2,841
Views
39
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

The use of bisphosphonates in cancer patients

, &
Pages 581-591 | Received 22 Sep 2007, Published online: 08 Jul 2009

Abstract

Skeletal-related events resulting from bone metastases or osteoporosis can significantly contribute to morbidity and mortality in cancer patients. Expert opinion on the effectiveness of bisphosphonates in this setting is evolving. Here we review current evidence on the risks and benefits of bisphosphonate therapy for a wide variety of cancers, as well as clinical management of its adverse effects. A MEDLINE search of English-language literature (1966 through May 2006) was conducted using the terms bisphosphonate, cancer, multiple myeloma, malignancy, and randomized controlled clinical studies. Studies were selected based on clinical pertinence, with an emphasis on phase III clinical trials. We reviewed bibliographies for other relevant articles. Accumulating evidence reveals that bisphosphonate therapy has a significant effect in preventing skeletal complications in multiple myeloma, breast cancers, and prostate cancer, and in reducing skeletal complications in other metastatic bone malignancies. Emerging data indicate that bisphosphonates are useful for preventing bone loss resulting from cancer or its therapy. The efficacy of bisphosphonates for early-stage breast cancers remains controversial. Significant risks of bisphosphonate therapy include nephrotoxicity, electrolyte abnormalities, and osteonecrosis of the jaw. Bisphosphonate therapy has a clear role in the management of skeletal metastases associated with a variety of cancers. However, significant side effects require ongoing monitoring and treatment.

Bone metastases lead to significant morbidity and mortality in cancer patients. They are associated with almost every type of malignant tumor, but most frequently with prostate, breast, and lung cancers () Citation[1]. Even though bone metastases are often categorized into osteoblastic (prostate cancer), osteolytic (myeloma), and mixed types (breast cancer) according to radiographic appearances, the underlying pathogenesis regularly involves both osteoblastic and osteolytic processes Citation[2]. In addition, primary or secondary osteoporosis is often comorbid in cancer patients, leading to significantly increased risk of fracture. Numerous factors contribute to cancer-related osteoporosis, including radiation, cytotoxic agents, gonadotropin-releasing hormone analogs, and aromatase inhibitors Citation[3]. While the mechanism for bone metastasis and osteoporosis is different, osteoclastic activity is known to play an important role in both processes.

Table I.  Incidence of bone metastases in cancer patients with metastatic disease Citation[1], Citation[78], Citation[79].

Bisphosphonates, which are analogs of endogenous pyrophosphates with the common structural element of a phosphorus-carbon-phosphorus (P-C-P) bond Citation[4], have been used extensively to treat and prevent osteoporosis, and to treat Paget's disease and hypercalcemia of malignancy () Citation[5], Citation[6]. Bisphosphonates have high affinity for bone. They bind to hydroxyapatite crystals on bone surfaces, where they are resorbed by osteoclasts. After they are internalized by osteoclasts through endocytosis, they reduce the activity of those osteoclasts and thus inhibit bone resorption. Each bisphosphonate has unique side chains (R1 and R2) attached to the central carbon atom. The R1 chain and the P-C-P backbone determine its binding to bone minerals, while the R2 side chain determines the antiresorptive potency, side effects, probably the mechanism of action, and possibly antineoplastic activity. Bisphosphonates with R2 side chains containing a primary nitrogen are more potent than nonnitrogen counterparts () Citation[7] and can inhibit prenylation of GTP-binding proteins by farnesyl diphosphonate synthetase, which is critical for osteoclast function and survival Citation[8]. Nonnitrogen-containing bisphosphonates are incorporated into nonhydrolysable cytotoxic analogues of ATP Citation[9]. In cancer patients, bisphosphonates may affect bone microenvironment to reduce the invasion of tumor cells Citation[10], thus delaying bone metastases in high-risk early-stage cancers. They may also have direct tumor cytotoxicity Citation[11–13] and antiangiogenic activity Citation[14–16]. However, the biological diversity of cancers and their bone metastases may result in different responses to bisphosphonate therapy. Furthermore, bisphosphonate treatment may expose patients to serious side effects that primary care physicians should recognize. We here review existing evidence regarding the benefits and risks of bisphosphonate use in the contemporary care of patients with various cancers.

Table II.  Bisphosphonates in clinical use Citation[7].

Methods

We performed MEDLINE searches of the English-language literature (1966 through May 2006) using the terms bisphosphonate, cancer, multiple myeloma, malignancy, and randomized controlled clinical studies. We manually reviewed relevant bibliographies for additional material. We obtained further information from presentations at the 2006 American Society of Clinical Oncology (ASCO) Meeting and 2006 Prostate Cancer Symposium, and reviewed published guidelines from the National Comprehensive Cancer Network and ASCO. In evaluating the benefits of bisphosphonate therapy, we focused on phase III randomized controlled trials. On review of clinical trials, endpoints were prevention or delay of skeletal complications and palliation of bone pain. No studies have been designed to evaluate survival as a primary endpoint for bisphosphonate therapy.

Efficacy of bisphosphonate therapy

Bisphosphonate therapy was initially evaluated in 1996 in large randomized controlled phase III clinical trials for multiple myeloma and breast cancer Citation[17], Citation[18], then in 2002 and 2003 for prostate and many other malignancies Citation[19], Citation[20]. To date, the use of bisphosphonates has been demonstrated to reduce skeletal-related events (SREs) in many malignancies, including multiple myeloma, breast, prostate, and other solid tumors (); however, the impact of bisphosphonate treatment on overall survival has been very limited for patients with metastatic cancers in these clinical trials Citation[17–20]. Thus, the main focus of this review is the role of bisphosphonates in reducing skeletal complications of malignancies, as well as potential toxicities of bisphosphonates.

Table III.  Major randomized double-blind placebo-controlled clinical studies of bisphosphonate therapy in metastatic bone disease, selected based on their importance in the clinical development of bisphosphonate therapy.

