Skip to main content
Current Oncology logoLink to Current Oncology
. 2020 Aug 1;27(4):220–224. doi: 10.3747/co.27.6631

Practical update for the use of bone-targeted agents in patients with bone metastases from metastatic breast cancer or castration-resistant prostate cancer

D Southcott *, A Awan , K Ghate , M Clemons , R Fernandes *,
PMCID: PMC7467800  PMID: 32905286

Abstract

Bone metastases are a significant source of morbidity and mortality for patients with breast and prostate cancer. In this review, we discuss key practical themes regarding the use of bone-targeted agents (btas) such as bisphosphonates and denosumab for managing bony metastatic disease. The btas both delay the onset and reduce the incidence of skeletal-related events (sres), defined as any or all of a need for radiation therapy or surgery to bone, pathologic fracture, spinal cord compression, or hypercalcemia of malignancy. They have more modest benefits for pain and other quality-of-life measures.

Regardless of the benefits of btas, it should always be remembered that the palliative management of metastatic bone disease is multimodal and multidisciplinary. The collaboration of all disciplines is essential for optimal patient care.

Special consideration is given to these key questions:

  • ■ What are btas, and what is their efficacy?

  • ■ What are their common toxicities?

  • ■ When should they be initiated?

  • ■ How do we choose the appropriate bta?

  • ■ What is the appropriate dose, schedule, and duration of btas?

Keywords: Bony metastases, bisphosphonates, skeletal-related events, bone-targeted agents, denosumab

INTRODUCTION

Bone is the most common site of metastasis in many malignancies, including breast (70%), prostate (70%–90%), thyroid (60%), lung (35%), and renal cell carcinomas (35%), and bone metastases can be associated with morbidity and mortality1,2. Since the start of the 2000s, there has been sustained growth in the understanding of the underlying biology of bone metastases and widespread incorporation of inhibitors of osteoclast function—namely, bisphosphonates and receptor activator of nuclear factor κB ligand inhibitors (denosumab)—into clinical practice35.

Bone metastases can be either osteolytic, osteoblastic, or mixed, and they most commonly occur in the long bones (femur and humerus) and axial skeleton6,7. These lesions lead to bone destruction, resulting in significant morbidity and impaired quality of life (qol). In clinical trials, the efficacy of bone-targeted agents (btas) is commonly assessed using the rate of skeletal-related events (sres), which occur in approximately 27%–40% of patients with metastatic castration-resistant prostate cancer (crpc) and metastatic breast cancer (mbca)2,8. Given that sres and impaired patient qol are both so important for patient care, it is essential that the full range of multimodal and multidisciplinary therapies are readily available for patients912.

BTAs FOR METASTASES TO BONE

Types of BTAs

For the purposes of this review, we focus on the two types of btas that are approved for use in clinical practice: bisphosphonates and denosumab. Bisphosphonates act primarily as inhibitors of osteoclast activity and have been studied in the management of a variety of malignancies. In oncology, the most commonly used agents are zoledronate and pamidronate. In patients with mbca and bone metastases, bisphosphonates have been shown to significantly reduce the incidence of sres, but have not been shown to improve progression-free survival or overall survival1315. A 2017 meta-analysis showed that bisphosphonates reduce the risk of sres in advanced breast cancer by 14%16. Additionally, a Cochrane review concluded that there is some evidence to support modest pain relief associated with the use of bisphosphonates, although current guidelines do not recommend such use exclusively for analgesic purposes17,18. In patients with crpc metastatic to bone, zoledronate has been shown to reduce sres by 36%19.

Denosumab is a fully human monoclonal antibody that acts by inhibiting the receptor activator of nuclear factor κB ligand, which stimulates the activity and development of osteoclasts, thereby reducing their activity and growth. Denosumab was shown to be superior to zoledronate in patients with bone metastasis from mbca and crpc by delaying the onset of first and subsequent sres14,20. In a meta-analysis of three randomized clinical trials, denosumab reduced the risk of a first sre by 17% and delayed the time to first sre by a median of 8.2 months21. In one report in patients with mbca, denosumab, compared with zoledronate, also showed a benefit in health-related qol measures22.

