Skip to main content
Erschienen in: Calcified Tissue International 2/2018

Open Access 20.01.2018 | Review

Cancer Treatment and Bone Health

verfasst von: Catherine Handforth, Stella D’Oronzo, Robert Coleman, Janet Brown

Erschienen in: Calcified Tissue International | Ausgabe 2/2018

Abstract

Considerable advances in oncology over recent decades have led to improved survival, while raising concerns about long-term consequences of anticancer treatments. In patients with breast or prostate malignancies, bone health is a major issue due to the high risk of bone metastases and the frequent prolonged use of hormone therapies that alter physiological bone turnover, leading to increased fracture risk. Thus, the onset of cancer treatment-induced bone loss (CTIBL) should be considered by clinicians and recent guidelines should be routinely applied to these patients. In particular, baseline and periodic follow-up evaluations of bone health parameters enable the identification of patients at high risk of osteoporosis and fractures, which can be prevented by the use of bone-targeting agents (BTAs), calcium and vitamin D supplementation and modifications of lifestyle. This review will focus upon the pathophysiology of breast and prostate cancer treatment-induced bone loss and the most recent evidence about effective preventive and therapeutic strategies.

Introduction

Advances in systemic cancer therapies and diagnostic techniques have led to consistent improvements in cancer survival, and, as a result, the long-term sequelae of cancer treatment have become an increasingly important consideration. Treatment-related adverse effects significantly impact the quality of life of patients, many of whom will live for years following their cancer diagnosis. They are also associated with significant economic costs to health and social care providers.
Bone health is an area of particular importance for patients with breast and prostate tumours. These are the commonest malignancies to affect females and males, respectively. More than 460,000 women are diagnosed with breast cancer (BC) in Europe each year, and BC is responsible for 130,000 deaths [1]. More than 420,000 men are diagnosed with prostate cancer (PC) in Europe each year, with more than half of cases occurring in men aged over 70 years [2]. Survival rates from both BC and PC have significantly improved over the past 40 years as a result of both treatment advances and earlier diagnosis, due to improved diagnostic techniques and screening campaigns.
In BC and PC, both the disease itself and the treatments pose specific challenges to skeletal integrity. This review will focus upon bone loss that occurs as a direct result of cancer therapy (cancer treatment-induced bone loss, CTIBL) for BC and PC. We will describe the factors that regulate bone remodelling processes, the pathophysiology and clinical complications of CTIBL. Evidence to support the use of bone-targeted agents (BTAs) will also be discussed, along with current guidelines for CTIBL management.

Influence of Sex Steroids on Bone

Both androgens and oestrogens have important roles in bone growth and maturation, and maintenance of skeletal integrity. However, accumulating evidence suggests a role for other reproductive hormones, such as activins and inhibins, in the preservation of bone health.

Androgens

Testosterone is the most abundant circulating androgen in men, 95% of which is secreted by the testis. The remaining 5% is formed from the enzymatic conversion of adrenal androgens dehydroepiandrosterone (DHEA) and DHEA sulphate (DHEAS) [3]. In women, the major circulating androgens are produced by the adrenal glands and ovaries and include DHEAS and DHEA, androstenedione (pro-androgens), testosterone and dihydrotestosterone (DHT). Female testosterone production also occurs via peripheral aromatisation of androstenedione. In both men and women, the majority of circulating testosterone is protein bound (to either sex hormone-binding globulin or albumin).
Testosterone may act directly on androgen receptors (AR), or indirectly via aromatisation to oestradiol and activation of oestrogen receptors (ER). The indirect actions of testosterone may also occur following conversion to the more potent DHT by 5α-reductase in peripheral tissues [4].
In bone, androgens exert direct effects on growth plate chondrocytes and promote longitudinal bone growth [5]. Both testosterone and DHT stimulate the proliferation of osteoblast precursors via AR signalling [6]. Binding of androgens with AR also upregulates osteoblast AR expression and promotes their differentiation [7]. Androgens also prevent osteoblast and osteocyte apoptosis and regulate osteoclast activity by inhibiting the interaction of receptor activator of nuclear factor kappa-B ligand (RANK-L) with its receptor (RANK), expressed on osteoclast precursors [8, 9]. Bone formation is promoted by androgen-mediated upregulation of growth factors, such as insulin-like growth factor and transforming growth factor beta, TGFβ [6]; on the other hand, downregulation of interleukin 6 inhibits osteoclast activity [10] through the reduction in levels of osteoprotegerin (OPG), which is produced by osteoblasts and acts as a soluble decoy receptor to RANK-L [11].

Oestrogens

The net action of oestrogens on bone is to decrease bone resorption. Their actions are exerted via ERα and ERβ receptors, expressed by osteoblasts and osteoclasts. Oestrogens increase osteoblast number and activity, inhibit the maturation of osteoclast precursors (via increased OPG production) and also cause osteoclast apoptosis [12, 13]. Oestrogen deficiency is known to increase the rate of osteocyte apoptosis with a consequent increase in skeletal fragility [14, 15].
In women, oestrogens are critical for the maintenance of normal bone mass. At the menopause, loss of ovarian follicular activity causes a significant fall in circulating oestrogens, with a consequent disruption of bone remodelling. The most rapid bone loss occurs in the first 3 years post-menopause (2–5%/year), after which skeletal metabolism becomes “acclimatised” to the low-oestrogen environment and bone loss slows to around 0.5–1.0% per annum. A greater proportion of bone loss occurs at sites containing trabecular bone (such as the spine) than cortical sites (such as the hip) [16].
Oestrogens have been identified as the sex steroids primarily responsible also for the regulation of bone resorption in men. In studies of young male patients unable to produce or respond to oestrogens, there was an increased rate of bone turnover and osteopaenia [17, 18]. Both oestrogens and testosterone are known to be important in the regulation of male bone formation [19].

Activins and Inhibins

Activins and inhibins belong to the TGFβ superfamily and exert their biological activity by interacting with a type II serine/threonine kinase receptor; once the latter associates with the corresponding type I receptor, an intracellular pathway is activated, leading to the regulation of DNA transcription, mediated by Smad proteins [20].
Activin A and B are homodimeric peptides, made up of βA and βB subunits, respectively, and regulated by gonadotropins from the pituitary. Activin A is produced in the gonad, while activin B is secreted by the pituitary gland itself; in the circulation, they are bound to follistatin in order to prevent excessive interaction with the activin type II receptor (ACTRII). Interestingly, neither activin A nor follistatin serum levels vary according to the menopausal status [21]. BC cells have been shown to produce activin, which in turn inhibits the proliferation of ER + tumour cell lines [22]; however, its role in the establishment of bone metastases has not been clarified yet. Moreover, while its capability to promote osteoclastogenesis via RANK upregulation has been ascertained, conflicting data exist about the role of activin in osteoblastogenesis [21].
Inhibin A and B are heterodimeric proteins, including αβA or αβB subunits. Due to their capability to prevent the interaction of activin with ACTRII, inhibins suppress gonadotropin secretion from the pituitary gland. Inhibin B levels tend to decline with age in both women and men, thus representing a good marker for fertility [23]. Similarly to activins, inhibins are secreted by BC cells [24] although their role in carcinogenesis seems contradictory. As activin inhibitors, one would expect them to promote tumour cell proliferation, but in vivo and clinical data suggest a possible tumour-suppressive activity [22]. With respect to bone turnover, this seems to be counteracted by inhibins through the suppression of both osteoblastogenesis and osteoclastogenesis [21]. Moreover, in a cross-sectional study involving 188 women with different ages (from 21 to 85 years), inhibins turned out as the best predictors of bone marker fluctuations, potentially able to indicate the initial bone loss that characterises the perimenopausal period, even before sex steroid and gonadotropin variations could be detected [25].

Bone Loss in Prostate Cancer

Pathophysiology

The growth of PC is initially androgen dependant, and the cornerstone of treatment is androgen deprivation therapy (ADT). This is most often achieved by the administration of luteinising hormone-releasing hormone (LHRH) agonists, LHRH antagonists and antiandrogens. ADT provides excellent initial disease control in virtually all patients; however, many of them subsequently develop castration resistance. Following the initiation of ADT, sex steroid levels decrease rapidly and significantly, reaching a nadir within 2–4 weeks (less than 5 and 20% of normal testosterone and oestrogen levels, respectively). This leads to an accelerated and disorganised bone turnover process and net bone loss that is most rapid during the first year of ADT (studies have reported between 5 and 10% loss of bone mineral density, BMD) [2628]. BMD loss also continues gradually throughout ADT duration. In a study of 390 men aged 54–89, the prevalence of osteoporosis was found to be 35% in hormone-naïve patients, 43% after 2 years of ADT and 81% after 10 years of ADT [29].
The pattern of CTIBL associated with ADT has been shown to differ from that seen with other conditions, being more likely to affect the radius than the hip or spine [30]. However, recent evidence suggests that this phenomenon could be partially attributed to the frequent overestimation of central BMD in men with skeletal degenerative disorders, which commonly occur in the elderly. On the other hand, densitometers have been found to over-read radius osteoporosis when using a male normative database, and thus forearm BMD should be re-analysed and evaluated according to larger normative databases [31].
In addition to ADT, men with newly diagnosed hormone-sensitive metastatic PC may receive chemotherapy [32]. Docetaxel is given for 18 weeks, along with daily 10 mg prednisolone and 24 mg dexamethasone every 3 weeks. It is well established that long-term use of glucocorticoids is associated with osteoporosis; indeed, it is the most common cause of secondary osteoporosis. Evidence suggests that the risk of hip and vertebral fractures increases up to 7- and 17-fold, respectively, when doses equivalent to 10–12 mg prednisolone are given for more than 3 months. The risk of fracture is also increased even when small (2.5–3 mg) daily doses are given [33]. No studies to date have investigated the impact of the combination of chemotherapy and glucocorticoids with ADT on BMD and fracture risk in men with PC.

Clinical Sequelae

CTIBL that is associated with ADT exponentially increases the risk of fracture. Indeed, the loss of 10–15% of BMD doubles the fracture risk [34] and men receiving ADT for PC are five times more likely to develop a fracture than healthy age-matched controls [35].
In particular, Shahinian et al. retrospectively evaluated the fracture risk of 50,613 PC patients listed in the Surveillance Epidemiology and End Results (SEER) programme between 1992 and 1997. Among men who survived at least 5 years after PC diagnosis, those receiving ADT exhibited a significantly higher risk of fracture, as compared to untreated men (19.4 vs. 12.6%, p < 0.001). Moreover, patients undergoing orchiectomy or receiving at least nine doses of LHRH agonists had the lowest fracture-free survival, although the analysis did not exclude bone metastasis-related fractures [36].
Another similar analysis involved 11,661 patients with non-metastatic PC and confirmed the significantly higher fracture rate in men undergoing ADT (7.88 vs. 6.51%/year of controls, p < 0.001), and the highest hazard ratio (HR) in those receiving LHRH agonists for at least 12 months (1.16, 95% CI 1.08–1.26; p < 0.001). Interestingly, when considering fractures at specific sites, both vertebral and hip/femur ones were more frequent in patients undergoing ADT, as compared to controls (p < 0.001 and p = 0.002, respectively) [37].
Not only are such events associated with subsequent fractures and loss of independence, but they also represent an independent adverse predictor of survival. Indeed, the relative risk of death is sevenfold greater in men with PC receiving ADT and who have a previous fracture compared to those with no fracture history [38].

Bone Loss in BC

Since the early 1990s, although the incidence of BC has steadily risen, BC mortality has progressively decreased. This “good news” has, however, raised concerns about the possible long-term effects of cancer treatments. In particular, both chemotherapy and hormone treatment have an impact on bone health, with different mechanisms that will be discussed in the next sections.

Pathophysiology of Chemotherapy-Induced Bone Loss and Clinical Data

Adjuvant chemotherapy has a significant impact on bone health in BC, due to both the induction of premature menopause and direct effects on bone turnover [39]. For example, cyclophosphamide exerts gonadotoxic effects (the metabolite phosphoramide mustard damages at first granulosa cells and then oocytes in a dose-dependent manner) [40] and also affects cell division of both osteoclast and osteoblast precursors, thus interfering with physiological bone turnover [41]. Additionally, doxorubicin, although less gonadotoxic, can also induce both premature ovarian failure in vivo [42] and increased osteoclast differentiation in vitro, at the expense of osteoblasts [43].
Studies investigating the effects of adjuvant chemotherapy on BMD in women with early BC have all reported a decrease in lumbar spine (LS) BMD, as compared to baseline [4447]. In particular, Shapiro and coworkers investigated the effects of adjuvant chemotherapy (containing cyclophosphamide and/or doxorubicin) on the bone health of 49 premenopausal patients with early BC; interestingly, all women experiencing premature ovarian failure (35/49) showed a significant BMD reduction at the LS within the first 6 months (− 4.0%, p = 0.0001); this was greater than the bone loss seen in women who retained menses (− 1.0%) [44].
In another study conducted by Cameron et al., 41 premenopausal patients received adjuvant chemotherapy for early BC and were monitored in their bone health and ovarian function for 1 year. During the first 6 months, all patients experienced LS BMD decrease (p < 0.0001), independently of age, ovarian function and type of chemotherapy; during the following 6 months, LS BMD further decreased only in women experiencing amenorrhea and/or low estradiol levels (p < 0.0001) [47].
A recent prospective study reported BMD changes in 97 women (mean age: 53 years) with early BC who had completed anthracycline- or taxane-based adjuvant chemotherapy [48]. All patients received high-dose prednisolone as antiemetic prophylaxis and supplementation with calcium and vitamin D. At completion of chemotherapy, BMD at LS increased significantly in postmenopausal women compared to baseline (+ 2.35%, 95% CI 1.6–3.3, p < 0.001), whereas the BMD of premenopausal women did not change. Such effects could be partially explained by the vitamin supplementation which could prevent the typical calcium deficiency occurring in postmenopausal women.
Based on these data, chemotherapy-induced bone loss is currently interpreted more as the consequence of premature ovarian failure, rather than the result of direct bone damage.