Multiple myeloma

Multiple myeloma is the third most common hematologic malignancy in the United States. At the time of diagnosis, nearly 80% of patients have osteolytic lesions, osteoporosis, or fractures Citation[21]. Pamidronate, zoledronic acid, clodronate, and ibandronate have been evaluated in the treatment of myeloma. In a trial performed by Myeloma Aredia Study Group, 392 patients with advanced multiple myeloma were randomized to placebo or pamidronate (90 mg as a 4-hour iv infusion) every 4 weeks for 21 cycles Citation[17], Citation[22]. The primary endpoints were reduction of SREs and evaluation of safety and survival after 9 and 21 cycles of therapy. SREs were defined as pathologic fractures, irradiation of or surgery to bone, and spinal cord compression. At the 9-month follow-up, the mean number of SREs per year was reduced from 2.1 in the placebo group to 1.1 in the pamidronate group (p = 0.0006). At the 21-month follow-up, the number of SREs remained stable at 2.2 per year in the placebo group and 1.3 per year in the pamidronate group (p = 0.008). In addition, median time to the first SRE was 10 months in the placebo group and 21 months in the pamidronate group (p < 0.001).

A large phase III randomized trial reported by Rosen et al. compared 4 or 8 mg of zoledronic acid to 90 mg of pamidronate iv every 3–4 weeks Citation[23]. The trial included 1 648 eligible patients, of whom 513 had multiple myeloma. As increases in creatinine occurred more often among patients randomized to 8 mg zoledronic acid, the dose was reduced to 4 mg. Median time to first SRE was not statistically different between the 4 mg zoledronic acid and pamidronate treatment groups (380 days vs. 286 days, p = 0.538). Thus, this trial met its primary endpoint of demonstrating non-inferiority of zoledronic acid to pamidronate.

An MRC study evaluated clodronate in 536 patients with newly diagnosed multiple myeloma who received either oral clodronate (1 600 mg/day) (n = 264) or placebo (n = 272) Citation[24]. Clodronate was associated with significantly lower incidence of vertebral (38% vs. 55%, p = 0.01) and nonvertebral fractures (6.8% vs. 13.2%, p = 0.04). A smaller benefit was noted in an earlier Finnish multicenter trial (n = 350) in which clodronate was associated with a significantly lower rate of radiological skeletal progression at 24 months, but no improvement in fracture incidence or pain relief Citation[25].

Ibandronate has not been demonstrated to be effective in reducing SREs in advanced myeloma. Stage II or III myeloma patients (n = 214) receiving chemotherapy were randomly assigned to receive monthly iv ibandronate at 2 mg or placebo for 24 months in a study performed by Menssen et al. Citation[26]. Neither incidence nor time to first SRE differed significantly between the 2 groups.

As there have been no direct comparisons between clodronate and pamidronate or clodronate and zoledronic acid, it is unclear which bisphosphonate is more effective in multiple myeloma. ASCO guidelines recommend only pamidronate and zoledronic acid because studies have more thoroughly assessed skeletal complications with their use. Clodronate is not available in the USA Citation[27].

Breast cancer

Bone metastases from breast cancer encompass both osteolytic and osteoblastic lesions. Bisphosphonates have been studied extensively in metastatic breast cancer. In women with osteolytic metastatic breast cancer on systemic chemotherapy (n = 382), iv pamidronate (90 mg) monthly for 2 years delayed the appearance of clinical skeletal complications, with a median time to first SRE of 14 months vs. 7 months with placebo (p < 0.001), as reported by Protocol 19 Aredia Breast Cancer Study Group Citation[18], Citation[28]. In women with metastatic osteolytic breast cancer (n = 372) on concurrent hormonal therapy, pamidronate was associated with prolonged time to first SRE (median 10.4 months vs. 6.9 months, p = 0.049) and reduced skeletal complications at 24 months (56% vs. 67%, p = 0.027) Citation[29]. Kohno et al. observed a similar effect of zoledronic acid on reducing skeletal complications in Japanese women with metastatic breast cancer Citation[30]. Finally, Body et al. found that ibandronate at 6 mg every 3–4 weeks reduced the risk of SREs by 38% over a 12-week period in breast cancer patients with bone metastases Citation[31].

A phase III double-blind study by Rosen et al. compared zoledronic acid to pamidronate in the treatment of metastatic bone disease of breast cancer (n = 1 130) Citation[23]. The proportion of patients who experienced SREs was similar between zoledronic acid and pamidronate in the breast cancer chemotherapy stratum (RR = 0.945, p = 0.745); however, it was associated with 30% additional risk reduction of SREs in the breast cancer hormonal therapy stratum in comparison with pamidronate (RR = 0.693, p = 0.009) Citation[32].

A pooled analysis by Cochrane Database of Systematic Reviews showed that oral bisphosphonates, including pamidronate and clodronate or ibandronate, reduced the risk of SREs by 16% (p = 0.001) in breast cancer patients with metastatic bone disease Citation[33].

Three studies have evaluated the use of clodronate and pamidronate in advanced breast cancer without clinically evident bone metastases Citation[34–36]. There was no significant reduction in incidence of SREs (RR 0.99, p = 0.97) by meta-analyses Citation[33].

The utility of bisphosphonates as an adjuvant treatment for breast cancer remains controversial. In 3 studies of oral clodronate that included 1 653 women with early-stage breast cancer but high risk for metastatic disease, there was no significant reduction in the risk of developing skeletal metastases (RR 0.82, p = 0.07) Citation[33]. However, there was evidence of improved survival (RR 0.82, p = 0.02), a finding that has prompted a phase III National Surgical Adjuvant Breast and Bowel Project (NSABP) trial comparing clodronate with zoledronate or ibandronate as adjuvants for women with resected stage I–III breast cancer.

