the same time induce metastasis in mouse tumour models. Host systemic effects in response to antiangiogenic drugs are suggested to be responsible for this phenomenon.

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Kerbel, R. S. & Ebos, J. M. Peering into the aftermath: the inhospitable host? Nat. Med. 16, 1084–1085 (2010).

    CAS  PubMed  Google Scholar 

  141. Rahbari, N. N. et al. Anti-VEGF therapy induces ECM remodeling and mechanical barriers to therapy in colorectal cancer liver metastases. Sci. Transl Med. 8, 360ra135 (2016). This study demonstrates that anti-angiogenic drugs may support liver metastasis in patients with colorectal cancer. Specifically, anti-VEGF therapy promotes the secretion of ECM molecules, which in turn contribute to tumour stiffness and metastasis.

    PubMed  PubMed Central  Google Scholar 

  142. Ando, N. et al. Surgery plus chemotherapy compared with surgery alone for localized squamous cell carcinoma of the thoracic esophagus: a Japan Clinical Oncology Group study—JCOG9204. J. Clin. Oncol. 21, 4592–4596 (2003).

    PubMed  Google Scholar 

  143. Hofman, V. et al. Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch Assay™ and the isolation by size of epithelial tumor cell method. Int. J. Cancer 129, 1651–1660 (2011).

    CAS  PubMed  Google Scholar 

  144. Retsky, M. et al. NSAID analgesic ketorolac used perioperatively may suppress early breast cancer relapse: particular relevance to triple negative subgroup. Breast Cancer Res. Treat 134, 881–888 (2012).

    CAS  PubMed  Google Scholar 

  145. Takemoto, Y. et al. The mobilization and recruitment of c-kit+ cells contribute to wound healing after surgery. PLOS ONE 7, e48052 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Ceelen, W., Pattyn, P. & Mareel, M. Surgery, wound healing, and metastasis: recent insights and clinical implications. Crit. Rev. Oncol. Hematol. 89, 16–26 (2014).

    PubMed  Google Scholar 

  147. Krall, J. A. et al. The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl Med. 10, eaan3464 (2018). This mechanistic study describes how surgery-induced immune modulation contributes to the outgrowth of dormant tumours at metastatic sites.

    PubMed  PubMed Central  Google Scholar 

  148. Panigrahy, D. et al. Preoperative stimulation of resolution and inflammation blockade eradicates micrometastases. J. Clin. Invest. 129, 2964–2979 (2019).

    PubMed  PubMed Central  Google Scholar 

  149. van der Bij, G. J. et al. The perioperative period is an underutilized window of therapeutic opportunity in patients with colorectal cancer. Ann. Surg. 249, 727–734 (2009).

    PubMed  Google Scholar 

  150. Jemal, A. et al. Cancer statistics, 2003. CA Cancer J. Clin. 53, 5–26 (2003).

    PubMed  Google Scholar 

  151. Demicheli, R., Abbattista, A., Miceli, R., Valagussa, P. & Bonadonna, G. Time distribution of the recurrence risk for breast cancer patients undergoing mastectomy: further support about the concept of tumor dormancy. Breast Cancer Res. Treat 41, 177–185 (1996).

    CAS  PubMed  Google Scholar 

  152. Shibue, T. & Weinberg, R. A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 14, 611–629 (2017).

    PubMed  PubMed Central  Google Scholar 

  153. Roodhart, J. M. et al. Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum-induced fatty acids. Cancer Cell 20, 370–383 (2011).

    CAS  PubMed  Google Scholar 

  154. Timaner, M. et al. Therapy-educated mesenchymal stem cells enrich for tumor-initiating cells. Cancer Res. 78, 1253–1265 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Chan, T. S. et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J. Exp. Med. 213, 2967–2988 (2016). This paper provides an explanation for tumour resistance following MTD chemotherapy in desmoplastic tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Su, S. et al. CD10+GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 172, 841–856.e16 (2018). This study demonstrates that only a subset of CAFs promote the enrichment of CSCs in response to chemotherapy.

    CAS  PubMed  Google Scholar 

  157. Olson, O. C., Kim, H., Quail, D. F., Foley, E. A. & Joyce, J. A. Tumor-associated macrophages suppress the cytotoxic activity of antimitotic agents. Cell Rep. 19, 101–113 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Beyar-Katz, O. et al. Pro-inflammatory macrophages promote multiple myeloma resistance to bortezomib therapy. Mol. Cancer Res. https://doi.org/10.1158/1541-7786.MCR-19-0487 (2019).