Pathophysiology of Hormone Treatment-Induced Bone Loss and Clinical Data

Long-Acting Gonadotropin-Releasing Hormone Agonists

Long-acting gonadotropin-releasing hormone (GnRH) agonists may be administered to premenopausal individuals with hormone receptor-positive (HR+) BC. Persistent activation of GnRH receptors leads to their desensitisation and consequent suppression of ovarian function. Induction of hypogonadism in premenopausal women is associated with accelerated bone loss which is usually, at least in part, reversible once treatment is stopped, especially in those who resume menses [49].
The Zoladex Early Breast Cancer Research Association (ZEBRA) bone sub-study included 96 women with premenopausal node-positive BC who had been randomised to receive either goserelin or CMF chemotherapy (cyclophosphamide, methotrexate and 5-fluorouracil). After 2 years of hormone treatment, goserelin was associated with a significantly greater reduction in BMD at both LS (mean BMD loss − 10.5 vs. − 6.5%, p = 0.0005) and femoral neck (FN) (− 6.4 vs. − 4.5%, p = 0.04), as compared to the CMF group. After a 3-year follow-up, partial BMD recovery was observed in goserelin-treated patients, but this did not occur in the CMF group [50].
However, the strongest evidence of goserelin impact on bone health derives from the Austrian Breast and Colorectal Cancer Study Group trial-12 (ABCSG-12), in which 404 premenopausal women with BC were randomised to receive adjuvant hormone treatment with or without zoledronate. Endocrine therapy consisted of either goserelin + tamoxifen or goserelin + anastrozole combination and, once administered without the bisphosphonate (BP), was associated with significant BMD reduction, evaluated after 36 and 60 months and compared to baseline values (after 36 months: LS BMD − 11.3%, FN BMD − 7.3%, p < 0.0001 in both instances; after 60 months: LS BMD − 6.3%, FN BMD − 4.1%, p = 0.0001 and p = 0.058, respectively). On the other hand, women receiving zoledronate exhibited stable and increased BMD after 36 and 60 months, respectively [51].

Tamoxifen

Tamoxifen is a selective oestrogen receptor modulator (SERM) exerting both antagonist (breast) and partial agonist effects (bone and uterus), that is widely used as treatment for women with HR + BC and as chemoprevention in women at high risk for developing BC. A placebo-controlled BC chemoprevention trial investigated the effects of tamoxifen on bone health in 179 women. In premenopausal women treated with tamoxifen, there was a mean annual BMD loss of 1.44% at LS, compared with a gain of 0.24% in the placebo group (p < 0.001). Tamoxifen had the opposite effect in postmenopausal patients, in whom it was associated with increased BMD at the LS (mean annual increase of 1.17%) compared with placebo (insignificant BMD loss) [52]. Subsequent studies involving postmenopausal women treated with tamoxifen reported reductions in bone turnover markers (BTM) [53] and significantly increased LS trabecular bone score (TBS), a novel parameter which reflects bone microarchitecture and is associated with resistance to fractures [54].

Aromatase Inhibitors

Third-generation aromatase inhibitors (AIs) induce a rapid decrease in circulating estradiol levels by preventing the conversion of androgens to oestrogens. Therefore, they represent a standard adjuvant treatment for postmenopausal patients with HR+ BC. Steroidal AIs (e.g. exemestane) irreversibly bind to the aromatase enzyme, while non-steroidal AIs (e.g. anastrozole, letrozole) are competitive inhibitors which bind to the heme moiety of the aromatase cytochrome P450 complex [55]. Table 1 summarises the most important studies investigating the effects of AIs on bone health.
Table 1
Clinical trials investigating the effects of aromatase inhibitors on fracture incidence
Clinical trial
Number of patients
Experimental arm
Control arm
Overall/annual fracture incidence
References
ATAC
9366
Anastrozole
Tamoxifen
Overall: 11 vs. 7.7%
Annual: 2.93 vs. 1.90%
[57, 58]
IBIS II bone sub-study
1410
Anastrozole
Placebo
Annual: 1.37 vs. 1.26%
[100]
BIG 1.98
4895
Letrozole
Tamoxifen
Overall: 9.3 vs. 6.5%
Annual: 2.71 vs. 1.872%
[59]
MA-17
5187
Letrozole
Placebo
Overall: 3.6 vs. 2.9%
[60]
IES
4274
Exemestane
Tamoxifen
Overall: 7 vs. 5%
Annual: 2.01 vs. 1.60%
[61]
ABCSG-18
3420
AI + denosumab
AI + placebo
Overall: 5 vs. 9.6%
[107]
AI aromatase inhibitor
The ATAC trial investigated the effects of adjuvant anastrozole on bone health [56]. The study reported an increased fracture risk in patients treated with anastrozole, as compared to tamoxifen (2.93 vs. 1.90% annual fracture rate, p < 0.0001). However, after the treatment was suspended, anastrozole-treated patients experienced BMD recovery at LS and no further loss at the hip, suggesting that AI-induced bone loss is reversible [57, 58].
Adjuvant letrozole has been found to be associated with an increase in annual (2.71 vs. 1.87%) and overall fractures (9.3 vs. 6.5%) when compared with tamoxifen [59]. The MA17 trial included over 5000 women and confirmed that the addition of letrozole, after standard 5-year adjuvant therapy with tamoxifen, was associated with significantly improved disease-free survival (DFS) in postmenopausal women [60]. However, as compared to placebo, letrozole was also associated with a slightly increased incidence of osteoporosis (5.8 vs. 4.5%, p = 0.07), although the rates of fracture were similar in the two treatment groups.
In the Intergroup Exemestane Study (IES), postmenopausal women completing 2–3 years of adjuvant tamoxifen were randomised to continue the treatment until the fifth year or switch to exemestane. Those in the exemestane arm experienced a greater decrease in BMD and, in the intention-to-treat population, higher annual fracture incidence than those who continued to take tamoxifen (2.01 vs. 1.60% with tamoxifen) [61]. Fracture rates associated with the use of non-steroidal and steroidal AIs appear to be similar [62].
A recent retrospective study evaluated LS BMD and TBS variations in 321 non-osteoporotic patients treated with AIs for at least 3 years. Both parameters significantly decreased from baseline to 5 years (− 6.15 and − 2.12%, respectively; p < 0.001 in both instances) although the annual reduction tended to slow after 3 and 1 years of treatment, respectively [63].
An interesting line of current research aims to investigate CYP19A1 (aromatase) gene polymorphisms potentially associated with higher susceptibility to AI-induced bone loss. It has been reported that G-to-A substitution at Val80 in the exon 3 (rs700518) is apparently related to significant BMD decrease at LS and hip at 12 months [64]. More recently, a significant correlation between another polymorphism (rs4646, GG genotype) and osteoporosis during AI treatment has been described [65].

Guidelines for Bone Health Assessment

Prostate Cancer

European guidelines for PC were published jointly by the European Association of Urology (EAU), International Society of Geriatric Oncology (SIOG) and European Society for Radiotherapy and Oncology (ESTRO) in 2016 [66]. These recommend that all men starting long-term ADT should undergo dual-energy X-ray absorptiometry (DXA) assessment and the result used in conjunction with the FRAX® tool to evaluate individual fracture risk. The FRAX® tool is based upon data from prospectively studied population cohorts [67]. This tool is available online and may be used for individuals aged 40–90 years across multiple countries and regions of the world to predict the 10-year risk of hip and major osteoporotic fractures, on the basis of multiple risk factors (e.g. age, sex, smoking, alcohol intake, history of fractures, familiar history of hip fractures, body mass index < 20 kg/m2, long-term use of corticosteroids, comorbidities, BMD).
However, these guidelines lack specific recommendations regarding the ongoing monitoring and follow-up of CTIBL. Previous EAU guidelines suggested that the initial T score should guide the frequency of BMD measurement at the time of ADT initiation (DXA to be repeated annually if the T score is between − 2.5 and − 1.0 at baseline, or every 2 years if the initial T score is above − 1.0) [68]. The International Society for Clinical Densitometry suggests that BMD measurement should be repeated after the first year of ADT, which seems appropriate given that the most rapid decline in BMD is observed during the first year of therapy [69].
Despite available guidelines, the timing and frequency of DXA assessments in men with PC are subject to significant variation and depend upon clinician awareness and preferences, access to DXA scans and patient factors. There is a need for comprehensive and consistent guidance for both the initial and the long-term management of bone health in men with PC receiving ADT.

Breast Cancer

Both the European Society for Medical Oncology (ESMO) and the American National Comprehensive Cancer Network (NCCN) recommend bone health assessment in women with iatrogenic premature ovarian failure and in patients treated with AIs for BC [70, 71]. The baseline evaluation should include a careful history to identify risk factors for fragility fractures [39, 72]. DXA assessment of BMD is recommended at baseline and should be repeated periodically, according to the local guidelines for osteoporosis management. The FRAX® tool is also useful to estimate the 10-year risk of fracture [67]; however, the algorithm does not include cancer treatment as a risk factor. Further information may be provided by laboratory assessment including calcium, phosphate, vitamin D, parathyroid hormone (PTH) and creatinine clearance. Bone biomarkers (such as serum bone alkaline phosphatase and urinary N-telopeptide of type I collagen) have been investigated for their ability to predict osteoporosis and fragility fractures in BC [73], but their routine use in clinical practice is currently limited by the high inter- and intra-individual variability.

Management of CTIBL

CTIBL in Prostate Cancer

Lifestyle Measures

Both smoking and excessive alcohol intake are associated with reduced BMD and should be avoided [74]. Other consequences of ADT are sarcopenia and fatigue, both of which increase the likelihood of frailty, falls and fractures [75]. Regular exercise is helpful to minimise this risk, and supervised aerobic and resistance exercise programmes, performed at least twice a week for 12 weeks, are currently recommended for all men undergoing ADT [7678].

Calcium and Vitamin D Supplementation

Men with PC are frequently deficient in both calcium and vitamin D [79]. Since vitamin D deficiency in men receiving ADT is independently associated with spinal fractures [80], supplementation with calcium and vitamin D should be considered in all men receiving ADT. However, the recommended doses (500–1000 mg calcium and 200–500 IU vitamin D per day) may be insufficient to prevent bone loss [81]. Further intervention studies to determine both the safety and efficacy of higher doses are required.

Bone-Targeted Agents (BTAs)

BTAs have an important role in the management of CTIBL. Bisphosphonates (BP) have high affinity for mineralised bone matrix, where they bind selectively to hydroxyapatite and are released during resorption. Ingestion of BP by osteoclasts results in their inhibition, either through induction of apoptosis (non-nitrogen-containing BP such as clodronate) or through inhibition of the mevalonate pathway required for osteoclastogenesis (nitrogen-containing BP such as zoledronate, ibandronate and pamidronate). Many BP are administered orally; however, the most comprehensively studied is zoledronate which is given intravenously. It requires dosage adjustment in patients with a creatinine clearance (CrCl) < 60 ml/min and is contraindicated in severe renal impairment (CrCl < 30 ml/min).
Denosumab is another BTA approved for the treatment of CTIBL. It is a fully humanised monoclonal IgG2 antibody that targets RANK-L and prevents its interaction with RANK on osteoclast precursors. Consequent inhibition of osteoclast differentiation and activation causes a rapid reduction in bone resorption [82].
Other antiresorptive drugs are currently widely used for the management of non-malignant bone disease, but are not specifically recommended in the CTIBL setting. These include calcitonin (the clinical investigation of which in cancer patients has been very limited) and teriparatide, a recombinant human PTH analogue. Teriparatide has been associated with an increased risk of osteosarcoma in patients receiving bone radiotherapy and is contraindicated for the treatment of CTIBL [83].
The potential role of SERM for the prevention of ADT-induced bone loss has been widely investigated by Smith and coworkers. In particular, 12-month treatment with either raloxifene [84] or toremifene [85] turned out to be capable to increase BMD at all sites, while reducing BTM levels, in men with non-metastatic PC undergoing ADT. When administered for 24 months, toremifene induced a 50% reduction in new vertebral fractures and significantly decreased the risk of fractures at all sites (p < 0.05 in both instances), as compared to placebo [86].
The role of denosumab in men with PC was evaluated in a randomised study of 1468 men receiving ADT for localised PC. Denosumab increased LS BMD by 5.6%, whereas those in the placebo group experienced a 1.0% loss (p < 0.001) [87]. Denosumab was also associated with a reduction in the incidence of new vertebral fractures after 36 months of treatment (1.5 vs. 3.9% with placebo; RR 0.38; 95% CI 0.19–0.78), and with a non-significant reduction in fractures at any site. Importantly, there were no between-group differences in adverse events.
Several studies have investigated the ability of BP to prevent CTIBL associated with ADT. BP have been found to increase LS and hip BMD when compared to placebo, and also to be associated with a significant reduction in BTM [8892]. However, available evidence is limited by small patient numbers, heterogeneous populations, variations in the type and frequency of BP administration and different schedules of follow-up, all of which preclude effective meta-analysis. No study had sufficient power to detect differences in the incidence of fractures between groups. Only one non-randomised study of 94 patients attempted to compare the efficacy of different BP [93]. Large, prospective randomised studies of BP in men receiving ADT are necessary in order to determine the efficacy of BP for fracture prevention in this setting.

