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.

  • Primer
  • Published:

Bone metastases

Abstract

Bone is the most frequent site for metastasis for many cancers, notably for tumours originating in the breast and the prostate. Tumour cells can escape from the primary tumour site and colonize the bone microenvironment. Within the bone, these disseminated tumour cells, as well as those arising in the context of multiple myeloma, may assume a state of dormancy, remaining quiescent for years before resuming proliferation and causing overt metastasis, which causes bone destruction via activation of osteoclast-mediated osteolysis. This structural damage can lead to considerable morbidity, including pain, fractures and impaired quality of life. Although treatment of bone metastases and myeloma bone disease is rarely curative, disease control is often possible for many years through the use of systemic anticancer treatments on a background of multidisciplinary supportive care. This care should include bone-targeted agents to inhibit tumour-associated osteolysis and prevent skeletal morbidity as well as use of appropriate local treatments such as radiation therapy, orthopaedic surgery and specialist palliative care to minimize the impact of metastatic bone disease on physical functioning. In this Primer, we provide an overview of the clinical features, the pathophysiology and the specific treatment approaches to prevent and treat bone metastases from solid tumours as well as myeloma bone disease.

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: Metastatic invasion of cancer cells to the bone.
Fig. 2: Physiological bone remodelling.
Fig. 3: Osteolytic bone metastases.
Fig. 4: Osteoblastic bone metastases.
Fig. 5: Myeloma bone disease.
Fig. 6: Relationships between imaging technologies and cellular biology of bone metastasis.
Fig. 7: Comparison of imaging techniques used for the diagnosis of bone metastases.
Fig. 8: Schematic algorithm for multidisciplinary treatment of bone metastases and myeloma bone disease.
Fig. 9: Bone metastasis response assessments on whole-body MRI.

Similar content being viewed by others

References

  1. Coleman R. E., Brown J. E., Holen I. in Clinical Oncology 6th edn Ch 51 (eds Niederhuber J. E., Armitage J. O., Doroshaw J. H., Kastan M. B. & Tepper J. E.) 809–830 (Elsevier, Churchill Livingstone, 2019).

  2. von Moos, R. et al. Management of bone health in solid tumours: from bisphosphonates to a monoclonal antibody. Cancer Treat. Rev. 76, 57–67 (2019).

    Google Scholar 

  3. Coleman, R. et al. Bone health in cancer: ESMO clinical practice guidelines. Ann. Oncol. https://doi.org/10.1093/annonc/mdu103 (2020). This paper describes the comprehensive current guidelines for diagnosis, prevention and treatment of bone metastases and muliple myeloma.

  4. Coleman, R. E. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin. Cancer Res. 12, 6243s–6249s (2006).

    PubMed  Google Scholar 

  5. D’Oronzo, S., Coleman, R., Brown, J. & Silvestris, F. Metastatic bone disease: pathogenesis and therapeutic options: up-date on bone metastasis management. J. Bone Oncol. 15, 004–004 (2018).

    PubMed  PubMed Central  Google Scholar 

  6. Bray, F. et al. Global cancer statistics 2018: GLOBOSCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68, 394–424 (2018).

    PubMed  Google Scholar 

  7. Boxer, D. I., Todd, C. E. C., Coleman, R. & Fogelman, I. Bone secondaries in breast cancer: the solitary metastasis. J. Nucl. Med. 30, 1318–1320 (1989).

    CAS  PubMed  Google Scholar 

  8. Hortobagyi, G. N. et al. Efficacy of pamidronate in reducing skeletal complications in patients with breast cancer and lytic bone metastases. N. Engl. J. Med. 335, 1785–1792 (1996). This study is the first RCT to clearly establish the impact of a bisphosphonate on skeletal morbidity from bone metastases.

    CAS  PubMed  Google Scholar 

  9. Berenson, J. R. et al. Long-term pamidronate treatment of advanced multiple myeloma patients reduces skeletal events. Myeloma Aredia Study Group. J. Clin. Oncol. 16, 593–602 (1998).

    CAS  PubMed  Google Scholar 

  10. Saad, F. et al. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. JNCI 94, 1458–1468 (2002).

    CAS  PubMed  Google Scholar 

  11. Rosen, L. S. et al. Long-term efficacy and safety of zoledronic acid in the treatment of skeletal metastases in patients with nonsmall cell lung carcinoma and other solid tumors. Cancer 100, 2613–2621 (2004).

    CAS  PubMed  Google Scholar 

  12. Oster, G. et al. Natural history of skeletal-related events in patients with breast, lung, or prostate cancer and metastases to bone: a 15-year study in two large US health systems. Support. Care Cancer 21, 3279–3286 (2013).

    PubMed  Google Scholar 

  13. Morrissey, C. et al. Effects of androgen deprivation therapy and bisphosphonate treatment on bone in patients with metastatic castration-resistant prostate cancer: results from the University of Washington Rapid Autopsy Series. J. Bone Min. Res. 28, 333–340 (2013).

    CAS  Google Scholar 

  14. Brown, J. E. et al. Bone turnover markers as predictors of skeletal complications in prostate cancer, lung cancer, and other solid tumors. J. Natl Cancer Inst. 97, 59–69 (2005).

    CAS  PubMed  Google Scholar 

  15. Herbst, R. S., Morgensztern, D. & Boshoff, C. The biology and management of non-small cell lung cancer. Nature. 553, 446–454 (2018).

    CAS  PubMed  Google Scholar 

  16. Melton, L. J. III et al. Fracture risk with multiple myeloma: a population-based study. J. Bone Min. Res. 20, 487–493 (2005).

    Google Scholar 

  17. Adamik, J., Galson, D. L. & Roodman, G. D. Osteoblast suppression in multiple myeloma bone disease. J. Bone Oncol. 13, 62–70 (2018).

    PubMed  PubMed Central  Google Scholar 

  18. Melton, L. J. et al. Fracture risk in monoclonal gammopathy of undetermined significance. J. Bone Miner. Res. 19, 25–30 (2004).

    PubMed  Google Scholar 

  19. Fornetti, J., Welm, A. L. & Stewart, S. A. Understanding the bone in cancer metastasis. J. Bone Min. Res. 33, 2099–2113 (2018).

    CAS  Google Scholar 

  20. Seeman, E. & Delmas, P. D. Bone quality — the material and structural basis of bone strength and fragility. N. Engl. J. Med. 354, 2250–2261 (2006).

    CAS  PubMed  Google Scholar 

  21. Sims, N. A. & Martin, T. J. Coupling the activities of bone formation and resorption: a multitude of signals within the basic multicellular unit. Bonekey Rep. 3, 481 (2014).

    PubMed  PubMed Central  Google Scholar 

  22. Hofbauer, L. C., Rachner, T. D., Coleman, R. E. & Jakob, F. Endocrine aspects of bone metastases. Lancet Diabetes Endocrinol. 2, 500–512 (2014).

    CAS  PubMed  Google Scholar 

  23. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 133, 571–573 (1889).

    Google Scholar 

  24. Joeckel, E. et al. High calcium concentration in bones promotes bone metastasis in renal cell carcinomas expressing calcium-sensing receptor. Mol. Cancer 13, 42 (2014).

    PubMed  PubMed Central  Google Scholar 

  25. Yin, J. J. et al. TGF-β signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J. Clin. Invest. 103, 197–206 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Brylka, L. & Schinke, T. Chemokines in physiological and pathological bone remodelling. Front. Immunol. 10, 2182 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Hosseini, H. et al. Early dissemination seeds metastasis in breast cancer. Nature 540, 555–558 (2016).

    Google Scholar 

  28. Mishra, A., Shiozawa, Y., Pienta, K. J. & Taichman, R. S. Homing of cancer cells to the bone. Cancer Microenviron. 4, 221–235 (2011).

    PubMed  PubMed Central  Google Scholar 

  29. Zhang, X. H. et al. Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma. Cell 154, 1060–1073 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Cox, T. R. et al. The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 522, 106–110 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zoni, E. & van der Pluijm, G. The role of microRNAs in bone metastasis. J. Bone Oncol. 5, 104–108 (2016).