    PubMed  Google Scholar 

  159. Ma, J., Song, X., Xu, X. & Mou, Y. Cancer-associated fibroblasts promote the chemo-resistance in gastric cancer through secreting IL-11 targeting JAK/STAT3/Bcl2 pathway. Cancer Res. Treat 51, 194–210 (2019).

    CAS  PubMed  Google Scholar 

  160. Kim, S. J. et al. Astrocytes upregulate survival genes in tumor cells and induce protection from chemotherapy. Neoplasia 13, 286–298 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Lehuédé, C. et al. Adipocytes promote breast cancer resistance to chemotherapy, a process amplified by obesity: role of the major vault protein (MVP). Breast Cancer Res. 21, 7 (2019).

    PubMed  PubMed Central  Google Scholar 

  162. Sun, Y. et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat. Med. 18, 1359–1368 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  164. Ahmed, K. M., Zhang, H. & Park, C. C. NF-κB regulates radioresistance mediated by β1-integrin in three-dimensional culture of breast cancer cells. Cancer Res. 73, 3737–3748 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Hanahan, D., Bergers, G. & Bergsland, E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J. Clin. Invest. 105, 1045–1047 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Pasquier, E., Kavallaris, M. & Andre, N. Metronomic chemotherapy: new rationale for new directions. Nat. Rev. Clin. Oncol. 7, 455–465 (2010).

    PubMed  Google Scholar 

  167. 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 

  168. Shaked, Y. et al. Evidence implicating immunological host effects in the efficacy of metronomic low-dose chemotherapy. Cancer Res. 76, 5983–5993 (2016).

    CAS  PubMed  Google Scholar 

  169. Pietras, K. & Hanahan, D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J. Clin. Oncol. 23, 939–952 (2005).

    CAS  PubMed  Google Scholar 

  170. Shaked, Y. et al. Low-dose metronomic combined with intermittent bolus-dose cyclophosphamide is an effective long-term chemotherapy treatment strategy. Cancer Res. 65, 7045–7051 (2005).

    CAS  PubMed  Google Scholar 

  171. Simkens, L. H. et al. Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): a phase 3 randomised controlled trial of the Dutch Colorectal Cancer Group. Lancet 385, 1843–1852 (2015).

    CAS  PubMed  Google Scholar 

  172. Takahashi, Y., Mai, M., Sawabu, N. & Nishioka, K. A pilot study of individualized maximum repeatable dose (iMRD), a new dose finding system, of weekly gemcitabine for patients with metastatic pancreas cancer. Pancreas 30, 206–210 (2005).

    CAS  PubMed  Google Scholar 

  173. Benguigui, M. et al. Dose- and time-dependence of the host-mediated response to paclitaxel therapy: a mathematical modeling approach. Oncotarget 9, 2574–2590 (2018).

    PubMed  Google Scholar 

  174. West, J. & Newton, P. K. Chemotherapeutic dose scheduling based on tumor growth rates provides a case for low-dose metronomic high-entropy therapies. Cancer Res. 77, 6717–6728 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Chen, C. S., Doloff, J. C. & Waxman, D. J. Intermittent metronomic drug schedule is essential for activating antitumor innate immunity and tumor xenograft regression. Neoplasia 16, 84–96 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Singh, M. et al. Anti-VEGF antibody therapy does not promote metastasis in genetically engineered mouse tumour models. J. Pathol. 227, 417–430 (2012).

    CAS  PubMed  Google Scholar 

  177. Rodenhuis, S. The status of high-dose chemotherapy in breast cancer. Oncologist 5, 369–375 (2000).

    CAS  PubMed  Google Scholar 

  178. Shaked, Y. & Kerbel, R. S. Antiangiogenic strategies on defense: blocking rebound by the tumor vasculature after chemotherapy. Cancer Res. 67, 7055–7058 (2007).

    CAS  PubMed  Google Scholar 

  179. Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005).

    CAS  PubMed  Google Scholar 

  180. Kerbel, R. S. Antiangiogenic therapy: a universal chemosensitization strategy for cancer? Science 312, 1171–1175 (2006).

    CAS  PubMed  Google Scholar 

  181. Scagliotti, G. et al. Phase III study of carboplatin and paclitaxel alone or with sorafenib in advanced non-small-cell lung cancer. J. Clin. Oncol. 28, 1835–1842 (2010).