Women undergoing treatment for breast cancer are at greater risk of bone loss as a result of decreased estrogen levels associated with therapy or postmenopausal status. Aromatase inhibitors including anastrozole, letrozole, and exemestane have a pronounced effect on bone mineral density, and are associated with a higher incidence of osteoporosis/osteopenia as demonstrated in several breast cancer adjuvant trials Citation[37–39]. Early results from several large randomized trials indicate that zoledronic acid may reduce bone loss associated with aromatase inhibitors Citation[40]. Adjuvant chemotherapy may be associated with premature menopause and early bone loss in premenopausal women. Short-term intermittent use of clodronate has not been shown to prevent bone loss in this setting Citation[41].

Bisphosphonates may have an analgesic effect in women with painful bone metastases secondary to breast cancer. Seven of 12 placebo-controlled trials showed a modest reduction in metastatic bone pain with bisphosphonates. The effect of bisphosphonates on patient-rated quality of life is not evident Citation[33].

In summary, bisphosphonates reduce the risk of skeletal complications and may also reduce pain in patients with breast cancer metastatic to the bone. ASCO guidelines recommend iv pamidronate or zoledronic acid for women with radiographic evidence of lytic metastatic bone disease Citation[42]. The utility of bisphosphonates in early breast cancer requires further study.

Prostate cancer

Bone metastases from prostate cancer are predominantly osteoblastic; however, there is increased osteoclastic activity associated with metastasis or androgen-deprivation therapy (ADT). Saad et al. evaluated zoledronic acid in a randomized placebo-controlled trial that enrolled 643 men with hormone-refractory prostate cancer and metastatic bone lesions Citation[19]. Patients received placebo or iv zoledronic acid at 4 mg or 8 mg (8 mg was subsequently reduced to 4 mg due to a high incidence of renal function deterioration) every 3 weeks for 15 months. A significant reduction in the frequency of SREs was found in men receiving zoledronic acid 4 mg compared with placebo (33% vs. 44%). Moreover, there was a 41% decrease in pathologic fractures seen with 4 mg zoledronic acid compared to placebo (13.1% vs. 22.1%, p = 0.015). At 24 months follow-up, reduction of SREs was stable (38% vs. 49%) and median time to develop an SRE was significantly longer in the treated group (488 days vs. 321 days). However, the therapy had little impact on pain, quality of life, or survival Citation[43]. In contrast, pamidronate and clodronate have shown no significant clinical benefit in men with metastatic prostate cancer Citation[44–46].

Clodronate was not effective in delaying bone metastasis in patients with nonmetastatic prostate cancers. In the MRC PR04 trial of 508 men with T2-4 prostate cancers on standard therapy, clodronate at 2 080 mg/day for up to 5 years was compared to placebo, with a primary endpoint of bone progression-free survival (BPFS), defined as the time to development of symptomatic bone metastasis or to death from prostate cancer Citation[47]. There was a trend to decreased BPFS in the clodronate arm (HR = 1.29, p = 0.13).

Bisphosphonates are also effective in preventing bone loss associated with ADT. In patients with metastatic and nonmetastatic prostate cancer, both pamidronate and zoledronic acid showed clear benefit in maintaining or improving bone mineral density compared with placebo, as reported by Smith et al. Citation[48], Citation[49]. Recent reports at the 2006 Prostate Cancer Symposium and ASCO Annual Meeting showed that weekly oral alendronate or annual zoledronic acid can prevent bone loss in men with nonmetastatic prostate cancer on ADT Citation[50], Citation[51].

In short, zoledronic acid is the only bisphosphonate that has been shown to reduce SREs in men with androgen-independent metastatic prostate cancer, and the only bisphosphonate approved for this use by the FDA. Pamidronate, zoledronic acid, and alendronate mitigate bone loss associated with ADT.

Other solid tumors

Few studies have been performed to evaluate the effect of bisphosphonates in metastatic cancers other than multiple myeloma and breast and prostate cancers. Zoledronic acid is the only bisphosphonate that has been evaluated in a wide variety of metastatic solid tumors. A randomized phase III double-blind placebo-controlled clinical trial (n = 733) by Rosen et al. evaluated zoledronic acid in various metastatic solid cancers other than breast and prostate cancers, including lung (50% non-small cell lung cancer and 8% small cell lung cancer), renal (10%), unknown primary (7%), thyroid (2%), and head and neck (2%) cancers Citation[20], Citation[52]. In comparison with placebo, 4 mg iv zoledronic acid significantly decreased the risk of SREs (38% vs. 47%, p = 0.039). Zoledronic acid also prolonged median time to first SRE by more than 2 months (p = 0.007). However, when hypercalcemia of malignancy was excluded from the analysis, the primary endpoint of reducing SREs did not reach statistical significance.

Due to the diversity of tumor types in the above-mentioned trial, the actual benefit of zoledronic acid in reducing skeletal complications of metastatic bone disease in an individual malignancy is not clear. However, it might not be feasible to evaluate the efficacy of zoledronic acid in all of these individual tumors. Despite the absence of direct evidence, bisphosphonate therapy may still be appropriate for patients with metastatic bone disease from solid tumors other than breast and prostate cancers, considering the significant benefit of bisphosphonate therapy in breast and prostate cancers, which encompass a wide range of osteolytic and osteoblastic lesions.

Risks and adverse effects of bisphosphonates

Bisphosphonates are generally well tolerated. However, significant side effects such as acute-phase reactions, renal toxicity, electrolyte abnormalities, and osteonecrosis of the jaw should be monitored regularly and treated in a timely manner.

Acute-phase reactions

Acute-phase reactions are self-limiting flu-like symptoms, including low-grade fever, arthralgia/myalgia, nausea, and increased bone pain. This is the most common adverse event, occurring in up to 18% of patients treated with either zoledronic acid or pamidronate Citation[23]. Symptoms occur within 24 hours after the first infusion, persist for less than 48 hours, and usually diminish or disappear by the second or third infusion. Management includes acetaminophen and fluids for flu-like symptoms and antiemetics for nausea.