Treatment Considerations and Toxicities

The toxicities of btas are well studied, and bisphosphonates and denosumab all share a number of adverse events, including hypocalcemia, osteonecrosis of the jaw, and atypical femur fractures. Patients who are starting a bta should receive appropriate calcium and vitamin D supplementation to reduce the risk of hypocalcemia, which occurs more frequently with denosumab (12.4%) than with bisphosphonates (5.3%)23,24. Risk of osteonecrosis of the jaw is rare with either therapy, occurring at rates less than 2% in the first year of treatment25. The risk is higher with denosumab than with bisphosphonates and increases with longer duration of therapy. Dental examination before bta initiation is recommended by most practice guidelines, and ongoing symptom inquiry is important throughout treatment for prompt identification of this serious adverse event18.

Bisphosphonates are specifically associated with nephrotoxicity, and renal function must be monitored26. The risk can be reduced by adhering to recommended doses, dosing intervals, and infusion rates as described later in this article27.

When to Initiate Treatment

It is widely accepted practice to initiate treatment with a bta at first radiographic evidence of bony metastases, because risk of sres is highest on diagnosis of metastatic disease and within the first year18,28,29. Whether all patients have to commence treatment as soon as there is a radiologic diagnosis has not been investigated. Clinical context should dictate management; in certain situations (for instance, in a patient with a low burden of disease or with visceral metastases and limited life expectancy), delaying the start of a bta might be more sensible4.

Aspects of the pathophysiology of bone lesions have led to some observations in the treatment of osteolytic compared with osteoblastic metastases. Bone metastases in patients with breast cancer are primarily osteolytic, and therefore treatment with inhibitors of osteoclast activity, such as bisphosphonates, is indicated13,30. On the other hand, bone metastases in patients with prostate cancer are primarily osteoblastic, leading to the deposition of calcium in new bone. The rationale for the use of bisphosphonates in the treatment of patients with prostate cancer comes from studies showing that abnormal osteoblastic bone formation is associated with osteoclastic bone resorption and an elevation in osteoclastic markers31,32. Multiple randomized trials have shown significant reductions in the frequency of sres in patients with bone metastases from a variety of solid tumours that are treated with an osteoclast inhibitor; the use of such agents is therefore recommended for most patients with bone metastases from solid tumours, regardless of whether those metastases are osteolytic or osteoblastic15,3335.

Choice of Agent and Dosing Schedule

The choice of bta depends on a number of factors, including tumour site, patient comorbidities, and cost-effectiveness. In mbca, a joint guideline from the American Society of Clinical Oncology and Ontario Health (Cancer Care Ontario) does not recommend one bta over another18. Although denosumab might be preferred based on its associated sre reduction and its subcutaneous route of administration, those benefits have to be balanced against its significantly higher drug acquisition cost. In a cost-effectiveness analysis, zoledronate given every 3 months was shown to be substantially more cost-effective than monthly denosumab in reducing sres36.

If a bisphosphonate is selected, zoledronate has the greatest demonstrated efficacy37. The joint 2017 guideline from the American Society of Clinical Oncology and Ontario Health (Cancer Care Ontario) for mbca recommends the use of zoledronate at a dose of 4 mg given intravenously over no less than 15 minutes18. Pamidronate 90 mg given intravenously also has demonstrated efficacy in reducing the risk of sres in mbca and would be a reasonable option, but it does have a longer infusion time (2 hours)38. In contrast, pamidronate did not demonstrate any benefit over placebo in the treatment of bony metastases from crpc39. A 2017 Genitourinary Disease Site Group guideline from Ontario Health (Cancer Care Ontario) recommended denosumab or zoledronic acid for preventing or delaying sres in men with metastatic crpc40,41.