    CAS  PubMed  Google Scholar 

  182. Cannarile, M. A. et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 5, 53 (2017).

    PubMed  PubMed Central  Google Scholar 

  183. Lyons, Y. A. et al. Macrophage depletion through colony stimulating factor 1 receptor pathway blockade overcomes adaptive resistance to anti-VEGF therapy. Oncotarget 8, 96496–96505 (2017).

    PubMed  PubMed Central  Google Scholar 

  184. Salvagno, C. et al. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat. Cell Biol. 21, 511–521 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Ruffell, B. & Coussens, L. M. Macrophages and therapeutic resistance in cancer. Cancer Cell 27, 462–472 (2015). This review focuses on the role of macrophages in promoting therapy resistance mainly by modulating the immune system, and also discusses the therapeutic potential of blocking macrophage recruitment to the treated tumour site.

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Harney, A. S. et al. The selective Tie2 inhibitor rebastinib blocks recruitment and function of Tie2Hi macrophages in breast cancer and pancreatic neuroendocrine tumors. Mol. Cancer Ther. 16, 2486–2501 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Voloshin, T. et al. G-CSF supplementation with chemotherapy can promote revascularization and subsequent tumor regrowth: prevention by a CXCR4 antagonist. Blood 118, 3426–3435 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Kim, J. et al. Chemokine receptor CXCR4 expression in colorectal cancer patients increases the risk for recurrence and for poor survival. J. Clin. Oncol. 23, 2744–2753 (2005).

    CAS  PubMed  Google Scholar 

  189. Bracci, L., Schiavoni, G., Sistigu, A. & Belardelli, F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 21, 15–25 (2014).

    CAS  PubMed  Google Scholar 

  190. Gartung, A. et al. Suppression of chemotherapy-induced cytokine/lipid mediator surge and ovarian cancer by a dual COX-2/sEH inhibitor. Proc. Natl Acad. Sci. USA 116, 1698–1703 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Sulciner, M. L. et al. Resolvins suppress tumor growth and enhance cancer therapy. J. Exp. Med. 215, 115–140 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Pueyo, G. et al. Cetuximab may inhibit tumor growth and angiogenesis induced by ionizing radiation: a preclinical rationale for maintenance treatment after radiotherapy. Oncologist 15, 976–986 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Fuentes-Antras, J., Provencio, M. & Diaz-Rubio, E. Hyperprogression as a distinct outcome after immunotherapy. Cancer Treat Rev. 70, 16–21 (2018).

    CAS  PubMed  Google Scholar 

  194. Kamada, T. et al. PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl Acad. Sci. USA 116, 9999–10008 (2019). This study is one of the first that provides a mechanism explaining hyperprogression during treatment with ICIs. The authors demonstrate that hyperprogressive gastric carcinomas from patients treated with anti-PD1 therapy are infiltrated with proliferating T reg cells that suppress immune activity.

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Lo Russo, G. et al. Antibody-Fc/FcR interaction on macrophages as a mechanism for hyperprogressive disease in non-small cell lung cancer subsequent to PD-1/PD-L1 blockade. Clin. Cancer Res. 25, 989–999 (2019).

    CAS  PubMed  Google Scholar 

  196. 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 

  197. Motzer, R. J. et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 380, 1103–1115 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Rini, B. I. et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 380, 1116–1127 (2019).

    CAS  PubMed  Google Scholar 

  199. Pastula, A. & Marcinkiewicz, J. Myeloid-derived suppressor cells: a double-edged sword? Int. J. Exp. Pathol. 92, 73–78 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Ma, Y. et al. Tumor necrosis factor is dispensable for the success of immunogenic anticancer chemotherapy. Oncoimmunology 2, e24786 (2013).

    PubMed  PubMed Central  Google Scholar 

  201. Aymeric, L. et al. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 70, 855–858 (2010).

    CAS  PubMed  Google Scholar 

  202. Ciampricotti, M., Hau, C. S., Doornebal, C. W., Jonkers, J. & de Visser, K. E. Chemotherapy response of spontaneous mammary tumors is independent of the adaptive immune system. Nat. Med. 18, 344–346 (2012).

    CAS  PubMed  Google Scholar 

  203. Baumeister, S. H., Freeman, G. J., Dranoff, G. & Sharpe, A. H. Coinhibitory pathways in immunotherapy for cancer. Annu. Rev. Immunol. 34, 539–573 (2016).

    CAS  PubMed  Google Scholar 

  204. Azami, A. et al. Abscopal effect following radiation monotherapy in breast cancer: a case report. Mol. Clin. Oncol. 9, 283–286 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Yan, Y. et al. Combining immune checkpoint inhibitors with conventional cancer therapy. Front. Immunol. 9, 1739 (2018).