Renal toxicity

Elevation of serum creatinine during bisphosphonate therapy is uncommon but potentially serious. Approximately 10% of patients treated with 4 mg zoledronic acid or 90 mg pamidronate experience deterioration of renal function (defined as an increase of 0.5 mg/dL in patients with normal baseline serum creatinine ≤ 1.4 mg/dL or an increase of 1.0 mg/dL in patients with baseline serum creatinine > 1.4 mg/dL) Citation[23]. Prolonged use of bisphosphonates (>2 years) is associated with notable serum creatinine increase in 12.5% of patients Citation[53]. Underlying risk factors for renal failure include multiple myeloma, diabetes mellitus, hypertension, advanced age (>65 years old), previous bisphosphonate exposure, and the use of NSAIDs. For zoledronic acid, renal impairment is also associated with 8-mg doses and infusion duration less than 15 minutes Citation[23], Citation[52]. The underlying tumor type may also contribute to renal failure. Most cases (42/72) have been reported in patients with multiple myeloma, but many cases (22/72) have been reported in patients with solid tumors Citation[54]. The impact of hydration status on the development of acute renal failure needs further study. The mechanism for bisphosphonate-associated renal failure is not well understood, but some case reports have suggested a link with acute tubular necrosis Citation[55].

Nephrotic syndrome was reported in 7 patients with multiple myeloma (n = 6) or breast cancer (n = 1) receiving pamidronate therapy Citation[56], Citation[57]. These patients had been treated with pamidronate for 15–48 months; 5 had received more than the standard dose of 90 mg per month. Average peak urine protein excretion was 12.4 g/day. Renal biopsy revealed collapsing focal segmental glomerulosclerosis (FSGS). FSGS is rare, occurring most commonly in African Americans with HIV infection. The study patients were white and HIV-negative, suggesting a possible association between pamidronate and FSGS. Pamidronate was stopped in 5 patients, with 2 requiring dialysis and 3 having stable renal insufficiency. Pamidronate was continued in 2 patients, both of whom required maintenance dialysis.

During iv bisphosphonate therapy, patients may need to avoid agents with potential renal side effects, such as radiographic contrast reagents and NSAIDs, due to the risk of nephrotoxicity. If these agents must be used in conjunction with bisphosphonates, they should ideally be given on different days, and patients’ renal function should be regularly assessed. Imaging studies with contrast could be carried out if renal function is adequate. In the presence of baseline renal insufficiency, the dose of bisphosphonates should be adjusted based on creatinine clearance, as per manufacturer recommendations () Citation[58]. However, for many bisphosphonates, optimal dosages in this situation have not been determined. In accordance with FDA labeling, ASCO guidelines recommend that creatinine be monitored prior to each dose of pamidronate or zoledronate Citation[42]. Unexplained azotemia should prompt temporary discontinuation of the bisphosphonate. Patients should be reassessed every 3–4 weeks. If renal function returns to baseline, therapy may be cautiously reinstituted.

Table IV.  Bisphosphonate dosing recommendations for patients with reduced baseline renal function.

Electrolyte abnormalities

Serum ion fluctuations, including hypocalcemia, hypophosphatemia, and hypermagnesemia, may occur during bisphosphonate therapy. They are usually mild but may have serious consequences such as seizure and tetany. Hypophosphatemia is most common during treatment of hypercalcemia of malignancy and can be present in 12.3% of patients receiving zoledronic acid infusion Citation[58]. Incidence of hypocalcemia varies according to malignancy. In studies of breast cancer patients, hypocalcemia was associated with 39% of patients treated with zoledronic acid, compared with 7% in the placebo arm Citation[30]. Hypocalcemia develops as a result of a deficient compensatory mechanism, such as prior parathyroidectomy, hypomagnesemic hypoparathyroidism or low vitamin D levels, and parathyroid hormone (PTH) resistance. Loop diuretics may further worsen hypocalcemia. Hypocalcemia is rarely symptomatic. Classic presentations include perioral paraesthesia, carpopedal spasm, tetany, and QT prolongation. Vague symptoms such as weakness and tingling may occur Citation[59]. Hypocalcemia may also promote PTH secretion and cause or aggravate secondary hyperparathyroidism Citation[60].

Vitamin D and calcium supplements are strongly recommended to minimize the incidence of hypocalcemia during bisphosphonate therapy. Any preexisting electrolyte abnormalities should be corrected before starting bisphosphonate treatment, and serum magnesium, calcium, and phosphate should be monitored periodically. Treatment of electrolyte abnormalities varies with severity and may necessitate iv replacement.

Osteonecrosis of the jaw

Osteonecrosis of the jaw (ONJ), also called avascular necrosis of the jaw, refers to the death of bone tissue in the jaw, probably due to impaired blood supply. The typical presenting lesion is a painful nonhealing ulcer, extraction socket, or exposed jawbone that has not responded to conservative debridement and antibiotic treatment. Prolonged use of bisphosphonates has been associated with ONJ in 5–6.7% of patients treated with bisphosphonates for bone metastases Citation[53], Citation[61]. Actual incidence may vary depending on the underlying malignancy, duration of bisphosphonate treatment, and clinician awareness. One series included 36 ONJ patients who received pamidronate or zoledronate for myeloma or breast cancer Citation[62]. A second series included 63 ONJ patients who received either iv bisphosphonates for at least one year (n = 56) or chronic oral bisphosphonates Citation[63]. Other notable cases have also been reported Citation[64–67]. ONJ may be a class effect of bisphosphonate therapy, occurring most commonly in second- and third-generation bisphosphonates. The FDA conducted a postmarketing safety review in 2004, and a search of the Adverse Event Reports database revealed 139 cases of bisphosphonate-associated ONJ: 47 patients taking pamidronate, 33 taking zoledronic acid, and 59 taking both drugs. Fewer cases were identified in patients taking alendronate or risedronate (n = 12 and 1 respectively). Possible risk factors of ONJ include length of exposure to bisphosphonates Citation[61], a diagnosis of cancer, concomitant therapies (e.g., chemotherapy, radiotherapy, corticosteroids), and comorbid conditions such as anemia, coagulopathies, infection, and preexisting oral disease Citation[68]. Previous dental procedures may be a precipitating factor. Possible mechanisms for bisphosphonate-associated ONJ include a compromised repair resulting from suppressed bone metabolism and angiogenesis secondary to bisphosphonates, and increased damage due to trauma or infection Citation[69].