    PubMed  PubMed Central  Google Scholar 

  32. Ell, B. et al. Tumor-induced osteoclast miRNA changes as regulators and biomarkers of osteolytic bone metastasis. Cancer Cell 24, 542–556 (2013).

    CAS  PubMed  Google Scholar 

  33. Woodward, J. K. L., Holen, I., Coleman, R. E. & Buttle, D. J. The roles of proteolytic enzymes in the development of tumour-induced bone disease in breast and prostate cancer. Bone 41, 912–927 (2007).

    CAS  PubMed  Google Scholar 

  34. Casimiro, S. et al. RANKL/RANK/MMP-1 molecular triad contributes to the metastatic phenotype of breast and prostate cancer cells in vitro. PLoS ONE 8, e63153 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Weilbaecher, K. N., Guise, T. A. & McCauley, L. K. Cancer to bone: a fatal attraction. Nat. Rev. Cancer 11, 411–425 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Braun, S. et al. A pooled analysis of bone marrow micrometastasis in breast cancer. N. Engl. J. Med. 353, 793–802 (2005).

    CAS  PubMed  Google Scholar 

  37. 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 

  38. Croucher, P. I., McDonald, M. M. & Martin, T. J. Bone metastasis: the importance of the neighbourhood. Nat. Rev. Cancer 16, 373–386 (2016). This paper presents a comprehensive review of our understanding of the mechanisms of metastasis to bone.

    CAS  PubMed  Google Scholar 

  39. Phan, T. G. & Croucher, P. I. The dormant cancer cell life cycle. Nat. Rev. Cancer 20, 398–411 (2020).

    CAS  PubMed  Google Scholar 

  40. Yin, J. J. et al. A causal role for endothelin-1 in the pathogenesis of osteoblastic bone metastases. Proc. Natl Acad. Sci. USA 100, 10954–10959 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Nelson, J. B. et al. Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat. Med. 1, 944–949 (1995).

    CAS  PubMed  Google Scholar 

  42. Liu, A. Y., Roudier, M. P. & True, L. D. Heterogeneity in primary and metastatic prostate cancer as defined by cell surface CD profile. Am. J. Pathol. 165, 1543–1556 (2004).

    PubMed  PubMed Central  Google Scholar 

  43. Roudier, M. P. et al. Histological, immunophenotypic and histomorphometric characterization of prostate cancer bone metastases. Cancer Treat. Res. 118, 311–339 (2004).

    PubMed  Google Scholar 

  44. Guise, T. A. et al. Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J. Clin. Invest. 98, 1544–1549 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003).

    CAS  PubMed  Google Scholar 

  46. Sethi, N., Dai, X., Winter, C. G. & Kang, Y. Tumor-derived JAGGED1 promotes osteolytic bone metastasis of breast cancer by engaging notch signaling in bone cells. Cancer Cell 19, 192–205 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Thomas, R. J. et al. Breast cancer cells interact with osteoblasts to support osteoclast formation. Endocrinology 140, 4451–4458 (1999).

    CAS  PubMed  Google Scholar 

  48. Teicher, B. A. & Fricker, S. P. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin. Cancer Res. 16, 2927–2931 (2010).

    CAS  PubMed  Google Scholar 

  49. Sethi, N. & Kang, Y. Unravelling the complexity of metastasis—molecular understanding and targeted therapies. Nat. Rev. Cancer 11, 735–748 (2011).

    CAS  PubMed  Google Scholar 

  50. Wang, H. et al. The osteogenic niche is a calcium reservoir of bone micrometastases and confers unexpected therapeutic vulnerability. Cancer Cell 34, 823–839.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Wang, H. et al. The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell 27, 193–210 (2015).

    PubMed  PubMed Central  Google Scholar 

  52. Eyre, R. et al. Microenvironmental IL1β promotes breast cancer metastatic colonisation in the bone via activation of Wnt signalling. Nat. Commun. 10, 5016 (2019).

    PubMed  PubMed Central  Google Scholar 

  53. Lara-Castillo, N. & Johnson, M. L. Bone–muscle mutual interactions. Curr. Osteoporos. Rep. 18, 408–421 (2020).

    PubMed  PubMed Central  Google Scholar 

  54. Hesse, E. et al. Sclerostin inhibition alleviates breast cancer-induced bone metastases and muscle weakness. JCI Insight 4, e125543 (2019).

    PubMed Central  Google Scholar 

  55. Ottewell, P. D. et al. Zoledronic acid has differential antitumor activity in the pre- and postmenopausal bone microenvironment in vivo. Clin. Cancer Res. 20, 2922–2932 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wright, L. E. et al. Aromatase inhibitor-induced bone loss increases the progression of estrogen receptor-negative breast cancer in bone and exacerbates muscle weakness in vivo. Oncotarget 8, 8406–8419 (2017).

    PubMed  Google Scholar 

  57. Campbell, J. P. et al. Stimulation of host bone marrow stromal cells by sympathetic nerves promotes breast cancer bone metastasis in mice. PLoS Biol. 10, e1001363 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Lang, J., Zhao, Q., He, Y. & Yu, X. Bone turnover markers and novel biomarkers in lung cancer bone metastases. Biomarkers 23, 518–526 (2018).

    CAS  PubMed  Google Scholar 

  59. Vicent, S. et al. Novel lung cancer signature mediates metastatic bone colonization by a dual mechanism. Cancer Res. 68, 2275–2285 (2008).

    CAS  PubMed  Google Scholar 

  60. Luis-Ravelo, D. et al. A gene signature of bone metastatic colonization sensitizes for tumor-induced osteolysis and predicts survival in lung cancer. Oncogene 33, 5090–5099 (2014).

    CAS  PubMed  Google Scholar 

  61. Johnson, R. W. et al. Wnt signaling induces gene expression of factors associated with bone destruction in lung and breast cancer. Clin. Exp. Metastasis 31, 945–959 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Chu, T., Teng, J., Jiang, L., Zhong, H. & Han, B. Lung cancer-derived Dickkopf1 is associated with bone metastasis and the mechanism involves the inhibition of osteoblast differentiation. Biochem. Biophys. Res. Commun. 443, 962–968 (2014).

    CAS  PubMed  Google Scholar 

  63. Pang, H. et al. The biological effects of Dickkopf1 on small cell lung cancer cells and bone metastasis. Oncol. Res. 25, 35–42 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. Hernández, I., Moreno, J. L., Zandueta, C., Montuenga, L. & Lecanda, F. Novel alternatively spliced ADAM8 isoforms contribute to the aggressive bone metastatic phenotype of lung cancer. Oncogene 29, 3758–3769 (2010).

    PubMed  Google Scholar 

  65. Chen, W. G. et al. Sema4D expression and secretion are increased by HIF-1α and inhibit osteogenesis in bone metastases of lung cancer. Clin. Exp. Metastasis 36, 39–56 (2019).

    CAS  PubMed  Google Scholar 

  66. Brunetti, G. et al. LIGHT/TNFSF14 promotes osteolytic bone metastases in non-small cell lung cancer patients. J. Bone Min. Res. 35, 671–680 (2020).

    CAS  Google Scholar 

  67. Miller, R. E., Jones, J. C., Tometsko, M., Blake, M. L. & Dougall, W. C. RANKL inhibition blocks osteolytic lesions and reduces skeletal tumor burden in models of non-small-cell lung cancer bone metastases. J. Thorac. Oncol. 9, 345–354 (2014).

    CAS  PubMed  Google Scholar 

  68. Futamura, N. et al. Hyaluronan synthesis inhibitor supplements the inhibitory effects of zoledronic acid on bone metastasis of lung cancer. Clin. Exp. Metastasis 30, 595–606 (2013).

    CAS  PubMed  Google Scholar 

  69. Ibrahim, T. et al. Pathogenesis of osteoblastic bone metastases from prostate cancer. Cancer 116, 1406–1418 (2010).

    CAS  PubMed  Google Scholar 

  70. Lin, S. C., Yu-Lee, L. Y. & Lin, S. H. Osteoblastic factors in prostate cancer bone metastasis. Curr. Osteoporos. Rep. 16, 642–647 (2018).