    PubMed  PubMed Central  Google Scholar 

  206. Derer, A., Frey, B., Fietkau, R. & Gaipl, U. S. Immune-modulating properties of ionizing radiation: rationale for the treatment of cancer by combination radiotherapy and immune checkpoint inhibitors. Cancer Immunol. Immunother. 65, 779–786 (2016).

    CAS  PubMed  Google Scholar 

  207. Postow, M. A. et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 366, 925–931 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Kasmann, L. et al. State of clinical research of radiotherapy/chemoradiotherapy and immune checkpoint inhibitor therapy combinations in solid tumours—a German radiation oncology survey. Eur. J. Cancer 108, 50–54 (2019).

    PubMed  Google Scholar 

  209. Wang, X. et al. Crosstalk between TEMs and endothelial cells modulates angiogenesis and metastasis via IGF1–IGF1R signalling in epithelial ovarian cancer. Br. J. Cancer 117, 1371–1382 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Oyama, T. et al. Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-κB activation in hemopoietic progenitor cells. J. Immunol. 160, 1224–1232 (1998).

    CAS  PubMed  Google Scholar 

  211. Ohm, J. E. et al. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood 101, 4878–4886 (2003).

    CAS  PubMed  Google Scholar 

  212. Osada, T. et al. The effect of anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer patients. Cancer Immunol. Immunother. 57, 1115–1124 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Fukumura, D., Kloepper, J., Amoozgar, Z., Duda, D. G. & Jain, R. K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 325–340 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Allen, E. et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci. Transl Med. 9, eaak9679 (2017).

    PubMed  PubMed Central  Google Scholar 

  215. Valpione, S. et al. Sex and interleukin-6 are prognostic factors for autoimmune toxicity following treatment with anti-CTLA4 blockade. J. Transl Med. 16, 94 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Tsukamoto, H. et al. Combined blockade of IL6 and PD-1/PD-L1 signaling abrogates mutual regulation of their immunosuppressive effects in the tumor microenvironment. Cancer Res. 78, 5011–5022 (2018).

    CAS  PubMed  Google Scholar 

  217. Li, J. et al. Targeting interleukin-6 (IL-6) sensitizes anti-PD-L1 treatment in a colorectal cancer preclinical model. Med. Sci. Monit. 24, 5501–5508 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Ilieva, K. M. et al. Effects of BRAF mutations and BRAF inhibition on immune responses to melanoma. Mol. Cancer Ther. 13, 2769–2783 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  219. Sasada, T., Azuma, K., Ohtake, J. & Fujimoto, Y. Immune responses to epidermal growth factor receptor (EGFR) and their application for cancer treatment. Front. Pharmacol. 7, 405 (2016).

    PubMed  PubMed Central  Google Scholar 

  220. Concha-Benavente, F. & Ferris, R. L. Reversing EGFR mediated immunoescape by targeted monoclonal antibody therapy. Front. Pharmacol. 8, 332 (2017).

    PubMed  PubMed Central  Google Scholar 

  221. Luke, J. J. Comprehensive clinical trial data summation for BRAF–MEK inhibition and checkpoint immunotherapy in metastatic melanoma. Oncologist https://doi.org/10.1634/theoncologist.2018-0876 (2019).

    PubMed  PubMed Central  Google Scholar 

  222. Krieg, C. et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat. Med. 24, 144–153 (2018).

    CAS  PubMed  Google Scholar 

  223. Sathyanarayanan, V. & Neelapu, S. S. Cancer immunotherapy: strategies for personalization and combinatorial approaches. Mol. Oncol. 9, 2043–2053 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Cassetta, L. & Kitamura, T. Macrophage targeting: opening new possibilities for cancer immunotherapy. Immunology 155, 285–293 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Zhu, Y. et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 74, 5057–5069 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Al-Lazikani, B., Banerji, U. & Workman, P. Combinatorial drug therapy for cancer in the post-genomic era. Nat. Biotechnol. 30, 679–692 (2012).

    CAS  PubMed  Google Scholar 

  227. Schadendorf, D. et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J. Clin. Oncol. 33, 1889–1894 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Harris, S. J., Brown, J., Lopez, J. & Yap, T. A. Immuno-oncology combinations: raising the tail of the survival curve. Cancer Biol. Med. 13, 171–193 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Bertolini, F., Sukhatme, V. P. & Bouche, G. Drug repurposing in oncology—patient and health systems opportunities. Nat. Rev. Clin. Oncol. 12, 732–742 (2015).