Guidelines for the Use of BTA in PC

European guidelines are available to guide the approach to CTIBL in PC patients [70]. These include a treatment algorithm for assessment and management of bone health (Fig. 1). Although a range of BP have been shown to prevent bone loss in men with locally advanced PC receiving ADT, denosumab is currently the only BTA that is licensed for the prevention of CTIBL in PC.

CTIBL in BC

Lifestyle Measures

Limitation of alcohol consumption and smoking cessation are recommended. Moderate weight-bearing exercise should be practised regularly to take advantage of the beneficial effects of exercise on BMD [94].

Calcium and Vitamin D Supplementation

If dietary intake is inadequate, calcium supplementation is recommended (1000 mg/day) together with vitamin D supplementation (800–1000 IU/day). Concomitant steroid uptake interferes with vitamin D absorption and requires higher dosage [39, 95]. Elderly patients, and those with reduced sunlight exposure and/or physical activity, should be assessed for vitamin D serum levels and deficient levels treated with high-dose vitamin D followed by ongoing supplementation [94].

BTA

Several clinical trials have investigated the role of BP in the management of BC-associated CTIBL; the most important are summarised in Table 2. The earliest results came from small studies exploring the efficacy of oral clodronate. Postmenopausal women treated with adjuvant tamoxifen or toremifene were randomised to the clodronate or control group for 3 years, where clodronate was associated with a small BMD improvement at both LS (+ 1.0 vs. − 1.7%, p = 0.01) and FN (+ 2.4 vs. − 0.4%, p = 0.12), as compared to patients not receiving the BP [96].
Table 2
Clinical trials investigating the efficacy of bisphosphonates in preventing AI-induced CTIBL
Clinical trial
No. of patients
Follow-up period (months)
Experimental arm
Control arm
BMD variations at LS
References
ARIBON
131
24
Anastrozole + ibandronate
Anastrozole + placebo
+ 2.98 vs. − 3.22%
[98]
SABRE
234
24
Anastrozole + risedronate
Anastrozole + placebo
+ 2.2 vs. − 1.8%
[99]
IBIS II bone sub-study
1410
36
Anastrozole + risedronate
Anastrozole + placebo
+ 1.1 vs. − 2.6%
[100]
Z-FAST
602
61
Letrozole + upfront ZA
Letrozole + upfront ZA
+ 6.18–6.22 vs. − 2.42%
[101]
ZO-FAST
1065
60
Letrozole + upfront ZA
Letrozole + delayed ZA
+ 4.3 vs. − 5.4%
[102]
E-ZO-FAST
527
12
Letrozole + upfront ZA
Letrozole + delayed ZA
+ 2.72 vs. − 2.71%
[103]
BMD bone mineral density, LS lumbar spine, ZA zoledronic acid
The ARIBON trial reported that 2-year treatment with ibandronate induced a significant BMD gain at both LS and hip (+ 2.98 and + 0.60%, p < 0.01 for both) in osteopaenic postmenopausal women treated with adjuvant anastrozole [97]. Patients who continued ibandronate treatment up to 5 years underwent a mean lumbar BMD increase of + 3.19%, while those stopping the BP showed a BMD reduction at both LS (− 3.21%) and hip (− 5%) at the 5-year follow-up [98].
The efficacy of oral risedronate in postmenopausal women was investigated in both the SABRE trial and the IBIS II study. SABRE reported that those at moderate or high risk of fragility fractures significantly benefited from the addition of risedronate to adjuvant AI treatment [99]. IBIS II investigated the effect of anastrozole or placebo for BC prevention in women who were at high risk of BC. Those with osteopaenia or osteoporosis at baseline were included in a sub-study to assess bone protection with risedronate. After 3 years, the addition of risedronate to anastrozole was associated with a 1.1% (95% CI 0.2–2.1) increase in LS BMD compared to a 2.6% (95% CI − 4.0 to − 1.3) decrease with placebo (p < 0.0001) in osteopaenic women; among osteoporotic patients, those receiving placebo and risedronate experienced a greater gain in BMD at LS (3.9 vs. 1.2%, p = 0.006) and hip (1.5 vs. 0.3%, p = 0.12) than those treated with anastrozole with risedronate, but the BP still counterbalanced the AI-induced bone loss [100].
Zoledronate is the most extensively investigated intravenous BP in the CTIBL setting, both in pre- and postmenopausal patients. In the bone sub-protocol of the ABCSG-12 trial, 404 premenopausal women were randomised to receive hormone treatment (goserelin + anastrozole vs. goserelin + tamoxifen) with or without zoledronate. After 3 years, patients receiving the BP exhibited stable BMD at both LS and trochanter (+ 0.4 and + 0.8%, respectively), while at 60 months zoledronate treatment was associated with BMD increases at both sites, compared to baseline (4.0% at LS, p = 0.02; 3.9% at trochanter, p = 0.07) [51]. Those who received endocrine therapy alone experienced significant bone loss at both time-points, with the greatest loss being observed in the goserelin + anastrozole arm.
In postmenopausal women undergoing AI therapy, the Z-FAST, ZO-FAST and E-ZO-FAST trials compared upfront zoledronate treatment (4 mg every 6 months) with delayed administration, started after the detection of fractures or BMD decrease. At the final analyses, all these studies showed significantly higher BMD values in the “upfront” arms at LS, FN and hip, as compared to “delayed-treatment” groups [101103]. In agreement with these data, Wagner-Johnston et al. described that upfront zoledronate addition to letrozole treatment, in postmenopausal patients previously treated with tamoxifen, was less frequently associated with LS BMD decrease as compared to delayed therapy. This difference was statistically significant after 5 years (p < 0.0001), while there were no significant differences in osteoporosis and fracture occurrence [104]. A recent study has also described a significant increase in LS BMD (+ 11.6%, p = 0.01) when zoledronate was given alongside adjuvant AI in women with pre-existing osteopaenia or osteoporosis [105].
Denosumab has also been investigated for its role in the prevention of AI-induced bone loss. In an initial randomised phase II study, 252 women with early HR + BC were randomised to receive an AI with or without denosumab (60 mg every 6 months for 2 years), and those in the denosumab arm experienced higher BMD gains at multiple sites, compared to controls [106]. The subsequent ABCSG-18 phase III trial prospectively evaluated the effects of adjuvant denosumab in postmenopausal patients with early HR + BC receiving AIs. A total of 3420 women were randomised to receive either denosumab or placebo every 6 months. Denosumab was associated with a significantly delayed time to first fracture and a reduced fracture incidence, compared to placebo (independently of baseline BMD) [107]. However, after denosumab discontinuation a rebound effect with accelerated bone loss was described [108, 109] leading to the proposal of a sequential BP administration to retain BMD values.

Guidelines for the Use of BTAs in BC

Current guidelines recommend adjuvant BTA treatment in those with a baseline t score < − 2 or at least two risk factors for fractures [110]. A panel of bone experts representing the major international societies that promote bone health and research (i.e. International Osteoporosis Foundation, IOF; Cancer and Bone Society, CABS; European Calcified Tissue Society, ECTS; International Expert Group for AIBL, IEG; European Society for Clinical and Economics Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases, ESCEO; International Menopause Society, IMS) has recently published a position statement that recommends BTA administration also to patients with a baseline t score < − 1.5 with an additional risk factor for fracture, while those women with a baseline t score > − 2 and no fracture risk factors should be managed according to the BMD variations which occurred during the first year of AI treatment [94]. There is no consensus about the optimal treatment duration, but the Cancer Care Ontario (CCO) and American Society of Clinical Oncology (ASCO) guidelines do not recommend the administration of zoledronate for more than 5 years, while clodronate should be given for up to 3 years [111].
In premenopausal patients undergoing ovarian suppression, 4 mg zoledronate every 6 months is recommended in addition to calcium and vitamin D supplementation. In postmenopausal women, either oral or intravenous BP have proven efficacious in preventing bone loss during AI treatment, and thus the treatment choice should depend upon local guidelines, the different toxicity profiles and concurrent therapies [49, 70]. Further clinical trials are needed to compare different BP doses and schedules [111].
Compliance to oral BP should be regularly assessed and, if unsatisfactory, switching to an intravenous BTA should be considered [94].
A recent prospective study investigated adherence to CTIBL treatment guidelines by both patients and clinicians. Vitamin D and calcium supplementation was prescribed in three quarters of those receiving AIs, but failure to comply with guidelines was identified in 11.6% of cases. A total of 12% (54 of 438 patients) received a BP, but seven patients who were osteoporotic did not receive antiresorptive medication [112]. These data underline the need for clarification and improved awareness of current guidelines.
There is still a debate about the administration of denosumab in the adjuvant setting; European bone experts suggest that it might be offered at the same dosage administered to patients with osteoporosis [49], while US clinicians underline the need for more consistent data, before making any recommendation [111].
Recent evidence suggests that adjuvant BP also reduce bone recurrence and improve survival in postmenopausal patients with BC [113]. Although the routine use of BP in this setting has not received regulatory approval, bone experts and oncologists recommend their administration to postmenopausal women with intermediate/high risk of BC (T2-4, N1-3, grade 2–3, ER negative or Her2 positive), regardless of BMD values [110, 111].

BTA Safety

BTAs are generally well tolerated, especially at the dosage recommended for CTIBL prevention [70].
Intravenous BP, such as zoledronate, seem to be more frequently associated with acute phase reactions than the oral ones. Symptoms include transient bone pain, arthralgia or myalgia, fever and nausea [111]. On the other hand, oral BP can cause esophagitis and other gastrointestinal disorders; for this reason, it is advised to take them on an empty stomach and remain upright for at least 30 min [111].
Hypocalcaemia is more likely to occur during denosumab treatment but can easily be prevented by the regular monitoring of serum calcium levels and the concomitant administration of calcium and vitamin D supplements [70].
Osteonecrosis of the jaw (ONJ) is one of the most important adverse events occurring during BTA treatment. However, it is more common (incidence of approximately 1.3%) in bone metastatic patients who receive BTAs on a monthly basis, rather than those undergoing less intensive treatment for osteoporosis or CTIBL management [49].
Nevertheless, a careful dental examination and appropriate preventive dentistry are recommended before BTA administration, and patients should be advised to maintain good oral hygiene. If possible, invasive dental procedures, such as tooth extractions and implants, should be avoided during BTA treatment [49, 70].

Conclusion

The improved survival of patients with BC and PC has led to increasing awareness of survivorship issues, including the long-term consequences of cancer treatment. Current treatments for both BC and PC adversely affect bone health via several mechanisms, increasing the risk of osteoporosis and fracture. These potentially devastating complications may be avoided by lifestyle changes and the use of BTAs, with zoledronate and denosumab being the most comprehensively studied agents. Zoledronate has been shown to prevent the loss of BMD in women with BC who experience premature menopause, in postmenopausal women receiving an AI and in men with PC undergoing ADT. However, these improvements have unfortunately not been translated into a clear reduction in fracture incidence. Denosumab is licensed for the prevention of CTIBL due to its proven ability to reduce the incidence of fractures and has become the treatment of choice for patients at high risk of fracture. Recently published guidelines are available to guide the assessment and treatment of CTIBL in both PC and BC. There is a need to improve awareness of CTIBL amongst clinicians and members of the multidisciplinary team, in order to ensure that these are widely and consistently implemented in clinical practice and that patients receive the best possible treatment.

Acknowledgements

We are grateful to Weston Park Cancer Charity for an award to JB providing financial support for CH.