    PubMed  PubMed Central  Google Scholar 

  71. Cher, M. L. et al. Cancer interaction with the bone microenvironment: a workshop of the National Institutes of Health Tumor Microenvironment Study Section. Am. J. Pathol. 168, 1405–1412 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Lin, S.-C. et al. Endothelial-to-osteoblast conversion generates osteoblastic metastasis of prostate cancer. Dev. Cell 41, 467–480.e3 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Wang, S. et al. FOXF2 reprograms breast cancer cells into bone metastasis seeds. Nat. Commun. 10, 2707 (2019).

    PubMed  PubMed Central  Google Scholar 

  74. Wan, X. et al. Prostate cancer cell–stromal cell cross-talk via FGFR1 mediates antitumor activity of dovitinib in bone metastases. Sci. Transl. Med. 6, 252ra122 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. Wang, W. et al. Prostate cancer promotes a vicious cycle of bone metastasis progression through inducing osteocytes to secrete GDF15 that stimulates prostate cancer growth and invasion. Oncogene 38, 4540–4559 (2019 Jun).

    CAS  PubMed  Google Scholar 

  76. Sottnik, J. L., Dai, J., Zhang, H., Campbell, B. & Keller, E. T. Tumor-induced pressure in the bone microenvironment causes osteocytes to promote the growth of prostate cancer bone metastases. Cancer Res. 75, 2151–2158 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Shiozawa, Y., Pienta, K. J. & Taichman, R. S. Hematopoietic stem cell niche is a potential therapeutic target for bone metastatic tumors. Clin. Cancer Res. 17, 5553–5558 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Owen, K. L. et al. Prostate cancer cell-intrinsic interferon signaling regulates dormancy and metastatic outgrowth in bone. EMBO Rep. 21, e50162 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Dudley, A. C. et al. Calcification of multi-potent, prostate tumor endothelium cancer cell. Cancer Cell 14, 201–211 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chen, Z., Orlowski, R. Z., Wang, M., Kwak, L. & McCarty, N. Osteoblastic niche supports the growth of quiescent multiple myeloma cells. Blood. 123, 2204–2208 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Lawson, M. A. et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat. Commun. 6, 8983 (2015). This important study demonstrates some of the processes by which multiple myeloma develops and affects the skeleton.

    CAS  PubMed  Google Scholar 

  82. Khoo, W. H. et al. A niche-dependent myeloid transcriptome signature defines dormant myeloma cells. Blood. 134, 30–43 (2019).

    CAS  PubMed  Google Scholar 

  83. Croucher, P. I. et al. Osteoprotegerin inhibits the development of osteolytic bone disease in multiple myeloma. Blood 98, 3534–3540 (2001).

    CAS  PubMed  Google Scholar 

  84. Pearse, R. N. et al. Multiple myeloma disrupts the TRANCE/osteoprotegerin cytokine axis to trigger bone destruction and promote tumor progression. Proc. Natl Acad. Sci. USA 98, 11581 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Giuliani, N. et al. Human myeloma cells stimulate the receptor activator of nuclear factor-κB ligand (RANKL) in T lymphocytes: a potential role in multiple myeloma bone disease. Blood 100, 4615–4621 (2002).

    CAS  PubMed  Google Scholar 

  86. Heider, U. et al. Expression of receptor activator of nuclear factor κB ligand on bone marrow plasma cells correlates with osteolytic bone disease in patients with multiple myeloma. Clin. Cancer Res. 9, 1436–1440 (2003).

    CAS  PubMed  Google Scholar 

  87. Terpos, E. et al. Soluble receptor activator of nuclear factor κB ligand–osteoprotegerin ratio predicts survival in multiple myeloma: proposal for a novel prognostic index. Blood 102, 1064–1069 (2003).

    CAS  PubMed  Google Scholar 

  88. Yaccoby, S. et al. Myeloma interacts with the bone marrow microenvironment to induce osteoclastogenesis and is dependent on osteoclast activity. Br. J. Haematol. 116, 278–290 (2002).

    PubMed  Google Scholar 

  89. Hoi, S. J. et al. Macrophage inflammatory protein 1α is a potential osteoclast stimulatory factor in multiple myeloma. Blood 96, 671–675 (2000).

    Google Scholar 

  90. Oyajobi, B. O. et al. Dual effects of macrophage inflammatory protein-1α on osteolysis and tumor burden in the murine 5TGM1 model of myeloma bone disease. Blood 102, 311–319 (2003).

    CAS  PubMed  Google Scholar 

  91. Hjertner, O. et al. Hepatocyte growth factor (HGF) induces interleukin-11 secretion from osteoblasts: a possible role for HGF in myeloma-associated osteolytic bone disease. Blood 94, 3883–3888 (1999).

    CAS  PubMed  Google Scholar 

  92. Marino, S. & Roodman, G. D. Multiple myeloma and bone: the fatal interaction. Cold Spring Harb. Perspect. Med. 8, a031286 (2018).

    PubMed  PubMed Central  Google Scholar 

  93. Tian, E. et al. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N. Engl. J. Med. 349, 2483–2494 (2003).

    CAS  PubMed  Google Scholar 

  94. Giuliani, N. et al. Production of Wnt inhibitors by myeloma cells: potential effects on canonical Wnt pathway in the bone microenvironment. Cancer Res. 67, 7665–7674 (2007).

    CAS  PubMed  Google Scholar 

  95. Heath, D. J. et al. Inhibiting Dickkopf-1 (Dkk1) removes suppression of bone formation and prevents the development of osteolytic bone disease in multiple myeloma. J. Bone Miner. Res. 24, 425–436 (2009).

    CAS  PubMed  Google Scholar 

  96. Yaccoby, S. et al. Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood 109, 2106–2111 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Politou, M. C. et al. Serum concentrations of Dickkopf-1 protein are increased in patients with multiple myeloma and reduced after autologous stem cell transplantation. Int. J. Cancer 119, 1728–1731 (2006).

    CAS  PubMed  Google Scholar 

  98. Delgado-Calle, J. et al. Bidirectional Notch signaling and osteocyte-derived factors in the bone marrow microenvironment promote tumor cell proliferation and bone destruction in multiple myeloma. Cancer Res. 76, 1089–1100 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Delgado-Calle, J. et al. Genetic deletion of Sost or pharmacological inhibition of sclerostin prevent multiple myeloma-induced bone disease without affecting tumor growth. Leukemia 31, 2686–2694 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Eda, H., Santo, L. & Wein, M. N. Regulation of sclerostin expression in multiple myeloma by Dkk-1: a potential therapeutic strategy for myeloma bone disease. J. Bone Min. Res. 31, 1225–1234 (2016).

    CAS  Google Scholar 

  101. Paton-Hough, J. et al. Preventing and repairing myeloma bone disease by combining conventional antiresorptive treatment with a bone anabolic agent in murine models. J. Bone Miner. Res. 34, 783–796 (2019).

    CAS  PubMed  Google Scholar 

  102. McDonald, M. M. et al. Inhibiting the osteocyte-specific protein sclerostin increases bone mass and fracture resistance in multiple myeloma. Blood 129, 3452–3464 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Vallet, S. et al. Activin A promotes multiple myeloma-induced osteolysis and is a promising target for myeloma bone disease. Proc. Natl Acad. Sci. USA 107, 5124–5129 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Shen, G., Deng, H., Hu, S. & Jia, Z. Comparison of choline-PET/CT, MRI, SPECT, and bone scintigraphy in the diagnosis of bone metastases in patients with prostate cancer: a meta-analysis. Skelet. Radiol. 4, 1503–1513 (2014).

    Google Scholar 

  105. Yang, H.-L., Liu, T., Wang, X.-M., Xu, Y. & Deng, S.-M. Diagnosis of bone metastases: a meta-analysis comparing 18FDG PET, CT, MRI and bone scintigraphy. Eur. Radiol. 21, 2604–2617 (2011).

    PubMed  Google Scholar 

  106. Ost, P. et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: a prospective, randomized, multicenter phase II trial. J. Clin. Oncol. 36, 446–453 (2018).