As no effective treatments have been established in prospective studies, the best approach to bisphosphonate-associated ONJ is prevention. Before initiating bisphosphonate therapy, it is recommended that patients be referred to a dentist familiar with existing guidelines for the prevention of oral complications of cancer therapy Citation[70]. Newly issued postmarketing guidelines for zoledronic acid and pamidronate also stress the importance of this regimen. Although the optimal waiting period for instituting bisphosphonate therapy following invasive dental procedures has not been determined, the standard of care prior to radiation therapy is a delay of at least 10 days to allow for healing. If possible, invasive dental procedures such as tooth extractions and bone biopsies should be postponed until after iv bisphosphonate therapy. Once ONJ develops, it is advisable to discontinue bisphosphonates until the lesions resolve, even though doing so may not actually promote healing of the lesions. Bisphosphonates should be reinstituted only if clinically indicated and after careful consideration of risks and benefits. Prompt referral to a dentist trained in oral medicine, a hospital dentist, or an oral/maxillofacial surgeon is recommended for patients with symptoms of ONJ. Conservative management includes culturing of any lesions, prescribing antibiotics as appropriate, and recommending an antiseptic oral rinse that contains chlorhexidine gluconate. Some patients will need hyperbaric treatments, debridement or resection of portions of the jaw, and subsequent reconstructive surgery.

Areas of controversy

Duration and frequency of bisphosphonate therapy

Data on the optimal duration of bisphosphonate therapy are limited. For multiple myeloma, the single randomized study was continued for 21 cycles. It has been recommended that therapy be continued even if the patient has an SRE such as compression fracture or pathologic fracture Citation[43]. Current ASCO guidelines recommend that bisphosphonate therapy be continued as long as it is well tolerated, or until there is a significant decrease in performance status in patients with multiple myeloma or bone metastasis from breast cancer Citation[27], Citation[42], Citation[71]. There are no consensus guidelines for the duration of bisphosphonate therapy in patients with prostate cancer. However, recommendations from a multidisciplinary panel suggest that bisphosphonate treatments should be indefinite in patients with androgen-independent prostate cancer and bone metastasis. This is supported by evidence that efficacy does not decrease during long-term bisphosphonate treatment Citation[43]. No formal recommendations have been published for patients with bone metastases from solid tumors other than breast or prostate cancer.

Pharmacoeconomic issues

The widespread use of bisphosphonates will have a major impact on pharmaceutical expenses. Costs will vary depending on the duration of bisphosphonate use, the specific bisphosphonate used, and how the bisphosphonate is delivered (oral vs. iv). Using Markov models, Ross et al. estimated that the overall cost of bisphosphonate therapy to prevent an SRE was GBP 250 and GBP 1 500 per event for patients with breast cancer and multiple myeloma respectively Citation[72]. Hillner et al. found cost-effective ratios associated with adding pamidronate of USD 108 200 per quality-adjusted life year in women concurrently treated with chemotherapy, and USD 305 300 per quality-adjusted life year in women treated with hormonal therapy Citation[73]. The cost of preventing one SRE was USD 9 350 in the chemotherapy cohort and USD 12 760 in the hormonal therapy cohort. These ratios are much higher than most routinely accepted medical therapies. However, a Canadian analysis concluded that pamidronate's incremental cost was USD 18 700 per quality-adjusted life year gained Citation[74]. The major differences in these results are explained primarily by: 1) the fact that the cost of pamidronate per treatment is approximately 50% lower in Canada than in the United States, and 2) by an assumption that a nonvertebral fracture would require in-patient treatment. The cost of treatment could be reduced by introducing bisphosphonate home therapy as part of long-term therapy Citation[75]. In a multicenter trial, patients with multiple myeloma (n = 37) were randomly allocated to receive 3 months of in-home treatment with iv pamidronate, followed by 3 months of in-hospital treatment with pamidronate or vice versa Citation[76]. Home therapy with pamidronate appeared feasible and safe. Similar results have been demonstrated with iv zoledronic acid Citation[77].

Conclusions

Accumulating evidence reveals that bisphosphonate therapy plays a significant role in preventing skeletal complications in patients with bone metastases from multiple myeloma, breast cancers, and hormone-refractory prostate cancers. In addition, zoledronic acid reduces skeletal complications in a wide variety of other metastatic malignancies, and emerging data suggest that bisphosphonates are useful for treatment-related bone loss in prostate cancer. The efficacy of bisphosphonates as adjuvants in the treatment of early-stage breast or prostate cancers has not been clarified. Significant side effects of long-term bisphosphonate therapy include renal toxicity, hypocalcemia, and osteonecrosis of the jaw. With proper vigilance, bisphosphonate therapy is safe and can have substantial benefits for cancer patients.

Acknowledgements

This research was supported in part by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research. We would like to thank Stan Appleton, DDS for providing expert advice in the preparation of this manuscript.