    PubMed  Google Scholar 

  230. Reinhold, W. C. et al. Using drug response data to identify molecular effectors, and molecular “omic” data to identify candidate drugs in cancer. Hum Genet. 134, 3–11 (2015).

    CAS  PubMed  Google Scholar 

  231. Spitzer, M. H. et al. Systemic immunity is required for effective cancer immunotherapy. Cell 168, 487–502.e15 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Gopalakrishnan, V., Helmink, B. A., Spencer, C. N., Reuben, A. & Wargo, J. A. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell 33, 570–580 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  233. McQuade, J. L., Daniel, C. R., Helmink, B. A. & Wargo, J. A. Modulating the microbiome to improve therapeutic response in cancer. Lancet. Oncol. 20, e77–e91 (2019).

    PubMed  Google Scholar 

  234. Day, D. & Siu, L. L. Approaches to modernize the combination drug development paradigm. Genome Med. 8, 115 (2016).

    PubMed  PubMed Central  Google Scholar 

  235. [No authors listed] Rationalizing combination therapies. Nat. Med. 23, 1113 (2017).

  236. Pastoriza, J. M. et al. Black race and distant recurrence after neoadjuvant or adjuvant chemotherapy in breast cancer. Clin. Exp. Metastasis 35, 613–623 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Martin, D. N. et al. Differences in the tumor microenvironment between African-American and European-American breast cancer patients. PLOS ONE 4, e4531 (2009).

    PubMed  PubMed Central  Google Scholar 

  238. Koru-Sengul, T. et al. Breast cancers from black women exhibit higher numbers of immunosuppressive macrophages with proliferative activity and of crown-like structures associated with lower survival compared to non-black Latinas and Caucasians. Breast Cancer Res. Treat 158, 113–126 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Casares, N. et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 202, 1691–1701 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Liu, W. M., Fowler, D. W., Smith, P. & Dalgleish, A. G. Pre-treatment with chemotherapy can enhance the antigenicity and immunogenicity of tumours by promoting adaptive immune responses. Br. J. Cancer 102, 115–123 (2010).

    CAS  PubMed  Google Scholar 

  241. Suzuki, E., Kapoor, V., Jassar, A. S., Kaiser, L. R. & Albelda, S. M. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin. Cancer Res. 11, 6713–6721 (2005).

    CAS  PubMed  Google Scholar 

  242. Reynders, K., Illidge, T., Siva, S., Chang, J. Y. & De Ruysscher, D. The abscopal effect of local radiotherapy: using immunotherapy to make a rare event clinically relevant. Cancer Treat Rev. 41, 503–510 (2015). This review focuses on the combination of immunotherapy and radiotherapy. The review raises the hypothesis that the abscopal effect occurring in a subset of patients who undergo radiotherapy can be enhanced with ICIs.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The author thanks the referees of this manuscript who provided excellent comments that substantially improved the Review. The author also thanks R. Kerbel, J. Condeelis and M. Oktay for their comments and suggestions throughout the writing process. It is with regret that not all relevant studies could be cited due to space limitation. This Review and many of the original studies published by the author on this specific subject have been supported by European Research Council (ERC) grants (current 771112).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yuval Shaked.

Ethics declarations

Competing interest

The author is a co-founder of OncoHost, a biotechnology company that utilizes the host response profile in the clinical setting in order to improve anticancer therapies. The author is listed on several patents related to host responses to anticancer drugs.

Additional information

Peer review information

Nature Reviews Cancer thanks M. De Palma 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

Tumour-associated macrophages

(TAMs). A heterogeneous population of differentiated monocytes found in the microenvironment of solid tumours, mainly consisting of cells with immunosuppressive functions (also known as M2-like macrophages) as well as a minor population of cells with pro-inflammatory functions (also known as M1-like macrophages).

ATP-binding cassette (ABC) transporters

Transporter proteins composed of transmembrane and ATPase protein subunits that take up or export various substrates across the cell membrane. In cancer cells, they contribute to therapy resistance via their ability to expel drugs from the cell.

Tubulin-binding agents

A family of anti-neoplastic chemotherapeutic drugs that interfere with the depolarization or polymerization of microtubules required for cell mitosis. As the rapidly proliferating malignant cells cannot divide in the presence of tubulin-binding agents, they eventually undergo apoptosis.