    CAS  PubMed  Google Scholar 

  107. Mottet N. et al. 2019 EAU–EANM–ESTRO–ESUR–SIOG guidelines on prostate cancer Vol. 53. EAU https://uroweb.org/guideline/prostate-cancer/#3 (2019).

  108. Crawford, E. D. et al. A clinician’s guide to next generation imaging in patients with advanced prostate cancer (RADAR III). J. Urol. 201, 682–692 (2019).

    PubMed  Google Scholar 

  109. Rajkumar, S. V. Updated diagnostic criteria and staging system for multiple myeloma. Am. Soc. Clin. Oncol. Educ. Book 35, e418–e423 (2016).

    PubMed  Google Scholar 

  110. Tateishi, U. et al. A meta-analysis of 18F-fluoride positron emission tomography for assessment of metastatic bone tumor. Ann. Nucl. Med. 24, 523–531 (2010).

    PubMed  Google Scholar 

  111. von Eyben, F. E. & Kairemo, K. Meta-analysis of 11C-choline and 18F-choline PET/CT for management of patients with prostate. Nucl. Med. Commun. 35, 221–230 (2014).

    Google Scholar 

  112. Wondergem, M., van der Zant, F. M., van der Ploeg, T. & Knol, R. J. J. A literature review of 18F-fluoride PET/CT and 18F-choline or 11C-choline PET/CT for detection of bone metastases in patients with prostate cancer. Nucl. Med. Commun. 34, 935–945 (2013).

    CAS  PubMed  Google Scholar 

  113. Perera, M. et al. Gallium-68 prostate-specific membrane antigen positron emission tomography in advanced prostate cancer—updated diagnostic utility, sensitivity, specificity, dnd Distribution of prostate-specific membrane antigen-avid lesions: a systematic review and meta-analysis. Eur Urol. 77, 403–417 (2019).

    PubMed  Google Scholar 

  114. Calais, J. et al. 18F-fluciclovine PET-CT and 68Ga-PSMA-11 PET-CT in patients with early biochemical recurrence after prostatectomy: a prospective, single-centre, single-arm, comparative imaging trial. Lancet Oncol. 2045, 1–9 (2019).

    Google Scholar 

  115. Jambor, I. et al. Prospective evaluation of planar bone scintigraphy, SPECT, SPECT/CT, 18F-NaF PET/CT and whole body 1.5 T MRI, including DWI, for the detection of bone metastases in high risk breast and prostate cancer patients: SKELETA clinical trial. Acta Oncol. 55, 59–67 (2016).

    PubMed  Google Scholar 

  116. Johnston, E. W. et al. Multiparametric whole-body 3.0-T MRI in newly diagnosed intermediate- and high-risk prostate cancer: diagnostic accuracy and interobserver agreement for nodal and metastatic staging. Eur. Radiol. 29, 3159–3169 (2019).

    PubMed  Google Scholar 

  117. Coleman, R. et al. Consensus on the utility of bone markers in the malignant bone disease setting. Crit. Rev. Oncol. Hematol. 80, 411–432 (2011).

    PubMed  Google Scholar 

  118. Coleman, R. E., Fogelman, I., Habibollahi, F., North, W. R. S. & Rubens, R. D. Selection of patients with breast cancer for routine follow-up bone scans. Clin. Oncol. 2, 328–332 (1990).

    CAS  Google Scholar 

  119. Mateo, J. et al. Managing nonmetastatic castration-resistant prostate cancer. Eur. Urol. 75, 285–293 (2019).

    PubMed  Google Scholar 

  120. Wilson, C. & Coleman, R. E. Adjuvant bone targeted therapies for postmenopausal breast cancer. JAMA Oncol. 2, 423–424 (2016).

    PubMed  Google Scholar 

  121. Coleman R. E. Clinical benefits of bone targeted agents in early breast cancer. Breast 48 (Suppl. 1), 92–96 (2019).

    Google Scholar 

  122. Gnant, M. et al. Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 62-month follow-up from the ABCSG-12 randomised trial. Lancet Oncol. 12, 631–641 (2011).

    CAS  PubMed  Google Scholar 

  123. Coleman, R. E. et al. Breast cancer adjuvant therapy with zoledronic acid. N. Engl. J. Med. 365, 1396–1405 (2011).

    CAS  PubMed  Google Scholar 

  124. Coleman, R. E. et al. Benefits and risks of adjuvant treatment with zoledronic acid in stage II/III breast cancer. 10 years follow-up of the AZURE randomized clinical trial (BIG 01/04). J. Bone Oncol. 13, 123–135 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Early Breast Cancer Trialists’ Collaborative Group. Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet 386, 1353–1361 (2015). This paper presents an individual patient meta-analysis of data from >18,000 women included in RCTs of adjuvant bisphosphonates in early breast cancer that demonstrated the reduction in bone metastases and breast cancer deaths in postmenopausal women.

    Google Scholar 

  126. Gralow J. R. et al. Phase III randomized trial of bisphosphonates as adjuvant therapy in breast cancer: S0307. J. Natl Cancer Inst. 112, 698–707 (2019).

    PubMed Central  Google Scholar 

  127. Gnant, M. et al. Adjuvant denosumab in postmenopausal patients with hormone receptor-positive breast cancer (ABCSG-18): disease-free survival results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 20, 339–351 (2019).

    CAS  PubMed  Google Scholar 

  128. Coleman, R. E. et al. Adjuvant denosumab in early breast cancer: primary results from the international, multicenter, randomized, phase 3, placebo-controlled D-CARE study. Lancet Oncol. 21, 60–72 (2020).

    CAS  PubMed  Google Scholar 

  129. Wirth, M. et al. Prevention of bone metastases in patients with high-risk nonmetastatic prostate cancer treated with zoledronic acid: efficacy and safety results of the Zometa European Study (ZEUS). Eur. Urol. 67, 482–491 (2015).

    CAS  PubMed  Google Scholar 

  130. Smith, M. R. et al. Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial. Lancet 379, 39–46 (2012).

    CAS  PubMed  Google Scholar 

  131. Scagliotti, G. V. et al. Zoledronic acid in patients with stage IIIA/B NSCLC: results of a randomized, phase III study. Ann. Oncol. 23, 2082–2087 (2012).

    CAS  PubMed  Google Scholar 

  132. Peters, S. et al. A randomised phase III trial evaluating the addition of denosumab to standard first-line treatment in advanced NSCLC—the ETOP and EORTC SPLENDOUR trial. Ann. Oncol. 29, viii493–viii547 (2018).

    Google Scholar 

  133. Coleman, R. E. et al. Bone scan flare predicts successful systemic therapy for bone metastases. J. Nucl. Med. 29, 1354–1359 (1988).

    CAS  PubMed  Google Scholar 

  134. Scher, H. I. et al. Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the Prostate Cancer Clinical Trials Working Group 3. J. Clin. Oncol. 34, 1–38 (2016).

    Google Scholar 

  135. Morris, M. J. et al. Radiographic progression-free survival as a response biomarker in metastatic castration-resistant prostate cancer: COU-AA-302 results. J. Clin. Oncol. 33, 1356–1363 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Armstrong, A. J. et al. Association between new unconfirmed bone lesions and outcomes in men with metastatic castration-resistant prostate cancer treated with enzalutamide: secondary analysis of the PREVAIL and AFFIRMvrandomized clinical trials. JAMA Oncol. 6, 217–225 (2019).

    PubMed Central  Google Scholar 

  137. Kumar, S. et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 17, e328–e346 (2016).

    PubMed  Google Scholar 

  138. Song, H., Jin, S., Xiang, P., Hu, S. & Jin, J. Prognostic value of the bone scan index in patients with metastatic castration-resistant prostate cancer: a systematic review and meta-analysis. BMC Cancer 20, 238 (2020).

    PubMed  PubMed Central  Google Scholar 

  139. Keller, L. & Pantel, K. Unraveling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Rev. Cancer 19, 553–567 (2019).

    CAS  PubMed  Google Scholar 

  140. Siravegna, G., Marsoni, S., Siena, S. & Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 14, 531–548 (2017).

    CAS  PubMed  Google Scholar 

  141. Wahl, R. L., Jacene, H., Kasamon, Y. & Lodge, M. A. From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J. Nucl. Med. 50, 122S–150S (2009).