In terms of dosing schedule, despite the fact that the half-life of bisphosphonates and bone turnover biomarkers exceed 1 year, the dosing interval has historically been more frequent, to coincide with every-3-weeks chemotherapy schedules42. Less-frequent dosing of btas in breast and prostate cancer is supported by data from three randomized controlled studies. In a meta-analysis, sre rates in patients with mbca were similar for zoledronate given every 12 weeks or every 3–4 weeks, regardless of whether patients started on standard dosing (every 3–4 weeks) or de-escalated dosing (every 12 weeks)43. A more recent trial evaluating the noninferiority of 12-weekly compared with 4-weekly dosing for btas in patients with breast cancer and crpc showed that a dose–de-escalated schedule can also be applied to pamidronate44.

Currently, the recommended dose and schedule for denosumab is 120 mg administered subcutaneously every 4 weeks4,18. Denosumab has also been tested in a dose–de-escalated schedule. A recent trial that evaluated the noninferiority of 12-weekly compared with 4-weekly dosing for btas suggested that the same dose–de-escalated schedule could also be applied to denosumab44. To direct future treatment recommendations, one ongoing clinical trial is currently comparing 12-weekly with 4-weekly denosumab in patients with mbca and crpc33,45. Data so far would suggest that de-escalation of all commonly used bone agents, including denosumab, to 12-weekly therapy is a reasonable clinical strategy. Table I summarizes the administration schedules and doses for those commonly used agents. Table II shows estimated prices for btas in Canada.

TABLE I.

Recommended doses and administration schedules for bone-targeted agents

Agent Indication Creatinine clearance (mL/min) Dose (mg) Route Schedule Infusion rate
Zoledronate mBCa and CRPC >60 4 Intravenous Every 12 weeks or every 3–4 weeks18 No less than 15 minutes
50–59 3.5
40–49 3.3
30–39 3
<30 Use not recommended

Pamidronate mBCa only >90 90 Intravenous Every 3–4 weeks; may consider every 12 weeks18 No less than 2 hours; 4 hours recommended
30–90 90
<30 Use not recommended

Denosumab mBCa and CRPC NA 120 Subcutaneous Every 3–4 weeks; may consider every 12 weeks36 NA

mBCa = metastatic breast cancer; CRPC = castration-resistant prostate cancer; NA = not applicable.

TABLE II.

Estimated price for bone-targeted agents in Canada

Agent Dose (mg) Route Schedule Infusion rate Price (CA$)

Per dosea Total annual
Zoledronate 4 Intravenous Every 3–4 weeks No less than 15 minutes 335.40b 4,024.80
415.56c 4,986.70

Pamidronate 90 Intravenous Every 3–4 weeks No less than 2 hours 86.78d 3,124.08

Denosumab 120 Subcutaneous Every 3–4 weeks NA 575.75 6,909.00
a

Per-dose prices represent a single infusion or injection per the Ontario Drug Benefit Exceptional Access Formulary, October 2015. Drug cost might be reimbursed through the Ontario Drug Benefit Plan. Drug prices are dynamic; thus, the prices listed here might not reflect current prices.

b

Zoledronic acid (Taro Pharmaceuticals, Brampton, ON).

c

Zoledronate (JAMP Pharma Corporation, Boucherville, QC).

d

10 mL Vial, 3 mg/mL injection (Pfizer Canada ULC, Kirkland, Québec)

NA = not applicable.

Duration of Treatment

American Society of Clinical Oncology guidelines recommend continuing treatment with btas indefinitely18. That recommendation must be balanced against long-term toxicity risks and patient-centred goals of care. Some data suggest that treatment should continue for 6 months at minimum to achieve a demonstrable reduction in the risk of sres19.

For patients who experience a sre while on treatment with a bta, therapy continuation is recommended to increase the time to a subsequent sre46. One study investigating the benefit of switching from pamidronate to zoledronic acid in patients with high-risk disease did not show any reduction in sres or qol and pain scores, and thus switching between bisphosphonates is not recommended47.

Treatment with newer anticancer therapies has extended life expectancy in patients with mbca and crpc by a number of years. As a result, the question of the duration of bta use should take into consideration life expectancy, overall burden of disease, and risk of sres in each individual patient. Most clinical trials have evaluated the use of btas for only a maximum of 2 years, and hence no high-quality evidence to support their use beyond 2 years has been developed3335. Duration of use is currently being further evaluated in a series of Canadian studies.