Conflict of interest

REC—research funding from Amgen. JB—consultancy/Advisory Board Membership for Amgen and Novartis. CH and SD—no conflicts of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Literatur
3.
Zurück zum Zitat Carnevale V, Romagnoli E, Cipriani C, Del Fiacco R, Piemonte S, Pepe J, Scillitani A, Minisola S (2010) Sex hormones and bone health in males. Arch Biochem Biophys 503(1):110–117PubMedCrossRef Carnevale V, Romagnoli E, Cipriani C, Del Fiacco R, Piemonte S, Pepe J, Scillitani A, Minisola S (2010) Sex hormones and bone health in males. Arch Biochem Biophys 503(1):110–117PubMedCrossRef
4.
Zurück zum Zitat Wilson JD (2001) The role of 5α reductase in steroid hormone physiology. Reprod Fertil Dev 13(7–8):673–678PubMedCrossRef Wilson JD (2001) The role of 5α reductase in steroid hormone physiology. Reprod Fertil Dev 13(7–8):673–678PubMedCrossRef
6.
Zurück zum Zitat Kasperk CH, Wakely GK, Hierl T, Ziegler R (1997) Gonadal and adrenal androgens are potent regulators of human bone cell metabolism in vitro. J Bone Miner Res 12(3):464–471PubMedCrossRef Kasperk CH, Wakely GK, Hierl T, Ziegler R (1997) Gonadal and adrenal androgens are potent regulators of human bone cell metabolism in vitro. J Bone Miner Res 12(3):464–471PubMedCrossRef
7.
Zurück zum Zitat Vanderschueren D, Vandenput L, Boonen S, Lindberg MK, Bouillon R, Ohlsson C (2012) Androgens and bone. Minerva Endocrinol 37(4):305–314 Vanderschueren D, Vandenput L, Boonen S, Lindberg MK, Bouillon R, Ohlsson C (2012) Androgens and bone. Minerva Endocrinol 37(4):305–314
8.
Zurück zum Zitat Huber DM, Bendixen AC, Pathrose P, Srivastava S, Dienger KM, Shevde NK, Pike JW (2001) Androgens suppress osteoclast formation induced by RANKL and macrophage-colony stimulating factor. Endocrinology 142(9):3800–3808PubMedCrossRef Huber DM, Bendixen AC, Pathrose P, Srivastava S, Dienger KM, Shevde NK, Pike JW (2001) Androgens suppress osteoclast formation induced by RANKL and macrophage-colony stimulating factor. Endocrinology 142(9):3800–3808PubMedCrossRef
9.
Zurück zum Zitat Boyle WJ, Simonet WS, Lacey DL (2003) Osteoclast differentiation and activation. Nature 423(6937):337–342PubMedCrossRef Boyle WJ, Simonet WS, Lacey DL (2003) Osteoclast differentiation and activation. Nature 423(6937):337–342PubMedCrossRef
10.
Zurück zum Zitat Hofbauer LC, Hicok KC, Chen D, Khosla S (2001) Regulation of osteoprotegerin production by androgens and anti-androgens in human osteoblastic lineage cells. Eur J Endocrinol 147(2):269–273CrossRef Hofbauer LC, Hicok KC, Chen D, Khosla S (2001) Regulation of osteoprotegerin production by androgens and anti-androgens in human osteoblastic lineage cells. Eur J Endocrinol 147(2):269–273CrossRef
11.
Zurück zum Zitat Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, Riggs BL (1999) Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 140(9):4367–4370PubMedCrossRef Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, Riggs BL (1999) Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 140(9):4367–4370PubMedCrossRef
12.
Zurück zum Zitat Kameda T, Mano H, Yuasa T, Mori Y, Miyazawa K, Shiokawa M, Nakamaru Y, Hiroi E, Hiura K, Kameda A, Yang NN, Hakeda Y, Kumegawa M (1997) Estrogen inhibits bone resorption by directly inducing apoptosis of the bone-resorbing osteoclasts. J Exp Med 186(4):489–495PubMedPubMedCentralCrossRef Kameda T, Mano H, Yuasa T, Mori Y, Miyazawa K, Shiokawa M, Nakamaru Y, Hiroi E, Hiura K, Kameda A, Yang NN, Hakeda Y, Kumegawa M (1997) Estrogen inhibits bone resorption by directly inducing apoptosis of the bone-resorbing osteoclasts. J Exp Med 186(4):489–495PubMedPubMedCentralCrossRef
13.
14.
Zurück zum Zitat Emerton KB, Hu B, Woo AA, Sinofski A, Hernandez C, Majeska RJ, Jepsen KJ, Schaffler MB (2010) Osteocyte apoptosis and control of bone resorption following ovariectomy in mice. Bone 46(3):577–583PubMedCrossRef Emerton KB, Hu B, Woo AA, Sinofski A, Hernandez C, Majeska RJ, Jepsen KJ, Schaffler MB (2010) Osteocyte apoptosis and control of bone resorption following ovariectomy in mice. Bone 46(3):577–583PubMedCrossRef
15.
Zurück zum Zitat Tomkinson A, Reeve J, Shaw RW, Noble BS (1997) The death of osteocytes via apoptosis accompanies estrogen withdrawal in human bone. J Clin Endocrinol Metab 82(9):3128–3135PubMed Tomkinson A, Reeve J, Shaw RW, Noble BS (1997) The death of osteocytes via apoptosis accompanies estrogen withdrawal in human bone. J Clin Endocrinol Metab 82(9):3128–3135PubMed
17.
Zurück zum Zitat Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K (1995) Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metab 80(12):3689–3698PubMed Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K (1995) Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metab 80(12):3689–3698PubMed
18.
Zurück zum Zitat Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS (1994) Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. New Engl J Med 331(16):1056–1061PubMedCrossRef Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS (1994) Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. New Engl J Med 331(16):1056–1061PubMedCrossRef
19.
Zurück zum Zitat Falahati-Nini A, Riggs BL, Atkinson EJ, O’Fallon WM, Eastell R, Khosla S (2000) Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J Clin Invest 106(12):1553–1560PubMedPubMedCentralCrossRef Falahati-Nini A, Riggs BL, Atkinson EJ, O’Fallon WM, Eastell R, Khosla S (2000) Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J Clin Invest 106(12):1553–1560PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Nicks KM, Perrien DS, Akel NS, Suva LJ, Gaddy D (2009) Regulation of osteoblastogenesis and osteoclastogenesis by the other reproductive hormones, Activin and Inhibin. Mol Cell Endocrinol 310(1–2):11–20PubMedPubMedCentralCrossRef Nicks KM, Perrien DS, Akel NS, Suva LJ, Gaddy D (2009) Regulation of osteoblastogenesis and osteoclastogenesis by the other reproductive hormones, Activin and Inhibin. Mol Cell Endocrinol 310(1–2):11–20PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Wilson C, Holen I, Coleman RE (2012) Seed, soil and secreted hormones: potential interactions of breast cancer cells with their endocrine/paracrine microenvironment and implications for treatment with bisphosphonates. Cancer Treat Rev 38:877–889PubMedCrossRef Wilson C, Holen I, Coleman RE (2012) Seed, soil and secreted hormones: potential interactions of breast cancer cells with their endocrine/paracrine microenvironment and implications for treatment with bisphosphonates. Cancer Treat Rev 38:877–889PubMedCrossRef
23.
Zurück zum Zitat Vale W, Wiater E, Gray P, arrison C, Bilezikjian L, Choe S (2004) Activins and Inhibins and their signalling. Ann N Y Acad Sci 1038:142–147PubMedCrossRef Vale W, Wiater E, Gray P, arrison C, Bilezikjian L, Choe S (2004) Activins and Inhibins and their signalling. Ann N Y Acad Sci 1038:142–147PubMedCrossRef
24.
Zurück zum Zitat Ying SY, Zhang Z (1996) Expression and localization of inhibin/activin subunits and activin receptors in MCF-7 cells, a human breast cancer cell line. Breast Cancer Res Treat 37(2):151–160PubMedCrossRef Ying SY, Zhang Z (1996) Expression and localization of inhibin/activin subunits and activin receptors in MCF-7 cells, a human breast cancer cell line. Breast Cancer Res Treat 37(2):151–160PubMedCrossRef
25.
Zurück zum Zitat Perrien DS, Achenbach SJ, Bledsoe SE, Walser B, Suva LJ, Khosla S, Gaddy D (2006) Bone turnover across the menopause transition: correlations with inhibins and follicle-stimulating hormone. J Clin Endocrinol Metab 91(5):1848–1854PubMedCrossRef Perrien DS, Achenbach SJ, Bledsoe SE, Walser B, Suva LJ, Khosla S, Gaddy D (2006) Bone turnover across the menopause transition: correlations with inhibins and follicle-stimulating hormone. J Clin Endocrinol Metab 91(5):1848–1854PubMedCrossRef
26.
Zurück zum Zitat Greenspan SL, Coates P, Sereika SM, Nelson JB, Trump DL, Resnick NM (2005) Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J Clin Endocrinol Metab 90(12):6410–6417PubMedCrossRef Greenspan SL, Coates P, Sereika SM, Nelson JB, Trump DL, Resnick NM (2005) Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J Clin Endocrinol Metab 90(12):6410–6417PubMedCrossRef
27.
Zurück zum Zitat Daniell HW, Dunn SR, Ferguson DW, Lomas G, Niazi Z, Stratte PT (2000) Progressive osteoporosis during androgen deprivation therapy for prostate cancer. J Urol 163(1):181–186PubMedCrossRef Daniell HW, Dunn SR, Ferguson DW, Lomas G, Niazi Z, Stratte PT (2000) Progressive osteoporosis during androgen deprivation therapy for prostate cancer. J Urol 163(1):181–186PubMedCrossRef
28.
Zurück zum Zitat Goldray D, Weisman Y, Jaccard N, Merdler C, Chen J, Matzkin H (1993) Decreased bone density in elderly men treated with the gonadotropin-releasing hormone agonist decapeptyl (D-Trp6-GnRH). J Clin Endocrinol Metab 76(2):288–290PubMed Goldray D, Weisman Y, Jaccard N, Merdler C, Chen J, Matzkin H (1993) Decreased bone density in elderly men treated with the gonadotropin-releasing hormone agonist decapeptyl (D-Trp6-GnRH). J Clin Endocrinol Metab 76(2):288–290PubMed
29.
Zurück zum Zitat Morote J, Morin JP, Orsola A, Abascal JM, Salvador C, Trilla E, Raventos CX, Cecchini L, Encabo G, Reventos J (2007) Prevalence of osteoporosis during long-term androgen deprivation therapy in patients with prostate cancer. Urology 69(3):500–504PubMedCrossRef Morote J, Morin JP, Orsola A, Abascal JM, Salvador C, Trilla E, Raventos CX, Cecchini L, Encabo G, Reventos J (2007) Prevalence of osteoporosis during long-term androgen deprivation therapy in patients with prostate cancer. Urology 69(3):500–504PubMedCrossRef
30.
Zurück zum Zitat Bruder JM, Ma JZ, Basler JW, Welch MD (2006) Prevalence of osteopenia and osteoporosis by central and peripheral bone mineral density in men with prostate cancer during androgen-deprivation therapy. Urology 67(1):152–155PubMedCrossRef Bruder JM, Ma JZ, Basler JW, Welch MD (2006) Prevalence of osteopenia and osteoporosis by central and peripheral bone mineral density in men with prostate cancer during androgen-deprivation therapy. Urology 67(1):152–155PubMedCrossRef
32.
Zurück zum Zitat James ND, Sydes MR, Clarke NW, Mason MD, Dearnaley DP, Spears MR, Ritchie AW, Parker CC, Russell JM, Attard G, de Bono J, Cross W, Jones RJ, Thalmann G, Amos C, Matheson D, Millman R, Alzouebi M, Beesley S, Birtle AJ, Brock S, Cathomas R, Chakraborti P, Chowdhury S, Cook A, Elliott T, Gale J, Gibbs S, Graham JD, Hetherington J, Hughes R, Laing R, McKinna F, McLaren DB, O’Sullivan JM, Parikh O, Peedell C, Protheroe A, Robinson AJ, Srihari N, Srinivasan R, Staffurth J, Sundar S, Tolan S, Tsang D, Wagstaff J, Parmar MK, STAMPEDE investigators (2016) 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 387(10024):1163–1177PubMedPubMedCentralCrossRef James ND, Sydes MR, Clarke NW, Mason MD, Dearnaley DP, Spears MR, Ritchie AW, Parker CC, Russell JM, Attard G, de Bono J, Cross W, Jones RJ, Thalmann G, Amos C, Matheson D, Millman R, Alzouebi M, Beesley S, Birtle AJ, Brock S, Cathomas R, Chakraborti P, Chowdhury S, Cook A, Elliott T, Gale J, Gibbs S, Graham JD, Hetherington J, Hughes R, Laing R, McKinna F, McLaren DB, O’Sullivan JM, Parikh O, Peedell C, Protheroe A, Robinson AJ, Srihari N, Srinivasan R, Staffurth J, Sundar S, Tolan S, Tsang D, Wagstaff J, Parmar MK, STAMPEDE investigators (2016) 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 387(10024):1163–1177PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Buehring B, Viswanathan R, Binkley N, Busse W (2013) Glucocorticoid-induced osteoporosis: an update on effects and management. J Allergy Clin Immunol 132(5):1019–1030PubMedCrossRef Buehring B, Viswanathan R, Binkley N, Busse W (2013) Glucocorticoid-induced osteoporosis: an update on effects and management. J Allergy Clin Immunol 132(5):1019–1030PubMedCrossRef