    CAS  PubMed  Google Scholar 

  142. Fanti, S. et al. Development of standardized image interpretation for 68Ga-PSMA PET/CT to detect prostate cancer recurrent lesions. Eur. J. Nucl. Med. Mol. Imaging 44, 1622–1635 (2017).

    CAS  PubMed  Google Scholar 

  143. Padhani, A. R. et al. METastasis Reporting and Data System for Prostate Cancer: practical guidelines for acquisition, interpretation, and reporting of whole-body magnetic resonance imaging-based evaluations of multiorgan involvement in advanced prostate cancer. Eur. Urol. 71, 81–92 (2017).

    PubMed  PubMed Central  Google Scholar 

  144. Patrick, D. L. et al. Pain outcomes in patients with bone metastases from advanced cancer: assessment and management with bone-targeting agents. Support. Care Cancer 23, 1157–1168 (2015).

    PubMed  Google Scholar 

  145. von Moos, R. et al. Pain and analgesic use associated with skeletal-related events in patients with advanced cancer and bone metastases. Support. Care Cancer 24, 1327–1337 (2016).

    Google Scholar 

  146. Roelofs, A. J., Thompson, K., Gordon, S. & Rogers, M. J. Molecular mechanisms of action of bisphosphonates: current status. Clin. Cancer Res. 12, 6222s–6230s (2006).

    CAS  PubMed  Google Scholar 

  147. Body, J. J. et al. Oral ibandronate reduces the risk of skeletal complications in breast cancer patients with metastatic bone disease: results from two randomised, placebo-controlled phase III studies. Br. J. Cancer 90, 1133–1137 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Paterson, A. H. et al. Double-blind controlled trial of oral clodronate in patients with bone metastases from breast cancer. J. Clin. Oncol. 11, 59–65 (1993).

    CAS  PubMed  Google Scholar 

  149. Morgan, G. J. et al. Long-term follow-up of MRC Myeloma IX Trial: survival outcomes with bisphosphonate and thalidomide treatment. Clin. Cancer Res. 19, 6030–6038 (2013).

    CAS  PubMed  Google Scholar 

  150. Todenhöfer, T., Stenzl, A., Hofbauer, L. C. & Rachner, T. D. Targeting bone metabolism in patients with advanced prostate cancer: current options and controversies. Int. J. Endocrinol. 2015, 838202 (2015).

    PubMed  PubMed Central  Google Scholar 

  151. Boyle, W. J., Simonet, W. S. & Lacey, D. L. Osteoclast differentiation and activation. Nature 423, 337–342 (2003).

    CAS  PubMed  Google Scholar 

  152. Stopeck, A. T. et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J. Clin. Oncol. 28, 5132–5139 (2010). This study is the first RCT to demonstrate the role of denosumab in patients with bone metastases and superior efficacy compared with a bisphosphonate.

    CAS  PubMed  Google Scholar 

  153. Fizazi, K. et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet 377, 813–822 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Henry, D. H. et al. Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J. Clin. Oncol. 29, 1125–1132 (2011).

    CAS  PubMed  Google Scholar 

  155. Raje, N. et al. Denosumab versus zoledronic acid in bone disease treatment of newly diagnosed multiple myeloma: an international, double-blind, double-dummy, randomised, controlled, phase 3 study. Lancet Oncol. 19, 370–381 (2018).

    CAS  PubMed  Google Scholar 

  156. Yang, M. & Yu, X. Management of bone metastasis with intravenous bisphosphonates in breast cancer: a systematic review and meta-analysis of dosing frequency. Support. Care Cancer 28, 2533–2540 (2020).

    PubMed  Google Scholar 

  157. Tsourdi, E. et al. Discontinuation of denosumab therapy for osteoporosis: a systematic review and position statement by ECTS. Bone 105, 11–17 (2017).

    PubMed  Google Scholar 

  158. Lamy, O., Stoll, D., Aubry-Rozier, B. & Rodriguez, E. G. Stopping denosumab. Curr. Osteoporos. Rep. 17, 8–15 (2019).

    PubMed  Google Scholar 

  159. Body, J. J. Dosing regimens and main adverse events of bisphosphonates. Semin. Oncol. 28, 49–53 (2001).

    CAS  PubMed  Google Scholar 

  160. Body, J.-J. et al. Hypocalcaemia in patients with metastatic bone disease treated with denosumab. Eur. J. Cancer 51, 1812–1821 (2015).

    CAS  PubMed  Google Scholar 

  161. Saad, F. et al. Incidence, risk factors, and outcomes of osteonecrosis of the jaw: integrated analysis from three blinded active-controlled phase III trials in cancer patients with bone metastases. Ann. Oncol. 23, 1341–1347 (2012).

    CAS  PubMed  Google Scholar 

  162. Nicolatou-Galitis, O. et al. Medication-related osteonecrosis of the jaw: definition and best practice for prevention, diagnosis, and treatment. Oral. Surg. Oral Med. Oral Pathol. Oral Radiol. 127, 117–135 (2019).

    PubMed  Google Scholar 

  163. Nakata, E., Sugihara, S., Yamashita, N. & Osumi, S. The incidence of atypical femoral fractures in breast cancer patients with bone metastases who received bisphosphonate treatment. J. Orthop. Sci. 22, 946–950 (2017).

    PubMed  Google Scholar 

  164. Himelstein, A. L. et al. Effect of longer-interval vs standard dosing of zoledronic acid on skeletal events in patients with bone metastases. JAMA 317, 48 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Hortobagyi, G. N. Van et al. Continued treatment effect of zoledronic acid dosing every 12 vs 4 weeks in women with breast cancer metastatic to bone. JAMA Oncol. 3, 906 (2017).

    PubMed  PubMed Central  Google Scholar 

  166. Rich, S. et al. Update of the systematic review of palliative radiation therapy fractionation for bone metastases. Radiother. Oncol. 126, 547–557 (2018). This paper reviews palliative radiotherapy for bone metastases, focusing on trials assessing different fractionation regimens.

    PubMed  Google Scholar 

  167. Ruhlmann, C. H. et al. 2016 updated MASCC/ESMO consensus recommendations: prevention of radiotherapy-induced nausea and vomiting. Support. Care Cancer 25, 309–316 (2017).

    PubMed  Google Scholar 

  168. McDonald, R. et al. Effect of radiotherapy on painful bone metastases: a secondary analysis of the NCIC Clinical Trials Group Symptom Control Trial SC.23. JAMA Oncol. 3, 953–959 (2017).

    PubMed  PubMed Central  Google Scholar 

  169. Chow, E. et al. Update on the systematic review of palliative radiotherapy trials for bone metastases. Clin. Oncol. 24, 112–124 (2012).

    CAS  Google Scholar 

  170. Lutz, S. et al. Palliative radiotherapy for bone metastases: an ASTRO evidence-based guideline. Int. J. Radiat. Oncol. Biol. Phys. 79, 965–976 (2011).

    PubMed  Google Scholar 

  171. Hahn, C. et al. Choosing wisely: the American Society for Radiation Oncology’s top 5 lists. Pract. Radiat. Oncol. 4, 349–355 (2014).

    PubMed  Google Scholar 

  172. Chow, E. et al. Single versus multiple fractions of repeat radiation for painful bone metastases: a randomised, controlled, non-inferiority trial. Lancet Oncol. 15, 164–171 (2014).

    PubMed  Google Scholar 

  173. Chow, E. et al. Impact of reirradiation of painful osseous metastases on quality of life and function: a secondary analysis of the NCIC CTG SC.20 randomized trial. J. Clin. Oncol. 32, 3867–3873 (2014).

    PubMed  PubMed Central  Google Scholar 

  174. Hird, A. et al. Determining the incidence of pain flare following palliative radiotherapy for symptomatic bone metastases: results from three Canadian cancer centers. Int. J. Radiat. Oncol. Biol. Phys. 75, 193–197 (2009).

    PubMed  Google Scholar 

  175. Chow, E. et al. Dexamethasone in the prophylaxis of radiation-induced pain flare after palliative radiotherapy for bone metastases: a double-blind, randomised placebo-controlled, phase 3 trial. Lancet Oncol. 16, 1463–1472 (2015).