Alkylating agents

A family of anti-neoplastic chemotherapeutic drugs that act to replace hydrogen with an alkyl group in the replicated DNA during cell proliferation. The alkyl group is attached to the guanine base of the DNA, thus causing DNA damage and, ultimately, cell apoptosis.

Anti-metabolite agents

A family of anti-neoplastic chemotherapeutic drugs that interfere with the metabolic pathway of DNA replication in S phase, ultimately inducing cell apoptosis. Among these agents are folic acid antagonists, and purine and pyrimidine anti-metabolites.

Myeloid cells

Leukocytes from the haematopoietic myeloid lineage. These are usually premature cells that differentiate into monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes and megakaryocytes.

Extracellular vesicles

(EVs). Lipid bilayer particles that contain DNA, RNA and proteins and that are released from the cell surface membrane. They vary in size (usually between 50 and 500 µm) and are categorized as exosomes, microparticles and apoptotic bodies.

Thymus-associated endothelial cells

Endothelial cells residing in the thymus that contribute to the regeneration of thymus tissue following radiation.

Bronchoalveolar lavage

An invasive medical procedure involving the instillation of saline solution into a subsegment of the lung area. The liquid is then retrieved by suction and analysed for cells, protein content and foreign materials.

Amphiregulin

A ligand of the epidermal growth factor receptor (EGFR) expressed by epithelial cells. Its signalling through EGFR contributes to the development and growth of normal epithelial cells. In cancer, it promotes tumour cell proliferation.

Epiregulin

A ligand of the epidermal growth factor receptor that contributes to inflammation, wound healing and tissue repair.

Lymphangiogenesis

The formation of new lymphatic vessels from pre-existing lymphatic vessels, in a process similar to angiogenesis. In cancer, lymphangiogenesis is associated with metastasis.

Vascular-disrupting agents

(VDAs). A family of drugs with potent tumour vessel disruption ability. These cytotoxic-like agents (usually tubulin-binding or tubulin-destabilizing agents) rapidly promote cancer vessel collapse leading to necrosis.

Haemangiocytes

Bone marrow-derived pro-angiogenic haematopoietic cells that were shown to contribute to tumour angiogenesis. They are usually localized in the perivascular zone of the growing blood vessel and contribute to its integrity.

Vasculogenesis

The process of systemic formation of blood vessels. As opposed to angiogenesis, which is a local process of blood vessel formation from pre-existing vessels, vasculogenesis is mediated by systemic mobilization of bone marrow-derived pro-angiogenic cells, such as endothelial precursor cells, which incorporate to generate blood vessels.

Fibrosis

The formation or thickening of connective tissue in response to injury or chronic inflammation.

Laparoscopy

A procedure in which a fibre-optic device is usually inserted into the abdomen in order to fulfil a surgical procedure. It is sometimes used instead of open surgery to minimize recovery time.

Tumour microenvironment of metastasis

(TMEM). An anatomical structure within solid tumours composed of a cancer cell and a perivascularly located macrophage expressing TIE2 and vascular endothelial growth factor (VEGF). When both of these cells tightly bind to a blood vessel endothelial cell, the secretion of local VEGF contributes to blood vessel permeability, opening a doorway through which tumour cells can extravasate and disseminate from the primary tumour site.

Pre-metastatic niche

A physical area in a secondary non-malignant site that provides a favourable environment for circulating tumour cells to seed and proliferate in order to establish a metastatic lesion.

Resolvins

Polyunsaturated fatty acid metabolites that, in response to injury and subsequent inflammation, resolve inflammation and help restore tissue homeostasis.

Desmoplastic tumours

Tumours containing a large amount of dense connective and fibrous tissue, such as pancreatic ductal adenocarcinoma.

Erythroleukaemia

A relatively rare haematological malignancy, classed as a subtype of acute myeloid leukaemia arising from malignant erythroid blasts. Owing to the lack of differentiated erythroid cells, the disease is characterized by anaemia, thrombocytopenia and leukopenia.

Epoxide hydrolase

A class of enzymes that metabolize epoxide residues into hydroxyls. There are several enzymes in this family, distinguished from each other by their preferred substrates. They are known to detoxify genotoxic compounds and play a role in physiological signalling.

Abscopal effect

A rare phenomenon in which local radiation causes tumour shrinkage not only at the irradiated tumour site but also at distant tumour sites (usually metastatic lesions) located outside the field of irradiation.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shaked, Y. The pro-tumorigenic host response to cancer therapies. Nat Rev Cancer 19, 667–685 (2019). https://doi.org/10.1038/s41568-019-0209-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-019-0209-6

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