34.
Zurück zum Zitat Guise TA (2006) Bone loss and fracture risk associated with cancer therapy. Oncologist 11:1121–1131PubMedCrossRef Guise TA (2006) Bone loss and fracture risk associated with cancer therapy. Oncologist 11:1121–1131PubMedCrossRef
35.
Zurück zum Zitat Oefelein MG, Ricchuiti V, Conrad W, Seftel A, Bodner D, Goldman H, Resnick M (2001) Skeletal fracture associated with androgen suppression induced osteoporosis: the clinical incidence and risk factors for patients with prostate cancer. J Urol 166(5):1724–1728PubMedCrossRef Oefelein MG, Ricchuiti V, Conrad W, Seftel A, Bodner D, Goldman H, Resnick M (2001) Skeletal fracture associated with androgen suppression induced osteoporosis: the clinical incidence and risk factors for patients with prostate cancer. J Urol 166(5):1724–1728PubMedCrossRef
36.
Zurück zum Zitat Shahinian VB, Kuo YF, Freeman JL, Goodwin JS (2005) Risk of fracture after androgen deprivation for prostate cancer. New Engl J Med 352:154–164PubMedCrossRef Shahinian VB, Kuo YF, Freeman JL, Goodwin JS (2005) Risk of fracture after androgen deprivation for prostate cancer. New Engl J Med 352:154–164PubMedCrossRef
37.
Zurück zum Zitat Smith MR, Lee WC, Brandman J, Wang Q, Botteman M, Pashos CL (2005) Gonadotropin-releasing hormone agonists and fracture risk: a claims-based cohort study of men with nonmetastatic prostate cancer. J Clin Oncol 23:7897–7903PubMedCrossRef Smith MR, Lee WC, Brandman J, Wang Q, Botteman M, Pashos CL (2005) Gonadotropin-releasing hormone agonists and fracture risk: a claims-based cohort study of men with nonmetastatic prostate cancer. J Clin Oncol 23:7897–7903PubMedCrossRef
38.
Zurück zum Zitat Nguyen PL, Alibhai SMH, Basaria S, D’Amico AV, Kantoff PW, Keating NL, Penson DF, Rosario DJ, Tombal B, Smith MR (2015) Adverse effects of androgen deprivation therapy and strategies to mitigate them. Eur Urol 67:825–836PubMedCrossRef Nguyen PL, Alibhai SMH, Basaria S, D’Amico AV, Kantoff PW, Keating NL, Penson DF, Rosario DJ, Tombal B, Smith MR (2015) Adverse effects of androgen deprivation therapy and strategies to mitigate them. Eur Urol 67:825–836PubMedCrossRef
39.
Zurück zum Zitat D’Oronzo S, Stucci S, Tucci M, Silvestris F (2015) Cancer treatment-induced bone loss (CTIBL): pathogenesis and clinical implications. Cancer Treat Rev 41(9):798–808PubMedCrossRef D’Oronzo S, Stucci S, Tucci M, Silvestris F (2015) Cancer treatment-induced bone loss (CTIBL): pathogenesis and clinical implications. Cancer Treat Rev 41(9):798–808PubMedCrossRef
40.
Zurück zum Zitat Pfeilschifter J, Diel IJ (2000) Osteoporosis due to cancer treatment: pathogenesis and management. J Clin Oncol 18:1570–1593PubMedCrossRef Pfeilschifter J, Diel IJ (2000) Osteoporosis due to cancer treatment: pathogenesis and management. J Clin Oncol 18:1570–1593PubMedCrossRef
41.
Zurück zum Zitat Stava CJ, Jimenez C, Hu MI, Vassilopoulou-Sellin R (2009) Skeletal sequelae of cancer and cancer treatment. J Cancer Surviv 3(1):75–88PubMedCrossRef Stava CJ, Jimenez C, Hu MI, Vassilopoulou-Sellin R (2009) Skeletal sequelae of cancer and cancer treatment. J Cancer Surviv 3(1):75–88PubMedCrossRef
42.
Zurück zum Zitat Soleimani R, Heytens E, Darzynkiewicz Z, Oktay K (2011) Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise. Ageing 3(8):782–793 Soleimani R, Heytens E, Darzynkiewicz Z, Oktay K (2011) Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise. Ageing 3(8):782–793
43.
Zurück zum Zitat Rana T, Chakrabarti A, Freeman M, Biswas S (2013) Doxorubicin-mediated bone loss in breast cancer bone metastases is driven by an interplay between oxidative stress and induction of TGFβ. PLoS ONE 8(10):e78043PubMedPubMedCentralCrossRef Rana T, Chakrabarti A, Freeman M, Biswas S (2013) Doxorubicin-mediated bone loss in breast cancer bone metastases is driven by an interplay between oxidative stress and induction of TGFβ. PLoS ONE 8(10):e78043PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Shapiro C, Manola J, Leboff M (2001) Ovarian failure after adjuvant chemotherapy is associated with rapid bone loss in women with early-stage breast cancer. J Clin Oncol 19(14):3306–3311PubMedCrossRef Shapiro C, Manola J, Leboff M (2001) Ovarian failure after adjuvant chemotherapy is associated with rapid bone loss in women with early-stage breast cancer. J Clin Oncol 19(14):3306–3311PubMedCrossRef
45.
Zurück zum Zitat Vehmanen L, Saarto T, Risteli J, Risteli L, Blomqvist C, Elomaa I (2004) Short-term intermittent intravenous clodronate in the prevention of bone loss related to chemotherapy-induced ovarian failure. Breast Cancer Res Treat 87(2):181–188PubMedCrossRef Vehmanen L, Saarto T, Risteli J, Risteli L, Blomqvist C, Elomaa I (2004) Short-term intermittent intravenous clodronate in the prevention of bone loss related to chemotherapy-induced ovarian failure. Breast Cancer Res Treat 87(2):181–188PubMedCrossRef
46.
Zurück zum Zitat Hershman DL, McMahon DJ, Crew KD, Cremers S, Irani D, Cucchiara G, Brafman L, Shane E (2008) Zoledronic acid prevents bone loss in premenopausal women undergoing adjuvant chemotherapy for early-stage breast cancer. Clin Oncol 26(29):4739–4745CrossRef Hershman DL, McMahon DJ, Crew KD, Cremers S, Irani D, Cucchiara G, Brafman L, Shane E (2008) Zoledronic acid prevents bone loss in premenopausal women undergoing adjuvant chemotherapy for early-stage breast cancer. Clin Oncol 26(29):4739–4745CrossRef
47.
Zurück zum Zitat Cameron DA, Douglas S, Brown JE, Anderson RA (2010) Bone mineral density loss during adjuvant chemotherapy in premenopausal women with early breast cancer: is it dependent on oestrogen deficiency? Breast Cancer Res Treat 123(3):805–814PubMedCrossRef Cameron DA, Douglas S, Brown JE, Anderson RA (2010) Bone mineral density loss during adjuvant chemotherapy in premenopausal women with early breast cancer: is it dependent on oestrogen deficiency? Breast Cancer Res Treat 123(3):805–814PubMedCrossRef
48.
Zurück zum Zitat Christensen C, Cronin-Fenton D, FrØslev T, Hermann AP, Ewertz M (2016) Change in bone mineral density during adjuvant chemotherapy for early-stage breast cancer. Support Care Cancer 24(10):4229–4236PubMedPubMedCentralCrossRef Christensen C, Cronin-Fenton D, FrØslev T, Hermann AP, Ewertz M (2016) Change in bone mineral density during adjuvant chemotherapy for early-stage breast cancer. Support Care Cancer 24(10):4229–4236PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Hadji P, Coleman RE, Wilson C, Powles TJ, Clézardin PA, Costa M, Body L, Markopoulos JJ, Santini C, Diel D, Di Leo I, Cameron A, Dodwell D, Smith D, Gnant I, Gray M, Harbeck R, Thurlimann N, Untch B, Cortes M, Martin J, Albert M, Conte US, Ejlertsen PF, Bergh B, Kaufmann J, Holen M I (2016) Adjuvant bisphosphonates in early breast cancer: consensus guidance for clinical practice from a European Panel. Ann Oncol 27:379–390PubMedCrossRef Hadji P, Coleman RE, Wilson C, Powles TJ, Clézardin PA, Costa M, Body L, Markopoulos JJ, Santini C, Diel D, Di Leo I, Cameron A, Dodwell D, Smith D, Gnant I, Gray M, Harbeck R, Thurlimann N, Untch B, Cortes M, Martin J, Albert M, Conte US, Ejlertsen PF, Bergh B, Kaufmann J, Holen M I (2016) Adjuvant bisphosphonates in early breast cancer: consensus guidance for clinical practice from a European Panel. Ann Oncol 27:379–390PubMedCrossRef
50.
Zurück zum Zitat Fogelman I, Blake GM, Blamey R, Palmer M, Sauerbrei W, Schumacher M, Serin D, Stewart A, Wilpshaar W (2003) Bone mineral density in premenopausal women treated for node-positive early breast cancer with 2 years of goserelin or 6 months of cyclophosphamide, methotrexate and 5-fluorouracil (CMF). Osteoporos Int 14:1001–1006PubMedCrossRef Fogelman I, Blake GM, Blamey R, Palmer M, Sauerbrei W, Schumacher M, Serin D, Stewart A, Wilpshaar W (2003) Bone mineral density in premenopausal women treated for node-positive early breast cancer with 2 years of goserelin or 6 months of cyclophosphamide, methotrexate and 5-fluorouracil (CMF). Osteoporos Int 14:1001–1006PubMedCrossRef
51.
Zurück zum Zitat Gnant M, Mlineritsch B, Luschin-Ebengreuth G, Kainberger F, Kässmann H, Piswanger-Sölkner JC, Seifert M, Ploner F, Menzel C, Dubsky P, Fitzal F, Bjelic-Radisic V, Steger G, Greil R, Marth C, Kubista E, Samonigg H, Wohlmuth P, Mittlböck M, Jakesz R; Austrian Breast and Colorectal Cancer Study Group (ABCSG) (2008) Austrian Breast and Colorectal Cancer Study Group (ABCSG). Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 5-year follow-up of the ABCSG-12 bone-mineral density substudy. Lancet Oncol 9(9):840–849PubMedCrossRef Gnant M, Mlineritsch B, Luschin-Ebengreuth G, Kainberger F, Kässmann H, Piswanger-Sölkner JC, Seifert M, Ploner F, Menzel C, Dubsky P, Fitzal F, Bjelic-Radisic V, Steger G, Greil R, Marth C, Kubista E, Samonigg H, Wohlmuth P, Mittlböck M, Jakesz R; Austrian Breast and Colorectal Cancer Study Group (ABCSG) (2008) Austrian Breast and Colorectal Cancer Study Group (ABCSG). Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 5-year follow-up of the ABCSG-12 bone-mineral density substudy. Lancet Oncol 9(9):840–849PubMedCrossRef
52.
Zurück zum Zitat Powles TJ, Hickish T, Kanis JA, Tidy A, Ashley S (1996) Effect of tamoxifen on bone mineral density measured by dual-energy X-Ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 14(1):78–84PubMedCrossRef Powles TJ, Hickish T, Kanis JA, Tidy A, Ashley S (1996) Effect of tamoxifen on bone mineral density measured by dual-energy X-Ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 14(1):78–84PubMedCrossRef
53.
Zurück zum Zitat Hadji P, Ziller M, Kieback DG, Menschik T, Kalder M, Kuck J, Hasenburg A (2009) The effect of exemestane or tamoxifen on markers of bone turnover: results of a German sub-study of the Tamoxifen Exemestane Adjuvant Multicentre (TEAM) trial. The Breast 18:159–164PubMedCrossRef Hadji P, Ziller M, Kieback DG, Menschik T, Kalder M, Kuck J, Hasenburg A (2009) The effect of exemestane or tamoxifen on markers of bone turnover: results of a German sub-study of the Tamoxifen Exemestane Adjuvant Multicentre (TEAM) trial. The Breast 18:159–164PubMedCrossRef
54.
Zurück zum Zitat Kalder M, Hans D, Kyvernitakis I, Lamy O, Bauer M, Hadji P (2014) Effects of exemestane and tamoxifen treatment on bone texture analysis assessed by TBS in comparison with bone mineral density assessed by DXA in women with breast cancer. J Clin Densitom Assess Manag Musculoskelet Health 17(1):66–71CrossRef Kalder M, Hans D, Kyvernitakis I, Lamy O, Bauer M, Hadji P (2014) Effects of exemestane and tamoxifen treatment on bone texture analysis assessed by TBS in comparison with bone mineral density assessed by DXA in women with breast cancer. J Clin Densitom Assess Manag Musculoskelet Health 17(1):66–71CrossRef