    CAS  PubMed  Google Scholar 

  176. van der Linden, Y. et al. Simple radiographic parameter predicts fracturing in metastatic femoral bone lesions: results from a randomized trial. Radiother. Oncol. 69, 21–31 (2003).

    PubMed  Google Scholar 

  177. Townsend, P. W. et al. Role of postoperative radiation therapy after stabilization of fractures caused by metastatic disease. Int. J. Radiat. Oncol. Biol. Phys. 31, 43–49 (1995).

    CAS  PubMed  Google Scholar 

  178. Drost, L. et al. Efficacy of postoperative radiation treatment for bone metastases in the extremities. Radiother. Oncol. 124, 45–48 (2017).

    PubMed  Google Scholar 

  179. Alghamdi, M. et al. Postoperative stereotactic body radiotherapy for spinal metastases. Chin. Clin. Oncol. 6, S18 (2017).

    PubMed  Google Scholar 

  180. Bhattacharya, I. S. & Hoskin, P. J. Stereotactic body radiotherapy for spinal and bone metastases. Clin. Oncol. 27, 298–306 (2015).

    CAS  Google Scholar 

  181. Sprave, T. et al. Randomized phase II trial evaluating pain response in patients with spinal metastases following stereotactic body radiotherapy versus three-dimensional conformal radiotherapy. Radiother. Oncol. 128, 274–282 (2018).

    PubMed  Google Scholar 

  182. Sprave, T. et al. Quality of life following stereotactic body radiotherapy versus three-dimensional conformal radiotherapy for vertebral metastases: secondary analysis of an exploratory phase II randomized trial. Anticancer. Res. 38, 4961–4968 (2018).

    PubMed  Google Scholar 

  183. Sprave, T. et al. Local response and pathologic fractures following stereotactic body radiotherapy versus three-dimensional conformal radiotherapy for spinal metastases — a randomized controlled trial. BMC Cancer 18, 859 (2018).

    PubMed  PubMed Central  Google Scholar 

  184. Nguyen, Q. N. et al. Single-fraction stereotactic vs conventional multifraction radiotherapy for pain relief in patients with predominantly nonspine bone metastases: a randomized phase 2 trial. JAMA Oncol. 5, 872–878 (2019).

    PubMed  PubMed Central  Google Scholar 

  185. Patchell, R. A. et al. Direct decompressive surgical resection in the treatment of spinal cord compression caused by metastatic cancer: a randomised trial. Lancet 366, 643–648 (2005). This paper is one of very few RCTs to evaluate radiotherapy versus surgical management of vertebral metastases associated with spinal cord compression.

    PubMed  Google Scholar 

  186. Maranzano, E. et al. Short-course versus split-course radiotherapy in metastatic spinal cord compression: results of a phase III, randomized, multicenter trial. J. Clin. Oncol. 23, 3358–3365 (2005).

    PubMed  Google Scholar 

  187. Hoskin, P. J. et al. Effect of single-fraction vs multifraction radiotherapy on ambulatory status among patients with spinal canal compression from metastatic cancer: the SCORAD randomized clinical trial. JAMA 322, 2084–2094 (2019).

    PubMed  PubMed Central  Google Scholar 

  188. Ballinger, J. R. Theranostic radiopharmaceuticals: established agents in current use. Br. J. Radiol. 91, 20170969 (2018).

    PubMed  PubMed Central  Google Scholar 

  189. Sartor, O., Hoskin, P. & Bruland, Ø. S. Targeted radio-nuclide therapy of skeletal metastases. Cancer Treat. Rev. 39, 18–26 (2013).

    CAS  PubMed  Google Scholar 

  190. Parker, C. et al. Targeted alpha therapy, an emerging class of cancer agents: a review. JAMA Oncol. 4, 1765–1772 (2018).

    PubMed  Google Scholar 

  191. Parker, C. et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N. Engl. J. Med. 18, 213–223 (2013). This study is the first RCT to demonstrate the benefits of a bone-seeking radiopharmaceutical on survival in patients with advanced cancer.

    Google Scholar 

  192. Smith, M. et al. Addition of radium-223 to abiraterone acetate and prednisone or prednisolone in patients with castration-resistant prostate cancer and bone metastases (ERA 223): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 20, 408–419 (2019).

    CAS  PubMed  Google Scholar 

  193. Tombal, B. F. et al. Decreased fracture rate by mandating bone-protecting agents in the EORTC 1333/PEACE III trial comparing enzalutamide and Ra223 versus enzalutamide alone: an interim safety analysis. J. Clin. Oncol. 37, 5007–5007 (2019).

    Google Scholar 

  194. Virgolini, I., Decristoforo, C., Haug, A., Fanti, S. & Uprimny, C. Current status of theranostics in prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 45, 471–495 (2018).

    PubMed  Google Scholar 

  195. Grubmüller, B. et al. Response assessment using 68Ga-PSMA ligand PET in patients undergoing 177Lu-PSMA radioligand therapy for metastatic castration-resistant prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 46, 1063–1072 (2019).

    PubMed  Google Scholar 

  196. Wüstemann, T., Haberkorn, U., Babich, J. & Mier, W. Targeting prostate cancer: prostate-specific membrane antigen based diagnosis and therapy. Med. Res. Rev. 39, 40–69 (2019).

    PubMed  Google Scholar 

  197. Hofman, M. S. et al. [177Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol. 19, 825–833 (2018).

    CAS  PubMed  Google Scholar 

  198. Thang, S. P. et al. Poor outcomes for patients with metastatic castration-resistant prostate cancer with low prostate-specific membrane antigen (PSMA) expression deemed ineligible for 177Lu-labelled PSMA radioligand therapy. Eur. Urol. Oncol. 2, 670–676 (2019).

    PubMed  Google Scholar 

  199. Weiss, K. R. et al. Fixation of pathological humeral fractures by the cemented plate technique. J. Bone Joint Surg. Br. 93, 1093–1097 (2011).

    CAS  PubMed  Google Scholar 

  200. Willeumier, J. J., van der Linden, Y. M., van de Sande, M. A. J. & Dijkstra, P. D. S. Treatment of pathological fractures of the long bones. EFORT Open. Rev. 1, 136–145 (2017).

    PubMed  Google Scholar 

  201. Damron T. & Mann K. Fracture risk assessment and clinical decision making for patients with metastatic bone disease. J. Orthop. Res. 38, 1175–1190 (2020).

    PubMed  PubMed Central  Google Scholar 

  202. Van der Linden, Y. M. et al. Comparative analysis of risk factors for pathological fracture with femoral metastases. J. Bone Joint Surg. Br. 86, 566–573 (2004).

    PubMed  Google Scholar 

  203. Mirels, H. Metastatic disease in long bones. A proposed scoring system for diagnosing impending pathologic fractures. Clin. Orthop. Relat. Res. 249, 256–264 (1989).

    Google Scholar 

  204. Disch, A. C., Kleber, C. & Redemann, D. Current surgical strategies for treating spinal tumors: results of a questionnaire survey among members of the German Spine Society (DWG). Eur. J. Surg. Oncol. 46, 89–94 (2020).

    CAS  PubMed  Google Scholar 

  205. Fisher, C. G. et al. A novel classification system for spinal instability in neoplastic disease: an evidence-based approach and expert consensus from the Spine Oncology Study Group. Spine 35, E1221–E1229 (2010).

    PubMed  Google Scholar 

  206. Health Quality Ontario. Vertebral augmentation involving vertebroplasty or kyphoplasty for cancer-related vertebral compression fractures: a systematic review. Ont. Health Technol. Assess. Ser. 16, 1–202 (2016).

    PubMed Central  Google Scholar 

  207. Middlemiss, T., Laird, B. & Fallon, M. Mechanisms of cancer-induced bone pain. Clin. Oncol. 23, 389–392 (2011).

    Google Scholar 

  208. Urch, C. The pathophysiology of cancer-induced bone pain: current understanding. Palliat. Med. 18, 267–274 (2004).

    PubMed  Google Scholar 

  209. Gordon-Williams, R. M. & Dickenson, A. H. Central neuronal mechanisms in cancer-induced bone pain. Curr. Opi. Support. Palliat. Care 1, 6–10 (2007).