References

  • Coleman RE. Metastatic bone disease: Clinical features, pathophysiology and treatment strategies. Cancer Treat Rev 2001; 27: 165–76
  • Roodman GD. Mechanisms of bone metastasis. N Engl J Med 2004; 350(16)1655–64
  • Pfeilschifter J, Diel IJ. Osteoporosis due to cancer treatment: Pathogenesis and management. J Clin Oncol 2000; 18: 1570–93
  • Fleisch H. Bisphosphonates–history and experimental basis. Bone 1987; 8(Suppl 1)S23–8
  • Gennari C, Nuti R, Agnusdei D, Camporeale A, Martini G. Management of osteoporosis and Paget's disease. An appraisal of the risks and benefits of drug treatment. Drug Saf 1994; 11: 179–95
  • Body JJ. Bisphosphonates for malignancy-related bone disease: Current status, future developments. Support Care Cancer 2006; 14: 408–18
  • Licata AA. Discovery, clinical development, and therapeutic uses of bisphosphonates. Ann Pharmacother 2005; 39: 668–77
  • Luckman SP, Hughes DE, Coxon FP, Graham R, Russell G, Rogers MJ. Nitrogen-containing bisphosphonates inhibit the mevalonate pathway and prevent post-translational prenylation of GTP-binding proteins, including Ras. J Bone Miner Res 1998; 13: 581–9
  • Frith JC, Monkkonen J, Blackburn GM, Russell RG, Rogers MJ. Clodronate and liposome-encapsulated clodronate are metabolized to a toxic ATP analog, adenosine 5′-(beta, gamma-dichloromethylene) triphosphate, by mammalian cells in vitro. J Bone Miner Res 1997; 12: 1358–67
  • Woodward JK, Coleman RE, Holen I. Preclinical evidence for the effect of bisphosphonates and cytotoxic drugs on tumor cell invasion. Anticancer Drugs 2005; 16: 11–9
  • Riebeling C, Forsea AM, Raisova M, Orfanos CE, Geilen CC. The bisphosphonate pamidronate induces apoptosis in human melanoma cells in vitro. Br J Cancer 2002; 87: 366–71
  • Witters LM, Crispino J, Fraterrigo T, Green J, Lipton A. Effect of the combination of docetaxel, zoledronic acid, and a COX-2 inhibitor on the growth of human breast cancer cell lines. Am J Clin Oncol 2003; 26: S92–7
  • Matsumoto S, Kimura S, Segawa H, Kuroda J, Yuasa T, Sato K, et al. Efficacy of the third-generation bisphosphonate, zoledronic acid alone and combined with anti-cancer agents against small cell lung cancer cell lines. Lung Cancer 2005; 47: 31–9
  • Fournier P, Boissier S, Filleur S, Guglielmi J, Cabon F, Colombel M, et al. Bisphosphonates inhibit angiogenesis in vitro and testosterone-stimulated vascular regrowth in the ventral prostate in castrated rats. Cancer Res 2002; 62: 6538–44
  • Wood J, Bonjean K, Ruetz S, Bellahcene A, Devy L, Foidart JM, et al. Novel antiangiogenic effects of the bisphosphonate compound zoledronic acid. J Pharmacol Exp Ther 2002; 302: 1055–61
  • Ferretti G, Fabi A, Carlini P, Papaldo P, Cordiali Fei P, Di Cosimo S, et al. Zoledronic-acid-induced circulating level modifications of angiogenic factors, metalloproteinases and proinflammatory cytokines in metastatic breast cancer patients. Oncology 2005; 69: 35–43
  • Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA, Bordoni R, George S, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. Myeloma Aredia Study Group. N Engl J Med 1996; 334: 488–93
  • Hortobagyi GN, Theriault RL, Porter L, Blayney D, Lipton A, Sinoff C, et al. Efficacy of pamidronate in reducing skeletal complications in patients with breast cancer and lytic bone metastases. Protocol 19 Aredia Breast Cancer Study Group. N Engl J Med 1996; 335: 1785–91
  • Saad F, Gleason DM, Murray R, Tchekmedyian S, Venner P, Lacombe L, et al. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst 2002; 94: 1458–68
  • Rosen LS, Gordon D, Tchekmedyian S, Yanagihara R, Hirsh V, Krzakowski M, et al. Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial–the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group. J Clin Oncol 2003; 21: 3150–7
  • Kyle RA. Multiple myeloma: Review of 869 cases. Mayo Clin Proc 1975; 50: 29–40
  • Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA, Bordoni R, George S, et al. Long-term pamidronate treatment of advanced multiple myeloma patients reduces skeletal events. Myeloma Aredia Study Group. J Clin Oncol 1998; 16: 593–602
  • Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey J, 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
  • McCloskey EV, MacLennan IC, Drayson MT, Chapman C, Dunn J, Kanis JA. A randomized trial of the effect of clodronate on skeletal morbidity in multiple myeloma. MRC Working Party on Leukaemia in Adults. Br J Haematol 1998; 100: 317–25
  • Lahtinen R, Laakso M, Palva I, Virkkunen P, Elomaa I. Randomised, placebo-controlled multicentre trial of clodronate in multiple myeloma. Finnish Leukaemia Group. Lancet 1992; 340(8827)1049–52
  • Menssen HD, Sakalova A, Fontana A, Herrmann Z, Boewer C, Facon T, et al. Effects of long-term intravenous ibandronate therapy on skeletal-related events, survival, and bone resorption markers in patients with advanced multiple myeloma. J Clin Oncol 2002; 20: 2353–9
  • Berenson JR, Hillner BE, Kyle RA, Anderson K, Lipton A, Yee GC, et al. American Society of Clinical Oncology clinical practice guidelines: The role of bisphosphonates in multiple myeloma. J Clin Oncol 2002; 20: 3719–36