55.
Zurück zum Zitat Bauer M, Bryce J, Hadji P (2012) Aromatase inhibitor-associated bone loss and its management with bisphosphonates in patients with breast cancer. Breast Cancer Targets Ther 4:91–101CrossRef Bauer M, Bryce J, Hadji P (2012) Aromatase inhibitor-associated bone loss and its management with bisphosphonates in patients with breast cancer. Breast Cancer Targets Ther 4:91–101CrossRef
56.
Zurück zum Zitat Forbes JF, Cuzick J, Buzdar A, Howell A, Tobias JS, Baum M, Arimidex, Tamoxifen, Alone or in Combination (ATAC) Trialists’ Group (2008) Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 100-month analysis of the ATAC trial. Lancet Oncol 9(1):45–53PubMedCrossRef Forbes JF, Cuzick J, Buzdar A, Howell A, Tobias JS, Baum M, Arimidex, Tamoxifen, Alone or in Combination (ATAC) Trialists’ Group (2008) Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 100-month analysis of the ATAC trial. Lancet Oncol 9(1):45–53PubMedCrossRef
57.
Zurück zum Zitat Eastell R, Adams JE, Coleman RE, Howell A, Hannon RA, Cuzick J, Mackey JR, Beckmann MW, Clack G (2008) Effect of anastrozole on bone mineral density: 5-year results from the anastrozole, tamoxifen, alone or in combination trial 18233230. J Clin Oncol 26(7):1051–1058PubMedCrossRef Eastell R, Adams JE, Coleman RE, Howell A, Hannon RA, Cuzick J, Mackey JR, Beckmann MW, Clack G (2008) Effect of anastrozole on bone mineral density: 5-year results from the anastrozole, tamoxifen, alone or in combination trial 18233230. J Clin Oncol 26(7):1051–1058PubMedCrossRef
58.
Zurück zum Zitat Eastell R, Adams J, Clack G, Howell A, Cuzick J, Mackey J, Beckmann MW, Coleman RE (2011) Long-term effects of anastrozole on bone mineral density: 7-year results from the ATAC trial. Ann Oncol 22(4):857–862PubMedCrossRef Eastell R, Adams J, Clack G, Howell A, Cuzick J, Mackey J, Beckmann MW, Coleman RE (2011) Long-term effects of anastrozole on bone mineral density: 7-year results from the ATAC trial. Ann Oncol 22(4):857–862PubMedCrossRef
59.
Zurück zum Zitat Rabaglio M, Sun Z, Price KN, Castiglione-Gertsch M, Hawle H, Thurlimann B, Mouridsen H, Campone M, Forbes JF, Paridaens RJ, Colleoni M, Pienkowski T, Nogaret JM, Láng I, Smith I, Gelber RD, Goldhirsch A, Coates AS, BIG 1–98 Collaborative and International Breast Cancer Study Groups (2009) Bone fractures among postmenopausal patients with endocrine-responsive early breast cancer treated with 5 years of letrozole or tamoxifen in the BIG 1–98 trial. Ann Oncol 20(9):1489–1498PubMedPubMedCentralCrossRef Rabaglio M, Sun Z, Price KN, Castiglione-Gertsch M, Hawle H, Thurlimann B, Mouridsen H, Campone M, Forbes JF, Paridaens RJ, Colleoni M, Pienkowski T, Nogaret JM, Láng I, Smith I, Gelber RD, Goldhirsch A, Coates AS, BIG 1–98 Collaborative and International Breast Cancer Study Groups (2009) Bone fractures among postmenopausal patients with endocrine-responsive early breast cancer treated with 5 years of letrozole or tamoxifen in the BIG 1–98 trial. Ann Oncol 20(9):1489–1498PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Goss PE, Ingle JN, Martino S, Robert NJ, Muss HB, Piccart MJ, Castiglione M, Tu D, Shepherd LE, Pritchard KI, Livingston RB, Davidson NE, Norton L, Perez EA, Abrams JS, Therasse P, Palmer MJ, Pater JL (2003) A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. New Engl J Med 349(19):557–568CrossRef Goss PE, Ingle JN, Martino S, Robert NJ, Muss HB, Piccart MJ, Castiglione M, Tu D, Shepherd LE, Pritchard KI, Livingston RB, Davidson NE, Norton L, Perez EA, Abrams JS, Therasse P, Palmer MJ, Pater JL (2003) A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. New Engl J Med 349(19):557–568CrossRef
61.
Zurück zum Zitat Coleman RE, Banks LM, Girgis SI, Kilburn LS, Vrdoljak E, Fox J, Cawthorn SJ, Patel A, Snowdon CF, Hall E, Bliss JM, Coombes RC, Intergroup Exemestane Study Group (2007) Skeletal effects of exemestane on bone-mineral density, bone biomarkers, and fracture incidence in postmenopausal women with early breast cancer participating in the Intergroup Exemestane Study (IES): a randomised controlled study. Lancet Oncol 8(2):119–127PubMedCrossRef Coleman RE, Banks LM, Girgis SI, Kilburn LS, Vrdoljak E, Fox J, Cawthorn SJ, Patel A, Snowdon CF, Hall E, Bliss JM, Coombes RC, Intergroup Exemestane Study Group (2007) Skeletal effects of exemestane on bone-mineral density, bone biomarkers, and fracture incidence in postmenopausal women with early breast cancer participating in the Intergroup Exemestane Study (IES): a randomised controlled study. Lancet Oncol 8(2):119–127PubMedCrossRef
62.
Zurück zum Zitat Goss PE, Ingle JN, Pritchard KI, Ellis MJ, Sledge GW, Budd GT, Rabaglio M, Ansari RH, Johnson DB, Tozer R, D’Souza DP, Chalchal H, Spadafora S, Stearns V, Perez EA, Liedke PE, Lang I, Elliott C, Gelmon KA, Chapman JA, Shepherd LE (2013) Exemestane versus anastrozole in postmenopausal women with early breast cancer: NCIC CTG MA.27-a randomized controlled phase III trial. J Clin Oncol 31(11):1398–1404PubMedPubMedCentralCrossRef Goss PE, Ingle JN, Pritchard KI, Ellis MJ, Sledge GW, Budd GT, Rabaglio M, Ansari RH, Johnson DB, Tozer R, D’Souza DP, Chalchal H, Spadafora S, Stearns V, Perez EA, Liedke PE, Lang I, Elliott C, Gelmon KA, Chapman JA, Shepherd LE (2013) Exemestane versus anastrozole in postmenopausal women with early breast cancer: NCIC CTG MA.27-a randomized controlled phase III trial. J Clin Oncol 31(11):1398–1404PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Hong AR, Kim JH, Lee KH, Kim TY, Im SA, Kim TY, Moon HG, Han WS, Noh DY, Kim SW, Shin CS (2017) Long-term effect of aromatase inhibitors on bone microarchitecture and macroarchitecture in non-osteoporotic postmenopausal women with breast cancer. Osteoporos Int 28(4):1413–1422PubMedCrossRef Hong AR, Kim JH, Lee KH, Kim TY, Im SA, Kim TY, Moon HG, Han WS, Noh DY, Kim SW, Shin CS (2017) Long-term effect of aromatase inhibitors on bone microarchitecture and macroarchitecture in non-osteoporotic postmenopausal women with breast cancer. Osteoporos Int 28(4):1413–1422PubMedCrossRef
64.
Zurück zum Zitat Napoli N, Rastelli A, Ma C, Yarramaneni J, Vattikutti S, Moskowitz G, Giri T, Mueller C, Kulkarny V, Qualls C, Ellis M, Armamento-Villareal R (2013) Genetic polymorphism at Val80 (rs700518) of the CYP19A1 gene is associated with aromatase inhibitor associated bone loss in women with ER (+) breast cancer. Bone 55(2):309–314PubMedPubMedCentralCrossRef Napoli N, Rastelli A, Ma C, Yarramaneni J, Vattikutti S, Moskowitz G, Giri T, Mueller C, Kulkarny V, Qualls C, Ellis M, Armamento-Villareal R (2013) Genetic polymorphism at Val80 (rs700518) of the CYP19A1 gene is associated with aromatase inhibitor associated bone loss in women with ER (+) breast cancer. Bone 55(2):309–314PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Mazzuca F, Botticelli A, Mazzotti E, La Torre M, Borro M, Marchetti L, Maddalena C, Gentile G, Simmaco M, Marchetti P (2016) CYP19A1 genetic polymorphisms rs4646 and osteoporosis in patients treated with aromatase inhibitor-based adjuvant therapy. Eurasian J Med 48(1):10–14PubMedCrossRef Mazzuca F, Botticelli A, Mazzotti E, La Torre M, Borro M, Marchetti L, Maddalena C, Gentile G, Simmaco M, Marchetti P (2016) CYP19A1 genetic polymorphisms rs4646 and osteoporosis in patients treated with aromatase inhibitor-based adjuvant therapy. Eurasian J Med 48(1):10–14PubMedCrossRef
69.
Zurück zum Zitat Mohler JL, Armstrong AJ, Bahnson RR, Boston B, Busby JE, D’Amico AV, Eastham JA, Enke CA, Farrington T, Higano CS, Horwitz EM, Kantoff PW, Kawachi MH, Kuettel M, Lee RJ, MacVicar GR, Malcolm AW, Miller D, Plimack ER, Pow-Sang JM, Roach M, Rohren E, Rosenfeld S, Srinivas S, Strope SA, Tward J, Twardowski P, Walsh PC, Ho M, Shead DA (2012) Prostate cancer, version 3.2012: featured updates to the NCCN guidelines. J Natl Compr Cancer Netw 10(9):1081–1087CrossRef Mohler JL, Armstrong AJ, Bahnson RR, Boston B, Busby JE, D’Amico AV, Eastham JA, Enke CA, Farrington T, Higano CS, Horwitz EM, Kantoff PW, Kawachi MH, Kuettel M, Lee RJ, MacVicar GR, Malcolm AW, Miller D, Plimack ER, Pow-Sang JM, Roach M, Rohren E, Rosenfeld S, Srinivas S, Strope SA, Tward J, Twardowski P, Walsh PC, Ho M, Shead DA (2012) Prostate cancer, version 3.2012: featured updates to the NCCN guidelines. J Natl Compr Cancer Netw 10(9):1081–1087CrossRef
70.
Zurück zum Zitat Coleman R, Body JJ, Aapro M, Hadji P, Herrstedt J, On behalf of the ESMO guidelines Working Group (2014) Bone health in cancer patients: ESMO clinical practice guidelines. Ann Oncol 25(S3):iii124–i37PubMedCrossRef Coleman R, Body JJ, Aapro M, Hadji P, Herrstedt J, On behalf of the ESMO guidelines Working Group (2014) Bone health in cancer patients: ESMO clinical practice guidelines. Ann Oncol 25(S3):iii124–i37PubMedCrossRef
72.
Zurück zum Zitat Kalder M, Hadji P (2014) Breast cancer and osteoporosis—management of cancer treatment-induced bone loss in postmenopausal women with breast cancer. Breast Care 9(5):312–317PubMedPubMedCentralCrossRef Kalder M, Hadji P (2014) Breast cancer and osteoporosis—management of cancer treatment-induced bone loss in postmenopausal women with breast cancer. Breast Care 9(5):312–317PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Lipton A, Smith MR, Ellis GK, Goessl C (2012) Treatment-induced bone loss and fractures in cancer patients undergoing hormone ablation therapy: efficacy and safety of denosumab. Clin Med Insights Oncol 6:287–303PubMedPubMedCentralCrossRef Lipton A, Smith MR, Ellis GK, Goessl C (2012) Treatment-induced bone loss and fractures in cancer patients undergoing hormone ablation therapy: efficacy and safety of denosumab. Clin Med Insights Oncol 6:287–303PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Agarwal MM, Khandelwal N, Mandal AK, Rana SV, Gupta V, Chandra Mohan V, Kishore GV (2005) Factors affecting bone mineral density in patients with prostate carcinoma before and after orchidectomy. Cancer 103(10):2042–2052PubMedCrossRef Agarwal MM, Khandelwal N, Mandal AK, Rana SV, Gupta V, Chandra Mohan V, Kishore GV (2005) Factors affecting bone mineral density in patients with prostate carcinoma before and after orchidectomy. Cancer 103(10):2042–2052PubMedCrossRef
75.
Zurück zum Zitat Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, McBurnie MA, Cardiovascular Health Study Collaborative Research Group (2001) Frailty in older adults: evidence for a phenotype. J Gerontol Ser A Biol Sci Med Sci 56(3):M146–M156CrossRef Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, McBurnie MA, Cardiovascular Health Study Collaborative Research Group (2001) Frailty in older adults: evidence for a phenotype. J Gerontol Ser A Biol Sci Med Sci 56(3):M146–M156CrossRef
76.
Zurück zum Zitat Galvão DA, Nosaka K, Taaffe DR, Spry N, Kristjanson LJ, McGuigan MR, Suzuki K, Yamaya K, Newton RU (2006) Resistance training and reduction of treatment side effects in prostate cancer. Med Sci Sports Exerc 38(12):2045–2052PubMedCrossRef Galvão DA, Nosaka K, Taaffe DR, Spry N, Kristjanson LJ, McGuigan MR, Suzuki K, Yamaya K, Newton RU (2006) Resistance training and reduction of treatment side effects in prostate cancer. Med Sci Sports Exerc 38(12):2045–2052PubMedCrossRef
77.