    Google Scholar 

  210. Gardner, K., Laird, B. J. A., Fallon, M. T. & Sande, T. A. A systematic review examining clinical markers and biomarkers of analgesic response to radiotherapy for cancer-induced bone pain. Crit. Rev. Oncol. Hematol. 133, 33–44 (2018).

    PubMed  Google Scholar 

  211. Zajączkowska, R., Kocot-Kępska, M., Leppert, W. & Wordliczek, J. Bone pain in cancer patients: mechanisms and current treatment. Int. J. Mol. Sci. 20, 6047–6066 (2019).

    PubMed Central  Google Scholar 

  212. McWilliams, K. & Fallon, M. Fast acting fentanyl preparations. Q. J. Med. 106, 887–890 (2013).

    CAS  Google Scholar 

  213. Fallon, M. et al. A randomised, double blind trial of pregabalin versus placebo in conjunction with palliative radiotherapy for cancer-induced bone pain. J. Clin. Oncol. 34, 550–556 (2016).

    CAS  PubMed  Google Scholar 

  214. Miller, S. P-126 effectiveness of gabapentin and pregabalin for cancer-induced bone pain: a systematic review. BMJ Support. Palliat. Care 7, A46–A47 (2017).

    Google Scholar 

  215. Hoskin, P. et al. A multicenter randomized trial of ibandronate compared with single-dose radiotherapy for localized metastatic bone pain in prostate cancer. J. Natl Cancer Inst. 107, djv197 (2015).

    PubMed  Google Scholar 

  216. Bruel, B. M. & Burton, A. W. Intrathecal therapy for cancer-related pain. Pain. Med. 17, 2404–2421 (2016).

    PubMed  PubMed Central  Google Scholar 

  217. Bennett, M. I. et al. Feasibility study of transcutaneous electrical nerve stimulation (TENS) for cancer bone pain. J. Pain 11, 351–359 (2010).

    PubMed  Google Scholar 

  218. Fallon, M. T. Neuropathic pain in cancer. Br. J. Anaesth. 111, 105–111 (2013).

    CAS  PubMed  Google Scholar 

  219. Laird, B. et al. Characterization of cancer induced bone pain: an exploratory study. J. Support. Care Cancer 19, 1393–1401 (2011).

    Google Scholar 

  220. Dennis, K. et al. Palliative radiotherapy for bone metastases in the last 3 months of life: worthwhile or futile? Clin. Oncol. 23, 709–715 (2011).

    CAS  Google Scholar 

  221. Tahara, R. K., Brewer, T. M., Theriault, R. L. & Ueno, N. T. Bone metastasis of breast cancer. Adv. Exp. Med. Biol. 1152, 105–129 (2019).

    CAS  PubMed  Google Scholar 

  222. Kinnane, N. Burden of bone disease. Eur. J. Oncol. Nurs. 11 (Suppl 2), S28–S31 (2007).

    PubMed  Google Scholar 

  223. Cleeland, C. S. The measurement of pain from metastatic bone disease: capturing the patient’s experience. Clin. Cancer Res. 12, 6236s–6242s (2006).

    PubMed  Google Scholar 

  224. Harris, K. et al. Patients’ and health care professionals’ evaluation of health-related quality of life issues in bone metastases. Eur. J. Cancer 45, 2510–2518 (2009).

    PubMed  Google Scholar 

  225. Ferrer Albiach, C. et al. Real-life management of patients with breakthrough cancer pain caused by bone metastases in Spain. J. Pain. Res. 12, 2125–2135 (2019).

    PubMed  PubMed Central  Google Scholar 

  226. Contartese, D., Salamanna, F., Veronesi, F. & Fini, M. Relevance of humanized three-dimensional tumor tissue models: a descriptive systematic literature review. Cell Mol Life Sci. https://doi.org/10.1007/s00018-020-03513-y (2020).

  227. Antonio, G. et al. Immune system and bone microenvironment: rationale for targeted cancer therapies. Oncotarget 11, 480–487 (2020).

    Google Scholar 

  228. Sousa, S. & Clézardin, P. Bone-targeted therapies in cancer-induced bone disease. Calcif. Tissue Int. 102, 227–250 (2018).

    CAS  PubMed  Google Scholar 

  229. Hadji, P., Coleman, R. & Gnant, M. Bone effects of mammalian target of rapamycin (mTOR) inhibition with everolimus. Crit. Rev. Oncol. Hematol. 87, 101–111 (2013).

    PubMed  Google Scholar 

  230. Hortobagyi, G. N. Everolimus plus exemestane for the treatment of advanced breast cancer: a review of subanalyses from BOLERO-2. Neoplasia 17, 279–288 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Liang, W. et al. Targeting cathepsin K diminishes prostate cancer establishment and growth in murine bone. J. Cancer Res. Clin. Oncol. 145, 1999–2012 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Jensen, A. B. et al. The cathepsin K inhibitor odanacatib suppresses bone resorption in women with breast cancer and established bone metastases: results of a 4-week, double-blind, randomized, controlled trial. Clin. Breast Cancer 10, 452–458 (2010).

    CAS  PubMed  Google Scholar 

  233. Uehara, S., Udagawa, N. & Kobayashi, Y. Regulation of osteoclast function via Rho–Pkn3–c-Src pathways. J. Oral. Biosci. 61, 135–140 (2019).

    PubMed  Google Scholar 

  234. Lee, R. J. & Smith, M. R. Targeting MET and vascular endothelial growth factor receptor signaling in castration-resistant prostate cancer. Cancer J. 19, 90–98 (2013).

    PubMed  PubMed Central  Google Scholar 

  235. Smith, D. C. et al. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. J. Clin. Oncol. 31, 412–419 (2013).

    CAS  PubMed  Google Scholar 

  236. Smith, M. et al. Phase III study of cabozantinib in previously treated metastatic castration-resistant prostate cancer: COMET-1. J. Clin. Oncol. 34, 3005–3013 (2016).

    CAS  PubMed  Google Scholar 

  237. Escudier, B. et al. Cabozantinib, a new standard of care for patients with advanced renal cell carcinoma and bone metastases? Subgroup analysis of the METEOR trial. J. Clin. Oncol. 36, 765–772 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Cosman, F. et al. Romosozumab treatment in postmenopausal women with osteoporosis. N. Engl. J. Med. 375, 1532–1543 (2016).

    CAS  PubMed  Google Scholar 

  239. Oyajobi, B. O. et al. Stimulation of new bone formation by the proteasome inhibitor, bortezomib: implications for myeloma bone disease. Br. J. Haematol. 139, 434–438 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Chantry, A. D. et al. Inhibiting activin-A signaling stimulates bone formation and prevents cancer-induced bone destruction in vivo. J. Bone Miner. Res. 25, 2633–2646 (2010).

    PubMed  Google Scholar 

  241. Rosanò, L., Spinella, F. & Bagnato, A. Endothelin 1 in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer 13, 637–651 (2013).

    PubMed  Google Scholar 

  242. Buijs, J. T., Stayrook, K. R. & Guise, T. A. The role of TGF-β in bone metastasis: novel therapeutic perspectives. Bonekey Rep. 1, 96 (2012).

    PubMed  PubMed Central  Google Scholar 

  243. Aiello, F., Carullo, G., Badolato, M. & Brizzi, A. TRPV1–FAAH–COX: the couples game in pain treatment. ChemMedChem 11, 1686–1694 (2016).

    CAS  PubMed  Google Scholar 

  244. Padhani, A. R. et al. Rationale for modernising imaging in advanced prostate cancer. Eur. Urol. Focus. 3, 198–205 (2017).

    Google Scholar 

  245. Perez-Lopez, R. et al. Multiparametric magnetic resonance imaging of prostate cancer bone disease correlation with bone biopsy histological and molecular features. Invest. Radiol. 53, 96–102 (2018).

    PubMed  PubMed Central  Google Scholar 

  246. Westbrook, J. A. et al. CAPG and GIPC1: breast cancer biomarkers for bone metastasis development and treatment. J. Natl Cancer Inst. 108, djv360 (2016).