SUMMARY

Bone-targeted agents are an integral component of the palliative management of bony metastatic disease in mbca and crpc. Knowledge of the available agents and their role in the treatment of patients with bony metastases is essential for general practitioners in oncology. The btas have demonstrated efficacy in reducing the risk of sres and in modestly improving pain and other qol outcomes. However, it is important to remember that btas have no effect on either progression-free or overall survival, and they have significant associated toxicities and costs. Further areas of study include clarifying the optimal dose, schedule, and duration of treatment for these agents.

We recommend the use of btas to reduce the risk of sres for most patients with bone metastases from solid tumours. For patients with a minimal bone tumour burden or limited expected survival, treatment with a bta should be discussed and individualized. We advise that the use of btas be specifically applied to crpc with bone metastases. In the absence of excessive toxicity, and if within the patient’s and the clinician’s joint treatment goals, btas should be continued indefinitely.

Key Points.

  • ■ Bisphosphonates and denosumab are osteoclast inhibitors that are recommended for the treatment of bony metastases in patients with breast cancer and crpc.

  • ■ Treatment with a bta can be considered at the first evidence of bony metastasis.

  • ■ Before therapy begins, guidelines recommend that patients undergo dental examination because of a risk—albeit low—of osteonecrosis of the jaw. Supplementation with vitamin D and calcium should also be initiated.

  • ■ If initiating a bisphosphonate, a dose–de-escalated schedule of administration every 12 weeks can be used. If initiating denosumab, the current recommendation is for administration every 4 weeks. However, the results of ongoing clinical trials suggest that a 12-weekly schedule of denosumab is reasonable.

  • ■ Bone-targeted agents may be continued beyond the 2 years evaluated in clinical trials. However, given the possibility that their toxicities might outweigh their benefits with longer duration of use, ongoing clinical trials are evaluating this approach.

Footnotes

CONFLICT OF INTEREST DISCLOSURES

We have read and understood Current Oncology’s policy on disclosing conflicts of interest, and we declare the following interests: RF has consulted for Novartis, Janssen, Merck, Pfizer, and Bayer. All other authors have no conflicts of interest to disclose.