  • Hortobagyi GN, Theriault RL, Lipton A, Porter L, Blayney D, Sinoff C, et al. Long-term prevention of skeletal complications of metastatic breast cancer with pamidronate. Protocol 19 Aredia Breast Cancer Study Group. J Clin Oncol 1998; 16: 2038–44
  • Theriault RL, Lipton A, Hortobagyi GN, Leff R, Gluck S, Stewart JF, et al. Pamidronate reduces skeletal morbidity in women with advanced breast cancer and lytic bone lesions: a randomized, placebo-controlled trial. Protocol 18 Aredia Breast Cancer Study Group. J Clin Oncol 1999; 17: 846–54
  • Kohno N, Aogi K, Minami H, Nakamura S, Asaga T, Iino Y, 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 2005; 23: 3314–21
  • Body JJ, Diel IJ, Lichinitser MR, Kreuser ED, Dornoff W, Gorbunova VA, et al. Intravenous ibandronate reduces the incidence of skeletal complications in patients with breast cancer and bone metastases. Ann Oncol 2003; 14: 1399–405
  • Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey J, et al. Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: A randomized, double-blind, multicenter, comparative trial. Cancer 2003; 98: 1735–44
  • Pavlakis, N, Schmidt, R, Stockler, M. Bisphosphonates for breast cancer. Cochrane Database Syst Rev. 2005: CD003474.
  • van Holten-Verzantvoort AT, Hermans J, Beex LV, Blijham G, Cleton FJ, van Eck-Smit BC, et al. Does supportive pamidronate treatment prevent or delay the first manifestation of bone metastases in breast cancer patients?. Eur J Cancer 1996; 32A: 450–4
  • Kanis JA, Powles T, Paterson AH, McCloskey EV, Ashley S. Clodronate decreases the frequency of skeletal metastases in women with breast cancer. Bone 1996; 19: 663–7
  • Mardiak J, Bohunicky L, Chovanec J, Salek T, Koza I. Adjuvant clodronate therapy in patients with locally advanced breast cancer–long term results of a double blind randomized trial. Slovak Clodronate Collaborative Group. Neoplasma 2000; 47: 177–80
  • Howell A, Cuzick J, Baum M, Buzdar A, Dowsett M, Forbes JF, et al. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years' adjuvant treatment for breast cancer. Lancet 2005; 365(9453)60–2
  • Kaufmann M, Rody A. Long-term risk of breast cancer recurrence: The need for extended adjuvant therapy. J Cancer Res Clin Oncol 2005; 131: 487–94
  • Lonning PE, Geisler J, Krag LE, Erikstein B, Bremnes Y, Hagen AI, et al. Effects of exemestane administered for 2 years versus placebo on bone mineral density, bone biomarkers, and plasma lipids in patients with surgically resected early breast cancer. J Clin Oncol 2005; 23: 5126–37
  • Aapro M. Improving bone health in patients with early breast cancer by adding bisphosphonates to letrozole: The Z-ZO-E-ZO-FAST program. Breast 2006; 15(Suppl 1)S30–S40
  • Vehmanen L, Saarto T, Risteli J, Risteli L, Blomqvist C, Elomaa I. Short-term intermittent intravenous clodronate in the prevention of bone loss related to chemotherapy-induced ovarian failure. Breast Cancer Res Treat 2004; 87: 181–8
  • Hillner BE, Ingle JN, Chlebowski RT, Gralow J, Yee GC, Janjan NA, et al. American Society of Clinical Oncology update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol ;21 2003; 2003: 4042–57
  • Saad F, Gleason DM, Murray R, Tchekmedyian S, Venner P, Lacombe L, et al. 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
  • 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
  • Dearnaley DP, Sydes MR, Mason MD, Stott M, Powell CS, Robinson AC, et al. A double-blind, placebo-controlled, randomized trial of oral sodium clodronate for metastatic prostate cancer (MRC PR05 Trial). J Natl Cancer Inst 2003; 95: 1300–11
  • Ernst DS, Tannock IF, Winquist EW, Venner PM, Reyno L, Moore MJ, et al. Randomized, double-blind, controlled trial of mitoxantrone/prednisone and clodronate versus mitoxantrone/prednisone and placebo in patients with hormone-refractory prostate cancer and pain. J Clin Oncol 2003; 21: 3335–42
  • Mason M. Development of bone metastases from prostate cancer: First results of the MRC PR04 trial (ISCRTN 61384873) [abstract]. J Clin Oncol 2004; 22(14S)4511
  • Smith MR, McGovern FJ, Zietman AL, Fallon MA, Hayden DL, Schoenfeld DA, et al. Pamidronate to prevent bone loss during androgen-deprivation therapy for prostate cancer. N Engl J Med 2001; 345: 948–55
  • Smith MR, Eastham J, Gleason DM, Shasha D, Tchekmedyian S, Zinner N. Randomized controlled trial of zoledronic acid to prevent bone loss in men receiving androgen deprivation therapy for nonmetastatic prostate cancer. J Urol 2003; 169: 2008–12
  • Nelson, J, Greenspan, S, Resnick, N, Trump, D, Parker, R. Once weekly oral alendronate prevents bone loss in men on androgen deprivation therapy for prostate cancer [abstract]. Prostate Cancer Symposium, San Francisco, CA. 2006. p 139.
  • Michaelson, M, Lee, H, Kaufman, D, Kantoff, P, Finkelstein, J, Smith, M. Annual zoledronic acid to prevent gonadotropin-releasing hormone agonist-induced bone loss in men with prostate cancer: A randomized placebo-controlled trial [abstract]. J Clin Oncol 2006;24(18S):4515.
  • Rosen LS, Gordon D, Tchekmedyian NS, Yanagihara R, Hirsh V, Krzakowski M, 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: A randomized, Phase III, double-blind, placebo-controlled trial. Cancer 2004; 100: 2613–21