    PubMed Central  Google Scholar 

  247. Pavlovic, M. et al. Enhanced MAF oncogene expression and breast cancer bone metastasis. J. Natl Cancer Inst. 107, djv256 (2015).

    PubMed  PubMed Central  Google Scholar 

  248. Coleman, R. et al. Effect of MAF amplification on treatment outcomes with adjuvant zoledronic acid in early breast cancer: a secondary analysis of the international, open-label, randomised, controlled, phase 3 AZURE (BIG 01/04) trial. Lancet Oncol. 18, 1543–1552 (2017).

    CAS  PubMed  Google Scholar 

  249. Theriault, R. L. et al. Pamidronate reduces skeletal morbidity in women with advanced breast cancer and lytic bone lesions: a randomised, placebo-controlled trial. J. Clin. Oncol. 17, 846–854 (1999).

    CAS  PubMed  Google Scholar 

  250. Kohno, N. et al. Zoledronic acid significantly reduces skeletal complications compared with placebo in Japanese women with bone metastases from breast cancer: a randomized, placebo-controlled trial. J. Clin. Oncol. 20, 3299–3301 (2005).

    Google Scholar 

  251. Rosen, L. S. et al. Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in treatment of skeletal complications in patients with advanced multiple myeloma or breast cancer: a randomized, double-blind, multicenter, comparative trial. Cancer 98, 1735–1744 (2003).

    CAS  PubMed  Google Scholar 

  252. Body, J. J. et al. Intravenous ibandronate reduces the incidence of skeletal complications in patients with breast cancer and bone metastases. Ann. Oncol. 14, 1399–1405 (2003).

    PubMed  Google Scholar 

  253. Rosen, L. S. et al. Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial. Cancer J. 7, 377–387 (2001).

    CAS  PubMed  Google Scholar 

  254. Lahtinen, R., Laakso, M., Palva, I., Virkkunen, P. & Elomaa, I. Randomised, placebo-controlled multicentre trial of clodronate in multiple myeloma. Finnish Leukaemia Group. Lancet 340, 1049–1052 (1992).

    CAS  PubMed  Google Scholar 

  255. Danson, S. et al. An exploratory randomized-controlled trial of the efficacy of the Src-kinase inhibitor saracatinib as a novel analgesic for cancer-induced bone pain. J. Bone Oncol. 19, 100261 (2019).

    PubMed  PubMed Central  Google Scholar 

  256. Koreckij, T. et al. Dasatinib inhibits the growth of prostate cancer in bone and provides additional protection from osteolysis. Br. J. Cancer 101, 263–268 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  257. Yu, E. Y. et al. Castration-resistant prostate cancer bone metastasis response measured by 18F-fluoride PET after treatment with dasatinib and correlation with progression-free survival: results from American College of Radiology Imaging Network 6687. J. Nucl. Med. 56, 354–360 (2015).

    CAS  PubMed  Google Scholar 

  258. Rizzo, S. et al. The effects of enzalutamide and abiraterone on skeletal related events and bone radiological progression free survival in castration resistant prostate cancer patients: an indirect comparison of randomized controlled trials. Crit. Rev. Oncol. Hematol. 120, 227–233 (2017).

    PubMed  Google Scholar 

  259. Iuliani, M. et al. Biological and clinical effects of abiraterone on anti-resorptive and anabolic activity in bone microenvironment. Oncotarget 6, 12520–12528 (2015).

    PubMed  PubMed Central  Google Scholar 

  260. Achrol, A. S. et al. Brain metastases. Nat. Rev. Dis. Prim. 5, 5 (2019).

    PubMed  Google Scholar 

  261. Coleman, R. E., Rathbone, E. & Brown, J. E. Management of cancer treatment-induced bone loss. Nat. Rev. Rheumatol. 9, 365–374 (2013).

    CAS  PubMed  Google Scholar 

  262. Berish, R. B. et al. Translational models of prostate cancer bone metastasis. Nat. Rev. Urol. 15, 403–421 (2018).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

P.I.C. acknowledges J. Gibson and the Ernest Heine Family Foundation, the Kay Stubbs Cancer Research Grant, the Cancer Council New South Wales, the Leukaemia Foundation, the Prostate Cancer Foundation of Australia, The Movember Foundation and the National Health and Medical Research Council for supporting his research. P.C. acknowledges INSERM, ‘Appel à Projets LIA/LEA 2016’ (grant no.: ASC17018CSA), Weston Park Cancer Charity (grant no.: CA163) and the LabEX DEVweCAN (ANR-10-LABX-61) from Université de Lyon, within the programme ‘Investissements d’Avenir’ (ANR-11-IDEX-0007) operated by the French National Research Agency (ANR), for funding his research.

Author information

Authors and Affiliations

Authors

Contributions

Introduction (R.E.C.); Epidemiology (R.E.C., L.C.); Mechanisms/pathophysiology (R.E.C., P.I.C., T.G.); Diagnosis, screening and prevention (R.E.C., A.R.P.); Management (R.E.C., E.C., L.C., M.F., A.R.P., S.C., R.C.); Quality of life (R.E.C.); Outlook (R.E.C., P.C.); Overview of Primer (R.E.C.).

Corresponding author

Correspondence to Robert E. Coleman.

Ethics declarations

Competing interests

R.E.C. has received honoraria from Amgen and Novartis, and has stock options with Inbiomotion related to a patented biomarker. P.I.C. is the recipient of a grant from Amgen, and received honoraria and participated in advisory boards from Amgen. P.C. has received honoraria from Amgen. L.C. has received research grants from Amgen, Bayer, Novartis and Roche, speaker honoraria from Amgen, Bayer, Janssen, Lilly and Roche, and is also a consultant for Amgen, Novartis and Servier. All other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Disease Primers thanks Jean-Yves Blay, Claire Edwards, Rachelle Johnson, Klaus Pantel, Julie Sterling 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

Bone resorption

The breakdown or dissolution of bone by osteoclasts.

Osteoclasts

Multinucleated cells derived from granulocyte–macrophage precursors that break down (resorb) bone.

Osteoblast

A bone-forming cell derived from mesenchymal, fibroblast-like cells that forms bone and usually works in close collaboration with osteoclasts to ensure bone resorption and formation are linked and balanced.

Bisphosphonates

A class of drugs that prevent bone loss by disrupting osteoclast-mediated bone resorption.

Denosumab

A fully humanized monoclonal antibody that binds to receptor activator of nuclear factor-κB ligand (RANKL) and inhibits osteoclast-mediated bone resorption.

Axial skeleton

The central portion of the skeleton comprising the skull, spine, pelvis, shoulders, hips and ribs.

Exosomes

Extracellular vesicles that may contain proteins, RNA or DNA.

Bone turnover

A measure of the speed of bone resorption and formation as determined by biomarkers or bone biopsy.

Endosteum

A thin vascular membrane of connective tissue that lines the inner surface of the bony tissue that forms the medullary cavity of long bones.

Leukoerythroblastic anaemia

Anaemia resulting from a structural or neoplastic problem in the bone marrow and the resultant appearance of immature erythrocyte and white cell precursors in the peripheral blood.

Diffusion weighted imaging

A form of MRI based upon measuring the random Brownian motion of water molecules within a voxel of tissue. Highly cellular tissues or those with cellular swelling exhibit lower diffusion coefficients.

Castration-resistant prostate cancer

(CRPC). A phase in the evolution of advanced prostate cancer that progresses despite androgen deprivation therapy.

Castration-sensitive prostate cancer

Prostate cancer that can be controlled by lowering androgen levels.

Oligometastasis

A single site or a few (<5) sites of metastasis within up to three different metastatic sites.

Curettage

Removal of tissue by scraping or scooping rather than surgical resection.

Analgesic titration

A stepwise increase in analgesics, especially opioids, to achieve pain control without excessive toxicity.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Coleman, R.E., Croucher, P.I., Padhani, A.R. et al. Bone metastases. Nat Rev Dis Primers 6, 83 (2020). https://doi.org/10.1038/s41572-020-00216-3

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41572-020-00216-3

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