Zurück zum Zitat Segal RJ, Reid RD, Courneya KS, Malone SC, Parliament MB, Scott CG, Venner PM, Quinney HA, Jones LW, D’Angelo ME, Wells GA (2003) Resistance exercise in men receiving androgen deprivation therapy for prostate cancer. J Clin Oncol 21(9):1653–1659PubMedCrossRef Segal RJ, Reid RD, Courneya KS, Malone SC, Parliament MB, Scott CG, Venner PM, Quinney HA, Jones LW, D’Angelo ME, Wells GA (2003) Resistance exercise in men receiving androgen deprivation therapy for prostate cancer. J Clin Oncol 21(9):1653–1659PubMedCrossRef
79.
Zurück zum Zitat Planas J, Morote J, Orsola A, Salvador C, Trilla E, Cecchini L, Raventós CX (2007) The relationship between daily calcium intake and bone mineral density in men with prostate cancer. BJU Int 99:812–816PubMedCrossRef Planas J, Morote J, Orsola A, Salvador C, Trilla E, Cecchini L, Raventós CX (2007) The relationship between daily calcium intake and bone mineral density in men with prostate cancer. BJU Int 99:812–816PubMedCrossRef
80.
Zurück zum Zitat Diamond TH, Bucci J, Kersley JH, Aslan P, Lynch WB, Bryant C (2004) Osteoporosis and spinal fractures in men with prostate cancer: risk factors and effects of androgen deprivation therapy. J Urol 172:529–532PubMedCrossRef Diamond TH, Bucci J, Kersley JH, Aslan P, Lynch WB, Bryant C (2004) Osteoporosis and spinal fractures in men with prostate cancer: risk factors and effects of androgen deprivation therapy. J Urol 172:529–532PubMedCrossRef
81.
Zurück zum Zitat Datta M, Schwartz GG (2012) Calcium and vitamin D supplementation during androgen deprivation therapy for prostate cancer: a critical review. Oncologist 17(9):1171–1179PubMedPubMedCentralCrossRef Datta M, Schwartz GG (2012) Calcium and vitamin D supplementation during androgen deprivation therapy for prostate cancer: a critical review. Oncologist 17(9):1171–1179PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Garg A, Leitzel K, Ali S, Lipton A (2015) Antiresorptive therapy in the management of cancer treatment-induced bone loss. Curr Osteoporos Rep 13(2):73–77PubMedCrossRef Garg A, Leitzel K, Ali S, Lipton A (2015) Antiresorptive therapy in the management of cancer treatment-induced bone loss. Curr Osteoporos Rep 13(2):73–77PubMedCrossRef
83.
Zurück zum Zitat Subbiah V, Madsen VS, Raymond AK, Benjamin RS, Ludwig JA (2010) Of mice and men: divergent risks of teriparatide-induced osteosarcoma. Osteoporos Int 21(6):1041–1045PubMedCrossRef Subbiah V, Madsen VS, Raymond AK, Benjamin RS, Ludwig JA (2010) Of mice and men: divergent risks of teriparatide-induced osteosarcoma. Osteoporos Int 21(6):1041–1045PubMedCrossRef
84.
Zurück zum Zitat Smith MR, Fallon MA, Lee H, Finkelstein JS (2004) Raloxifene to prevent gonadotropin-releasing hormone agonist-induced bone loss in men with prostate cancer: a randomized controlled trial. J Clin Endocrinol Metab 89(8):3841–3846PubMedCrossRef Smith MR, Fallon MA, Lee H, Finkelstein JS (2004) Raloxifene to prevent gonadotropin-releasing hormone agonist-induced bone loss in men with prostate cancer: a randomized controlled trial. J Clin Endocrinol Metab 89(8):3841–3846PubMedCrossRef
85.
Zurück zum Zitat Smith MR, Malkowicz SB, Chu F, Forrest J, Price D, Sieber P, Barnette KG, Rodriguez D, Steiner MS (2008) Toremifene increases bone mineral density in men receiving androgen deprivation therapy for prostate cancer: interim analysis of a multicenter phase 3 clinical study. J Urol 179(1):152–155PubMedCrossRef Smith MR, Malkowicz SB, Chu F, Forrest J, Price D, Sieber P, Barnette KG, Rodriguez D, Steiner MS (2008) Toremifene increases bone mineral density in men receiving androgen deprivation therapy for prostate cancer: interim analysis of a multicenter phase 3 clinical study. J Urol 179(1):152–155PubMedCrossRef
86.
Zurück zum Zitat Smith MR, Morton RA, Barnette KG, Sieber PR, Malkowicz SB, Rodriguez D, Hancock ML, Steiner MS (2010) Toremifene to reduce fracture risk in men receiving androgen deprivation therapy for prostate cancer. J Urol 184(4):1316–1321PubMedPubMedCentralCrossRef Smith MR, Morton RA, Barnette KG, Sieber PR, Malkowicz SB, Rodriguez D, Hancock ML, Steiner MS (2010) Toremifene to reduce fracture risk in men receiving androgen deprivation therapy for prostate cancer. J Urol 184(4):1316–1321PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Smith MR, Saad F, Egerdie B, Szwedowski M, Tammela TL, Ke C, Leder BZ, Goessl C (2009) Effects of denosumab on bone mineral density in men receiving androgen deprivation therapy for prostate cancer. J Urol 182(6):2670–2675PubMedPubMedCentralCrossRef Smith MR, Saad F, Egerdie B, Szwedowski M, Tammela TL, Ke C, Leder BZ, Goessl C (2009) Effects of denosumab on bone mineral density in men receiving androgen deprivation therapy for prostate cancer. J Urol 182(6):2670–2675PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Klotz LH, McNeill IY, Kebabdjian M, Zhang L, Chin JL (2013) A phase 3, double-blind, randomised, parallel-group, placebo-controlled study of oral weekly alendronate for the prevention of androgen deprivation bone loss in nonmetastatic prostate cancer: the Cancer and Osteoporosis Research with Alendronate and Leuprolide (CORAL) study. Eur Urol 63(5):927–935PubMedCrossRef Klotz LH, McNeill IY, Kebabdjian M, Zhang L, Chin JL (2013) A phase 3, double-blind, randomised, parallel-group, placebo-controlled study of oral weekly alendronate for the prevention of androgen deprivation bone loss in nonmetastatic prostate cancer: the Cancer and Osteoporosis Research with Alendronate and Leuprolide (CORAL) study. Eur Urol 63(5):927–935PubMedCrossRef
89.
Zurück zum Zitat Michaelson MD, Kaufman DS, Lee H, McGovern FJ, Kantoff PW, Fallon MA, Finkelstein JS, Smith MR (2007) Randomized controlled trial of annual zoledronic acid to prevent gonadotropin-releasing hormone agonist-induced bone loss in men with prostate cancer. J Clin Oncol 25(9):1038–1042PubMedPubMedCentralCrossRef Michaelson MD, Kaufman DS, Lee H, McGovern FJ, Kantoff PW, Fallon MA, Finkelstein JS, Smith MR (2007) Randomized controlled trial of annual zoledronic acid to prevent gonadotropin-releasing hormone agonist-induced bone loss in men with prostate cancer. J Clin Oncol 25(9):1038–1042PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Smith MR, Eastham J, Gleason DM, Shasha D, Tchekmedyians S, Zinner N (2003) Randomized controlled trial of zoledronic acid to prevent bone loss in men receiving androgen deprivation therapy for nonmetastatic prostate cancer. J Urol 169(6):2008–2012PubMedCrossRef Smith MR, Eastham J, Gleason DM, Shasha D, Tchekmedyians S, Zinner N (2003) Randomized controlled trial of zoledronic acid to prevent bone loss in men receiving androgen deprivation therapy for nonmetastatic prostate cancer. J Urol 169(6):2008–2012PubMedCrossRef
91.
Zurück zum Zitat Magno C, Anastasi G, Morabito N, Gaudio A, Maisano D, Franchina F, Galí A, Frisina N, Melloni D (2005) Preventing bone loss during androgen deprivation therapy for prostate cancer: early experience with neridronate. Eur Urol 47(5):575–580PubMedCrossRef Magno C, Anastasi G, Morabito N, Gaudio A, Maisano D, Franchina F, Galí A, Frisina N, Melloni D (2005) Preventing bone loss during androgen deprivation therapy for prostate cancer: early experience with neridronate. Eur Urol 47(5):575–580PubMedCrossRef
92.
Zurück zum Zitat Choo R, Lukka H, Cheung P, Corbett T, Briones-Urbina R, Vieth R, Ehrlich L, Kiss A, Danjoux C (2013) Randomized, double-blinded, placebo-controlled, trial of risedronate for the prevention of bone mineral density loss in nonmetastatic prostate cancer patients receiving radiation therapy plus androgen deprivation therapy. Int J Radiat Oncol Biol Phys 85(5):1239–1245PubMedCrossRef Choo R, Lukka H, Cheung P, Corbett T, Briones-Urbina R, Vieth R, Ehrlich L, Kiss A, Danjoux C (2013) Randomized, double-blinded, placebo-controlled, trial of risedronate for the prevention of bone mineral density loss in nonmetastatic prostate cancer patients receiving radiation therapy plus androgen deprivation therapy. Int J Radiat Oncol Biol Phys 85(5):1239–1245PubMedCrossRef
93.
Zurück zum Zitat Rodrigues P, Hering FO, Bruna P, Meller A, Afonso Y (2007) Comparative study of the protective effect of different intravenous bisphosphonates on the decrease in bone mineral density in patients submitted to radical prostatectomy undergoing androgen deprivation therapy. A prospective open-label controlled study. Int J Urol 14(4):317–320PubMedCrossRef Rodrigues P, Hering FO, Bruna P, Meller A, Afonso Y (2007) Comparative study of the protective effect of different intravenous bisphosphonates on the decrease in bone mineral density in patients submitted to radical prostatectomy undergoing androgen deprivation therapy. A prospective open-label controlled study. Int J Urol 14(4):317–320PubMedCrossRef
94.
Zurück zum Zitat Hadji P, Aapro MS, Body JJ, Gnant M, Brandi ML, Reginster JY, Zillikens MC, Glüer CC, de Villiers T, Baber R, Roodman GD, Cooper C, Langdahl B, Palacios S, Kanis J, Al-Daghri N, Nogues X, Eriksen EF, Kurth A, Rizzoli R, Coleman RE (2017) Management of aromatase inhibitor-associated bone loss (AIBL) in postmenopausal women with hormone sensitive breast cancer: Joint Position Statement of the IOF, CABS. J Bone Oncol 7:1–12PubMedPubMedCentralCrossRef Hadji P, Aapro MS, Body JJ, Gnant M, Brandi ML, Reginster JY, Zillikens MC, Glüer CC, de Villiers T, Baber R, Roodman GD, Cooper C, Langdahl B, Palacios S, Kanis J, Al-Daghri N, Nogues X, Eriksen EF, Kurth A, Rizzoli R, Coleman RE (2017) Management of aromatase inhibitor-associated bone loss (AIBL) in postmenopausal women with hormone sensitive breast cancer: Joint Position Statement of the IOF, CABS. J Bone Oncol 7:1–12PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Trémollieres FA, Ceausu I, Depypere H, Lambrinoudaki I, Mueck A, Pérez-López FR, van der Schouw YT, Senturk LM, Simoncini T, Stevenson JC, Stute P, Rees M (2017) Osteoporosis management in patients with breast cancer: EMAS position statement. Maturitas 95:66–71CrossRef Trémollieres FA, Ceausu I, Depypere H, Lambrinoudaki I, Mueck A, Pérez-López FR, van der Schouw YT, Senturk LM, Simoncini T, Stevenson JC, Stute P, Rees M (2017) Osteoporosis management in patients with breast cancer: EMAS position statement. Maturitas 95:66–71CrossRef
96.
Zurück zum Zitat Saarto T, Vehmanen L, Elomaa I, Välimäki M, Mäkelä P, Blomqvist C (2001) The effect of clodronate and antioestrogens on bone loss associated with oestrogen withdrawal in postmenopausal women with breast cancer. Br J Cancer 84(8):1047–1051PubMedPubMedCentralCrossRef Saarto T, Vehmanen L, Elomaa I, Välimäki M, Mäkelä P, Blomqvist C (2001) The effect of clodronate and antioestrogens on bone loss associated with oestrogen withdrawal in postmenopausal women with breast cancer. Br J Cancer 84(8):1047–1051PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Lester JE, Dodwell D, Purohit OP, Gutcher SA, Ellis SP, Thorpe R, Horsman JM, Brown JE, Hannon RA, Coleman RE (2008) Prevention of anastrozole-induced bone loss with monthly oral ibandronate during adjuvant aromatase inhibitor therapy for breast cancer. Clin Cancer Res 14(19):6336–6342PubMedCrossRef Lester JE, Dodwell D, Purohit OP, Gutcher SA, Ellis SP, Thorpe R, Horsman JM, Brown JE, Hannon RA, Coleman RE (2008) Prevention of anastrozole-induced bone loss with monthly oral ibandronate during adjuvant aromatase inhibitor therapy for breast cancer. Clin Cancer Res 14(19):6336–6342PubMedCrossRef