  • Guarneri V, Donati S, Nicolini M, Giovannelli S, D'Amico R, Conte PF. Renal safety and efficacy of i.v. bisphosphonates in patients with skeletal metastases treated for up to 10 Years. Oncologist 2005; 10: 842–8
  • Chang, JT, Green, L, Beitz, J. Renal failure with the use of zoledronic acid. N Engl J Med 2003;349:1676–9; Discussion –9.
  • Markowitz GS, Fine PL, Stack JI, Kunis CL, Radhakrishnan J, Palecki W, et al. Toxic acute tubular necrosis following treatment with zoledronate (Zometa). Kidney Int 2003; 64: 281–9
  • Markowitz GS, Appel GB, Fine PL, Fenves AZ, Loon NR, Jagannath S, et al. Collapsing focal segmental glomerulosclerosis following treatment with high-dose pamidronate. J Am Soc Nephrol 2001; 12: 1164–72
  • Desikan R, Veksler Y, Raza S, Stokes B, Sabir T, Li ZJ, et al. Nephrotic proteinuria associated with high-dose pamidronate in multiple myeloma. Br J Haematol 2002; 119: 496–9
  • Novartis. Zometa prescribing information. 2005.
  • Rosen CJ, Brown S. Severe hypocalcemia after intravenous bisphosphonate therapy in occult vitamin D deficiency. N Engl J Med 2003; 348: 1503–4
  • Lu KC, Yeung LK, Lin SH, Lin YF, Chu P. Acute effect of pamidronate on PTH secretion in postmenopausal hemodialysis patients with secondary hyperparathyroidism. Am J Kidney Dis 2003; 42: 1221–7
  • Bamias A, Kastritis E, Bamia C, Moulopoulos LA, Melakopoulos I, Bozas G, et al. Osteonecrosis of the jaw in cancer after treatment with bisphosphonates: Incidence and risk factors. J Clin Oncol 2005; 23: 8580–7
  • Marx RE. Pamidronate (Aredia) and zoledronate (Zometa) induced avascular necrosis of the jaws: A growing epidemic. J Oral Maxillofac Surg 2003; 61: 1115–7
  • Ruggiero SL, Mehrotra B, Rosenberg TJ, Engroff SL. Osteonecrosis of the jaws associated with the use of bisphosphonates: A review of 63 cases. J Oral Maxillofac Surg 2004; 62: 527–34
  • Migliorati CA. Bisphosphanates and oral cavity avascular bone necrosis. J Clin Oncol 2003; 21: 4253–4
  • Lugassy G, Shaham R, Nemets A, Ben-Dor D, Nahlieli O. Severe osteomyelitis of the jaw in long-term survivors of multiple myeloma: A new clinical entity. Am J Med 2004; 117: 440–1
  • Carter G, Goss AN, Doecke C. Bisphosphonates and avascular necrosis of the jaw: A possible association. Med J Aust 2005; 182: 413–5
  • Vannucchi AM, Ficarra G, Antonioli E, Bosi A. Osteonecrosis of the jaw associated with zoledronate therapy in a patient with multiple myeloma. Br J Haematol 2005; 128: 738
  • Tarassoff P, Csermak K. Avascular necrosis of the jaws: Risk factors in metastatic cancer patients. J Oral Maxillofac Surg 2003; 61: 1238–9
  • Woo SB, Hellstein JW, Kalmar JR. Narrative [corrected] review: Bisphosphonates and osteonecrosis of the jaws. Ann Intern Med 2006; 144: 753–61
  • Migliorati CA, Casiglia J, Epstein J, Jacobsen PL, Siegel MA, Woo SB. Managing the care of patients with bisphosphonate-associated osteonecrosis: An American Academy of Oral Medicine position paper. J Am Dent Assoc 2005; 136: 1658–68
  • Hillner BE, Ingle JN, Berenson JR, Janjan NA, Albain KS, Lipton A, et al. American Society of Clinical Oncology guideline on the role of bisphosphonates in breast cancer. American Society of Clinical Oncology Bisphosphonates Expert Panel. J Clin Oncol 2000; 18: 1378–91
  • Ross JR, Saunders Y, Edmonds PM, Patel S, Wonderling D, Normand C, et al. A systematic review of the role of bisphosphonates in metastatic disease. Health Technol Assess 2004; 8: 1–176
  • Hillner BE, Weeks JC, Desch CE, Smith TJ. Pamidronate in prevention of bone complications in metastatic breast cancer: A cost-effectiveness analysis. J Clin Oncol 2000; 18: 72–9
  • Dranitsaris G, Hsu T. Cost utility analysis of prophylactic pamidronate for the prevention of skeletal related events in patients with advanced breast cancer. Support Care Cancer 1999; 7: 271–9
  • Coyte PC, Dobrow MJ, Broadfield L. Incremental cost analysis of ambulatory clinic and home-based intravenous therapy for patients with multiple myeloma. Pharmacoeconomics 2001; 19: 845–54
  • Smith AG, Soutar RL, Schey S, Andrews CD, Baister ER, Bilbrough C, et al. Home care versus hospital care in patients with multiple myeloma treated with pamidronate. Int J Palliat Nurs 2004; 10: 144–9
  • Wardley A, Davidson N, Barrett-Lee P, Hong A, Mansi J, Dodwell D, et al. Zoledronic acid significantly improves pain scores and quality of life in breast cancer patients with bone metastases: A randomised, crossover study of community vs hospital bisphosphonate administration. Br J Cancer 2005; 92: 1869–76
  • Katoh M, Unakami M, Hara M, Fukuchi S. Bone metastasis from colorectal cancer in autopsy cases. J Gastroenterol 1995; 30: 615–8
  • Kanthan R, Loewy J, Kanthan SC. Skeletal metastases in colorectal carcinomas: A Saskatchewan profile. Dis Colon Rectum 1999; 42: 1592–7
  • Hultborn R, Gundersen S, Ryden S, Holmberg E, Carstensen J, Wallgren UB, et al. Efficacy of pamidronate in breast cancer with bone metastases: A randomized, double-blind placebo-controlled multicenter study. Anticancer Res 1999; 19(4C)3383–92
  • Body JJ, Diel IJ, Lichinitzer M, Lazarev A, Pecherstorfer M, Bell R, 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 2004; 90: 1133–7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.