REFERENCES

  • 1.Coleman RE. Bisphosphonates: clinical experience. Oncologist. 2004;9(suppl 4):14–27. doi: 10.1634/theoncologist.9-90004-14. [DOI] [PubMed] [Google Scholar]
  • 2.Coleman RE. Metastatic bone disease: clinical features, pathophysiology and treatment strategies. Cancer Treat Rev. 2001;27:165–76. doi: 10.1053/ctrv.2000.0210. [DOI] [PubMed] [Google Scholar]
  • 3.Clemons M, Gelmon KA, Pritchard KI, Paterson AHG. Bone-targeted agents and skeletal-related events in breast cancer patients with bone metastases: the state of the art. Curr Oncol. 2012;19:259–68. doi: 10.3747/co.19.1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bouganim N, Dranitsaris G, Amir E, Clemons M. Optimising the use of bone-targeted agents in patients with metastatic cancers: a practical guide for medical oncologists. Support Care Cancer. 2011;19:1687–96. doi: 10.1007/s00520-011-1230-9. [DOI] [PubMed] [Google Scholar]
  • 5.Bouganim N, Clemons MJ. Bone targeted agents in the treatment of bone metastases: rank outsider or new kid on the block? Future Oncol. 2011;7:381–3. doi: 10.2217/fon.10.192. [DOI] [PubMed] [Google Scholar]
  • 6.Coleman R, Body JJ, Aapro M, Hadji P, Herrstedt J on behalf of the esmo Guidelines Working Group. Bone health in cancer patients: esmo clinical practice guidelines. Ann Oncol. 2004;25(suppl 3):iii124–37. doi: 10.1093/annonc/mdu103. [DOI] [PubMed] [Google Scholar]
  • 7.Mastro AM, Gay CV, Welch DR. The skeleton as a unique environment for breast cancer cells. Clin Exp Metastasis. 2003;20:375–84. doi: 10.1023/A:1022995403081. [DOI] [PubMed] [Google Scholar]
  • 8.Kawai AT, Martinez D, Saltus CW, Vassilev ZP, Soriano-Gabarro M, Kaye JA. Incidence of skeletal related events in castration resistant prostate cancer: an observational retrospective cohort study in the US. Prostate Cancer. 2019;2019 doi: 10.1155/2019/5971615. 5971615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Coleman RE, Lipton A, Roodman GD, et al. Metastasis and bone loss: advancing treatment and prevention. Cancer Treat Rev. 2010;36:615–20. doi: 10.1016/j.ctrv.2010.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Neville-Webbe HL, Coleman RE. Bisphosphonates and rank ligand inhibitors or the treatment and prevention of metastatic bone disease. Eur J Cancer. 2010;46:1211–22. doi: 10.1016/j.ejca.2010.02.041. [DOI] [PubMed] [Google Scholar]
  • 11.Lipton A, Zheng M, Seaman J. Zoledronic acid delays the onset of skeletal-related events and progression of skeletal disease in patients with advanced renal cell carcinoma. Cancer. 2003;98:962–9. doi: 10.1002/cncr.11571. [DOI] [PubMed] [Google Scholar]
  • 12.Saad F, Eastham JA. Zoledronic acid use in patients with bone metastases from renal cell carcinoma or bladder cancer. Semin Oncol. 2010;37(suppl 1):S38–44. doi: 10.1053/j.seminoncol.2010.06.001. [DOI] [PubMed] [Google Scholar]
  • 13.Pavlakis N, Schmidt R, Stockler M. Bisphosphonates for breast cancer. Cochrane Database Syst Rev. 2005 doi: 10.1002/14651858.CD003474.pub2. CD003474. [DOI] [PubMed] [Google Scholar]
  • 14.Stopeck AT, Lipton A, Body JJ, 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. 2010;28:5132–9. doi: 10.1200/JCO.2010.29.7101. [DOI] [PubMed] [Google Scholar]
  • 15.Saad F, Gleason DM, Murray R, et al. on behalf of the Zoledronic Acid Prostate Cancer Study Group. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst. 2002;94:1458–68. doi: 10.1093/jnci/94.19.1458. [DOI] [PubMed] [Google Scholar]
  • 16.O’Carrigan B, Wong MH, Wilson ML, Stockler MR, Pavlakis N, Goodwin A. Bisphosphonates and other bone agents for breast cancer. Cochrane Database Syst Rev. 2017;10 doi: 10.1002/14651858.CD003474.pub4. CD003474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wong R, Wiffen PJ. Bisphosphonates for the relief of pain secondary to bone metastases. Cochrane Database Syst Rev. 2002;2002 doi: 10.1002/14651858.CD002068. CD002068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Van Poznack C, Somerfield MR, Barlow WE, et al. Role of bone-modifying agents in metastatic breast cancer: an American Society of Clinical Oncology–Cancer Care Ontario focused guideline update. J Clin Oncol. 2017;35:3978–86. doi: 10.1200/JCO.2017.75.4614. [DOI] [PubMed] [Google Scholar]