98.
Zurück zum Zitat Lester JE, Dodwell D, Brown JE, Purohit OP, Gutcher SA, Ellis SP, Thorpe R, Horsman JM, Coleman RE (2012) Prevention of anastrozole induced bone loss with monthly oral ibandronate: Final 5 year results from the ARIBON trial. J Bone Oncol 1:57–62PubMedPubMedCentralCrossRef Lester JE, Dodwell D, Brown JE, Purohit OP, Gutcher SA, Ellis SP, Thorpe R, Horsman JM, Coleman RE (2012) Prevention of anastrozole induced bone loss with monthly oral ibandronate: Final 5 year results from the ARIBON trial. J Bone Oncol 1:57–62PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Van Poznak C, Hannon RA, Mackey JR, Campone M, Apffelstaedt JP, Clack G, Barlow D, Makris A, Eastell R (2010) Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol 28(6):967–975PubMedCrossRef Van Poznak C, Hannon RA, Mackey JR, Campone M, Apffelstaedt JP, Clack G, Barlow D, Makris A, Eastell R (2010) Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol 28(6):967–975PubMedCrossRef
100.
Zurück zum Zitat Sestak I, Singh S, Cuzick J, Blake GM, Patel R, Gossiel F, Coleman R, Dowsett M, Forbes JF, Howell A, Eastell R (2014) Changes in bone mineral density at 3 years in postmenopausal women receiving anastrozole and risedronate in the IBIS-II bone substudy: an international, double-blind, randomised, placebo-controlled trial. Lancet Oncol 15:1460–1468PubMedCrossRef Sestak I, Singh S, Cuzick J, Blake GM, Patel R, Gossiel F, Coleman R, Dowsett M, Forbes JF, Howell A, Eastell R (2014) Changes in bone mineral density at 3 years in postmenopausal women receiving anastrozole and risedronate in the IBIS-II bone substudy: an international, double-blind, randomised, placebo-controlled trial. Lancet Oncol 15:1460–1468PubMedCrossRef
101.
Zurück zum Zitat Brufsky AM, Harker WG, Beck JT, Bosserman L, Vogel C, Seidler C, Jin L, Warsi G, Argonza-Aviles E, Hohneker J, Ericson SG, Perez EA (2012) Final 5-year results of Z-FAST trial: adjuvant zoledronic acid maintains bone mass in postmenopausal breast cancer patients receiving letrozole. Cancer 5:1192–1201CrossRef Brufsky AM, Harker WG, Beck JT, Bosserman L, Vogel C, Seidler C, Jin L, Warsi G, Argonza-Aviles E, Hohneker J, Ericson SG, Perez EA (2012) Final 5-year results of Z-FAST trial: adjuvant zoledronic acid maintains bone mass in postmenopausal breast cancer patients receiving letrozole. Cancer 5:1192–1201CrossRef
102.
Zurück zum Zitat Coleman R, de Boer R, Eidtmann H, Llombart A, Davidson N, Neven P, von Minckwitz G, Sleeboom HP, Forbes J, Barrios C, Frassoldati A, Campbell I, Paija O, Martin N, Modi A, Bundred N (2013) Zoledronic acid (zoledronate) for postmenopausal women with early breast cancer receiving adjuvant letrozole (ZO-FAST study): final 60-month results. Ann Oncol 24(2):398–405PubMedCrossRef Coleman R, de Boer R, Eidtmann H, Llombart A, Davidson N, Neven P, von Minckwitz G, Sleeboom HP, Forbes J, Barrios C, Frassoldati A, Campbell I, Paija O, Martin N, Modi A, Bundred N (2013) Zoledronic acid (zoledronate) for postmenopausal women with early breast cancer receiving adjuvant letrozole (ZO-FAST study): final 60-month results. Ann Oncol 24(2):398–405PubMedCrossRef
103.
Zurück zum Zitat Llombart A, Frassoldati A, Paija O, Sleeboom HP, Jerusalem G, Mebis J, Deleu I, Miller J, Schenk N, Neven P (2012) Immediate administration of zoledronic acid reduces aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer: 12-month analysis of the E-ZO-FAST trial. Clin Breast Cancer 12(1):40–48PubMedCrossRef Llombart A, Frassoldati A, Paija O, Sleeboom HP, Jerusalem G, Mebis J, Deleu I, Miller J, Schenk N, Neven P (2012) Immediate administration of zoledronic acid reduces aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer: 12-month analysis of the E-ZO-FAST trial. Clin Breast Cancer 12(1):40–48PubMedCrossRef
104.
Zurück zum Zitat Wagner-Johnston ND, Sloan JA, Liu H, Kearns AE, Hines SL, Puttabasavaiah S, Dakhil SR, Lafky JM, Perez EA, Loprinzi CL (2015) 5-Year follow-up of a randomized controlled trial of immediate versus delayed zoledronic acid for prevention of bone loss in postmenopausal women with breast cancer starting letrozole after Tamoxifen: N03CC (Alliance). Cancer 121(15):2537–2543PubMedPubMedCentralCrossRef Wagner-Johnston ND, Sloan JA, Liu H, Kearns AE, Hines SL, Puttabasavaiah S, Dakhil SR, Lafky JM, Perez EA, Loprinzi CL (2015) 5-Year follow-up of a randomized controlled trial of immediate versus delayed zoledronic acid for prevention of bone loss in postmenopausal women with breast cancer starting letrozole after Tamoxifen: N03CC (Alliance). Cancer 121(15):2537–2543PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Majithia N, Atherton PJ, Lafky JM, Wagner-Johnston N, Olson J, Dakhil SR, Perez EA, Loprinzi CL, Hines SL (2016) Zoledronic acid for treatment of osteopenia and osteoporosis in women with primary breast cancer undergoing adjuvant aromatase inhibitor therapy: a 5-year follow-up. Support Care Cancer 24:1219–1226PubMedCrossRef Majithia N, Atherton PJ, Lafky JM, Wagner-Johnston N, Olson J, Dakhil SR, Perez EA, Loprinzi CL, Hines SL (2016) Zoledronic acid for treatment of osteopenia and osteoporosis in women with primary breast cancer undergoing adjuvant aromatase inhibitor therapy: a 5-year follow-up. Support Care Cancer 24:1219–1226PubMedCrossRef
106.
Zurück zum Zitat Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Fan M, Kim D (2009) Effect of denosumab on bone mineral density in women receiving adjuvant aromatase inhibitors for non-metastatic breast cancer: subgroup analyses of a phase 3 study. Breast Cancer Res Treat 118:81–87PubMedCrossRef Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Fan M, Kim D (2009) Effect of denosumab on bone mineral density in women receiving adjuvant aromatase inhibitors for non-metastatic breast cancer: subgroup analyses of a phase 3 study. Breast Cancer Res Treat 118:81–87PubMedCrossRef
107.
Zurück zum Zitat Gnant M, Pfeile Gr, Dubsky PC, Hubalek M, Greil R, Jakesz R, Wette V, Balic M, Haslbauer F, Melbinger E, Bjelic-Radisic V, Artner-Matuschek S, Fitzal F, Marth C, Sevelda P, Mlineritsch B, Steger GG, Manfreda D, Exner R, Egle D, Bergh J, Kainberger F, Talbot S, Warner D, Fesl C, Singer CF, On behalf of the Austrian Breast and. Colorectal Cancer Study Group (2015) Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Oncol 386(9992):433–443CrossRef Gnant M, Pfeile Gr, Dubsky PC, Hubalek M, Greil R, Jakesz R, Wette V, Balic M, Haslbauer F, Melbinger E, Bjelic-Radisic V, Artner-Matuschek S, Fitzal F, Marth C, Sevelda P, Mlineritsch B, Steger GG, Manfreda D, Exner R, Egle D, Bergh J, Kainberger F, Talbot S, Warner D, Fesl C, Singer CF, On behalf of the Austrian Breast and. Colorectal Cancer Study Group (2015) Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Oncol 386(9992):433–443CrossRef
108.
Zurück zum Zitat Bone HG, Bolognese MA, Yuen CK, Kendler DL, Miller PD, Yang YC, Grazette L, San Martin J, Gallagher JC (2011) Effects of denosumab treatment and discontinuation on bone mineral density and bone turnover markers in postmenopausal women with low bone mass. J Clin Endocrinol Metab 96(4):972–80.119PubMedCrossRef Bone HG, Bolognese MA, Yuen CK, Kendler DL, Miller PD, Yang YC, Grazette L, San Martin J, Gallagher JC (2011) Effects of denosumab treatment and discontinuation on bone mineral density and bone turnover markers in postmenopausal women with low bone mass. J Clin Endocrinol Metab 96(4):972–80.119PubMedCrossRef
109.
Zurück zum Zitat Miller PD, Wagman RB, Peacock M, Lewiecki EM, Bolognese MA, Weinstein RL, Ding B, San Martin J, McClung MR (2011) Effect of denosumab on bone mineral density and biochemical markers of bone turnover: six-year results of a phase 2 clinical trial. J Clin Endocrinol Metab 96(2):394–402PubMedCrossRef Miller PD, Wagman RB, Peacock M, Lewiecki EM, Bolognese MA, Weinstein RL, Ding B, San Martin J, McClung MR (2011) Effect of denosumab on bone mineral density and biochemical markers of bone turnover: six-year results of a phase 2 clinical trial. J Clin Endocrinol Metab 96(2):394–402PubMedCrossRef
110.
Zurück zum Zitat Hadji P, Coleman RE, Wilson C, Powles TJ, Clézardin P, Aapro M, Costa L, Body JJ, Markopoulos C, Santini D, Diel I, Di Leo A, Cameron D, Dodwell D, Smith I, Gnant M, Gray R, Harbeck N, Thurlimann B, Untch M, Cortes J, Martin M, Albert US, Conte PF, Ejlertsen B, Bergh J, Kaufmann M, Holen I (2016) Adjuvant bisphosphonates in early breast cancer: consensus guidance for clinical practice from a European Panel. Ann Oncol 27(3):379–390PubMedCrossRef Hadji P, Coleman RE, Wilson C, Powles TJ, Clézardin P, Aapro M, Costa L, Body JJ, Markopoulos C, Santini D, Diel I, Di Leo A, Cameron D, Dodwell D, Smith I, Gnant M, Gray R, Harbeck N, Thurlimann B, Untch M, Cortes J, Martin M, Albert US, Conte PF, Ejlertsen B, Bergh J, Kaufmann M, Holen I (2016) Adjuvant bisphosphonates in early breast cancer: consensus guidance for clinical practice from a European Panel. Ann Oncol 27(3):379–390PubMedCrossRef
111.
Zurück zum Zitat Dhesy-Thind S, Fletcher GG, Blanchette PS, Clemons MJ, Dillmon MS, Frank ES, Gandhi S, Gupta R, Mates M, Moy B, Vandenberg T, van Poznack CH (2017) Use of adjuvant bisphosphonates and other bone-modifying agents in breast cancer: a Cancer Care Ontario and American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 35(18):2062–2081PubMedCrossRef Dhesy-Thind S, Fletcher GG, Blanchette PS, Clemons MJ, Dillmon MS, Frank ES, Gandhi S, Gupta R, Mates M, Moy B, Vandenberg T, van Poznack CH (2017) Use of adjuvant bisphosphonates and other bone-modifying agents in breast cancer: a Cancer Care Ontario and American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 35(18):2062–2081PubMedCrossRef
112.
Zurück zum Zitat Bošković L, Gašparić M, Petković M, Gugić D, Lovasić IB, Soldić Ž, Miše BP, Dabelić N, Vazdar L, Vrdoljak E (2017) Bone health and adherence to vitamin D and calcium therapy in early breast cancer patients on endocrine therapy with aromatase inhibitors. The Breast 31:16–19PubMedCrossRef Bošković L, Gašparić M, Petković M, Gugić D, Lovasić IB, Soldić Ž, Miše BP, Dabelić N, Vazdar L, Vrdoljak E (2017) Bone health and adherence to vitamin D and calcium therapy in early breast cancer patients on endocrine therapy with aromatase inhibitors. The Breast 31:16–19PubMedCrossRef
113.
Zurück zum Zitat Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) (2015) Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet 386(10001):1353–1361CrossRef Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) (2015) Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet 386(10001):1353–1361CrossRef
Metadaten
Titel
Cancer Treatment and Bone Health
verfasst von
Catherine Handforth
Stella D’Oronzo
Robert Coleman
Janet Brown
Publikationsdatum
20.01.2018
Verlag
Springer US
Erschienen in
Calcified Tissue International / Ausgabe 2/2018
Print ISSN: 0171-967X
Elektronische ISSN: 1432-0827
DOI
https://doi.org/10.1007/s00223-017-0369-x

Weitere Artikel der Ausgabe 2/2018

Calcified Tissue International 2/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.