  • 19.Saad F, Gleason DM, Murray R, et al. on behalf of the Zoledronic Acid Prostate Cancer Study Group. Long-term efficacy of zoledronic acid for the prevention of skeletal complications in patients with metastatic hormone refractory prostate cancer. J Natl Cancer Inst. 2004;96:879–82. doi: 10.1093/jnci/djh141. [DOI] [PubMed] [Google Scholar]
  • 20.Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration resistant prostate cancer: a randomized, double blind study. Lancet. 2011;377:813–22. doi: 10.1016/S0140-6736(10)62344-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lipton A, Fizazi K, Stopeck AT, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer. 2012;48:3082–92. doi: 10.1016/j.ejca.2012.08.002. [DOI] [PubMed] [Google Scholar]
  • 22.Martin M, Bell R, Bourgeois H, et al. Bone-related complications and quality of life in advanced breast cancer: results from a randomized phase iii trial of denosumab versus zoledronic acid. Clin Cancer Res. 2012;18:4841–9. doi: 10.1158/1078-0432.CCR-11-3310. [DOI] [PubMed] [Google Scholar]
  • 23.Wang-Gillam A, Miles DA, Hutchins LF. Evaluation of vitamin D deficiency in breast cancer patients on bisphosphonates. Oncologist. 2008;13:821–7. doi: 10.1634/theoncologist.2008-0013. [DOI] [PubMed] [Google Scholar]
  • 24.Body JJ, Bone HG, de Boer RH, et al. Hypocalcemia in patients with metastatic bone disease treated with denosumab. Eur J Cancer. 2015;51:1812–21. doi: 10.1016/j.ejca.2015.05.016. [DOI] [PubMed] [Google Scholar]
  • 25.Stopeck AT, Lipton A, Body JJ, 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. 2010;28:5132–9. doi: 10.1200/JCO.2010.29.7101. [DOI] [PubMed] [Google Scholar]
  • 26.Munier A, Gras V, Andrejak M, et al. Zoledronic acid and renal toxicity: data from French adverse effect reporting database. Ann Pharmacother. 2005;39:1194–7. doi: 10.1345/aph.1E589. [DOI] [PubMed] [Google Scholar]
  • 27.Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int. 2008;74:1385–93. doi: 10.1038/ki.2008.356. [DOI] [PubMed] [Google Scholar]
  • 28.Smith MR, Halabi S, Ryan CJ, et al. Randomized clinical trial of early zoledronic acid in men with castration-sensitive prostate cancer and bone metastases: results of calgb 90202 (Alliance) J Clin Oncol. 2014;32:1143–50. doi: 10.1200/JCO.2013.51.6500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.James ND, Sydes MR, Clarke NW, et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (stampede): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet. 2016;387:1163–77. doi: 10.1016/S0140-6736(15)01037-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Djulbegovic B, Wheatley K, Ross J, et al. Bisphosphonates in multiple myeloma. Cochrane Database Syst Rev. 2002:CD003188. doi: 10.1002/14651858.CD003188. [DOI] [PubMed] [Google Scholar]
  • 31.Adami S. Bisphosphonates in prostate carcinoma. Cancer. 1997;80(suppl):1674–9. doi: 10.1002/(SICI)1097-0142(19971015)80:8+&#x0003c;1674::AID-CNCR18&#x0003e;3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
  • 32.Koizumi M, Yamada Y, Takiguchi T, et al. Bone metabolic markers in bone metastases. J Cancer Res Clin Oncol. 1995;121:542–8. doi: 10.1007/BF01197767. [DOI] [PubMed] [Google Scholar]
  • 33.Templeton AJ, Stalder L, Bernhard J, et al. Prevention of symptomatic skeletal events with denosumab administered every 4 weeks versus every 12 weeks: a noninferiority phase iii trial (sakk 96/12, reduse) [abstract TPS5095] J Clin Oncol. 2014;20 [Available online at: https://ascopubs.org/doi/abs/10.1200/jco.2014.32.15_suppl.tps5095; cited 17 July 2020] [Google Scholar]
  • 34.Amadori D, Aglietta M, Alessi B, et al. Efficacy and safety of 12-weekly versus 4-weekly zoledronic acid for prolonged treatment of patients with bone metastases from breast cancer (zoom): a phase 3, open-label, randomised, non-inferiority trial. Lancet Oncol. 2013;14:663–70. doi: 10.1016/S1470-2045(13)70174-8. [DOI] [PubMed] [Google Scholar]
  • 35.Hortobagyi GN, Van Poznak C, Harker WG, et al. Continued treatment effect of zoledronic acid dosing every 12 vs 4 weeks in women with breast cancer metastatic to bone: the optimize-2 randomized clinical trial. JAMA Oncol. 2017;3:906–12. doi: 10.1001/jamaoncol.2016.6316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shapiro CL, Moriarty JP, Dusetzina S, et al. Cost-effectiveness analysis of monthly zoledronic acid, zoledronic acid every 3 months, and monthly denosumab in women with breast cancer and skeletal metastases calgb 70604 (Alliance) J Clin Oncol. 2017;35:3949–55. doi: 10.1200/JCO.2017.73.7437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Palimeri C, Fullarton JR, Brown J. Comparative efficacy of bisphosphonates in metastatic breast and prostate cancer and multiple myeloma: a mixed-treatment meta-analysis. Clin Cancer Res. 2013;19:6863–72. doi: 10.1158/1078-0432.CCR-13-2275. [DOI] [PubMed] [Google Scholar]
  • 38.Rosen LS, Gordon D, Kaminski M, 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. 2001;7:377–87. [PubMed] [Google Scholar]
  • 39.Small EJ, Smith MR, Seaman JJ, Petrone S, Kowalski MO. Combined analysis of two multicenter, randomized, placebo-controlled studies of pamidronate disodium for the palliation of bone pain in men with metastatic prostate cancer. J Clin Oncol. 2003;21:4277–84. doi: 10.1200/JCO.2003.05.147. [DOI] [PubMed] [Google Scholar]
  • 40.Alibhai SMH, Zukotynski K, Walker-Dilks C, et al. Bone health and bone-targeted therapies for prostate cancer: a Programme in Evidence-Based Care–Cancer Care Ontario clinical practice guideline. Clin Oncol (R Coll Radiol) 2017;29:348–55. doi: 10.1016/j.clon.2017.01.007. [DOI] [PubMed] [Google Scholar]
  • 41.Saylor PJ, Rumble RB, Tagawa S, et al. Bone health and bone-targeted therapies for prostate cancer: ASCO endorsement of a Cancer Care Ontario guideline. J Clin Oncol. 2020;38:1736–43. doi: 10.1200/JCO.19.03148. [DOI] [PubMed] [Google Scholar]
  • 42.Khan SA, Kanis JA, Vasikaran S, et al. Elimination and biochemical responses to intravenous alendronate in postmenopausal osteoporosis. J Bone Miner Res. 1997;12:1700–7. doi: 10.1359/jbmr.1997.12.10.1700. [DOI] [PubMed] [Google Scholar]
  • 43.Awan AA, Hutton B, Hilton K, et al. De-escalation of bone-modifying agents in patients with bone metastases from breast cancer: a systematic review and meta-analysis. Breast Cancer Res Treat. 2019;176:507–17. doi: 10.1007/s10549-019-05265-1. [DOI] [PubMed] [Google Scholar]
  • 44.Clemons M, Stober C, Mates M, Joy AA, Robinson A, Hilton J. A pragmatic, randomised, multicentre trial comparing 4-weekly vs 12-weekly administration of bone-targeted agents (denosumab, zoledronate, or pamidronate) in patients with bone metastases. Ann Oncol. 2019;30(suppl 3):iii72–3. doi: 10.1093/annonc/mdz118.002. [DOI] [Google Scholar]
  • 45.Clemons M, Ong M, Stober C, et al. A randomized trial comparing four-weekly versus 12-weekly administration of bone-targeted agents (denosumab, zoledronate, or pamidronate) in patients with bone metastases from either breast or castration-resistant prostate cancer [abstract 11501] J Clin Oncol. 2019;37 doi: 10.1200/JCO.2019.37.15_suppl.11501. [Available online at: https://ascopubs.org/doi/abs/10.1200/JCO.2019.37.15_suppl.11501; cited 19 July 2020] [DOI] [Google Scholar]
  • 46.Ross JR, Saunders Y, Edmonds PM, Patel S, Broadley KE, Johnston SR. Systematic review of role of bisphosphonates on skeletal morbidity in metastatic cancer. BMJ. 2003;327:469. doi: 10.1136/bmj.327.7413.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jacobs C, Kuchuk I, Bouganim N, et al. A randomized, double-blind, phase ii, exploratory trial evaluating the palliative benefit of either continuing pamidronate or switching zoledronic acid in patients with high-risk bone metastases from breast cancer. Breast Cancer Res Treat. 2016;155:77–84. doi: 10.1007/s10549-015-3646-2. [DOI] [PubMed] [Google Scholar]

Articles from Current Oncology are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES