Skip to main content
Erschienen in: Clinical and Translational Oncology 11/2022

Open Access 02.07.2022 | Special Article

Bone loss induced by cancer treatments in breast and prostate cancer patients

verfasst von: Santos Castañeda, Ana Casas, Aránzazu González-del-Alba, Guillermo Martínez-Díaz-Guerra, Xavier Nogués, Cristina Ojeda Thies, Óscar Torregrosa Suau, Álvaro Rodríguez-Lescure

Erschienen in: Clinical and Translational Oncology | Ausgabe 11/2022

Abstract

Cancer and cancer therapies are a major factor risk for osteoporosis due to bone loss and deterioration of bone microarchitecture. Both factors contribute to a decrease in bone strength and, consequently, increased bone fragility and risk of fracture. Cancer-associated bone loss is a multifactorial process, and optimal interdisciplinary management of skeletal health, accurate assessment of bone density, and early diagnosis are essential when making decisions aimed at reducing bone loss and fracture risk in patients who have received or are receiving treatment for cancer. In this document, a multidisciplinary group of experts collected the latest evidence on the pathophysiology of osteoporosis and its prevention, diagnosis, and treatment with the support of the Spanish scientific society SEOM. The aim was to provide an up-to-date and in-depth view of osteoporotic risk and its consequences, and to present a series of recommendations aimed at optimizing the management of bone health in the context of cancer.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12094-022-02872-1.
The original online version of this article was revised to correct a sentence in the Abstract section.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s12094-022-02905-9.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Osteoporosis is defined as a systemic skeletal disease characterized by low bone mass and a deterioration in bone microarchitecture, resulting in an increase in bone fragility and predisposition to fractures [1].
Cancer treatments can have significant negative effects on bone health and cause bone loss or secondary osteoporosis that increases the risk of fractures [24]. Moreover, cancer accentuates age-related loss of muscle mass, or sarcopenia, which increases the risk of falls and osteoporotic fractures [5]. This compromises the patient’s functional status, their quality of life, and their very survival.
The aim of this document, drawn up by a group of experts from the Spanish Society of Medical Oncology (SEOM), the Spanish Society for Bone and Mineral Metabolism Research (SEIOMM), the Spanish Society of Rheumatology (SER), and the Spanish Society of Orthopedic Surgery and Traumatology (SECOT) is to provide an up-to-date review of the pathophysiology of the metabolic bone comorbidity osteoporosis in cancer patients. We discuss the biomarkers most widely used in the diagnosis and monitoring of osteoporosis, and the main pharmacological and non-pharmacological measures aimed at preventing and treating bone loss and fractures. We also present advances in surgical and rehabilitation techniques and a series of recommendations based on our clinical experience and expertise to provide a practical up-to-date framework for specialists who routinely monitor these patients.

Pathophysiology of osteoporosis in cancer patients

The pathophysiology of osteoporosis is multifactorial and varies depending on the underlying disorder. There are multiple risk factors involved in bone loss and fracture (called “osteoporotic, fragility, or low-impact fractures”) (Table 1). Cancer itself and many treatments used in oncology (chemotherapy [CT], radiotherapy [RT], glucocorticoids [GC], or hormone therapies [HT]) are independent risk factors for the development of bone loss, osteoporosis, and fractures [24]. There are also other factors that alter bone health, such as prolonged immobilization and/or sedentary lifestyle, primary bone cancer, and bone metastases associated with other types of cancer. Although osteoporosis is a common manifestation in these patients, the pathogenic mechanisms are largely unknown, and most studies focus on patients with breast (BC) and prostate cancer (PC).
Table 1
General factors that increase the risk of osteoporosis and fractures
Non-modifiable risk factors
 Age
Personal history of previous fracture
 Female sex
Genetic (family history)
 Ethnicity (Asian or Caucasian)
Hip fractures in first-degree relatives
Modifiable risk factors
 Low levels of physical activity (prolonged immobilization and/or sedentary lifestyle)
Estrogen deficiency (early menopause, prolonged amenorrhea periods)
 Smoking
Low calcium intake or malnutrition
 Alcohol consumption (≥ 3 units per day)
Osteoporosis secondary to chronic or consumptive diseases
 Low weight (< 58 kg or 127 lb)
Chronic glucocorticoid use
Drugs used in oncology
 Aromatase inhibitors (BC)
Chemotherapy
 Steroidal (exemestane)
Alkylating agents
 Non-steroidal (anastrozole, letrozole)
Anthracyclines Docetaxel
 GnRH agonists (BC: goserelin, triptorelin)
Doxorrubicin
 Selective ER Modulators (BC)
5-fluorouracil
 Androgen deprivation therapy (PC)
Other
 LHRH analogues (goserelin, buserelin, leuprorelin, triptorelin)
Other drugs
 LHRH antagonists (goserelin)
Antidepressants and serotonin reuptake inhibitors
 Antiandrogens (enzalutamide, bicalutamide, flutamide, nilutamide)
Oral antidiabetics (thiazolidinediones)
Other osteopenizing drugs
 Methotrexate
NSAIDs
 Megestrol acetate
Estramustine
 Platinum compounds
Ifosfamide
 Cyclophosphamide
Radiotherapy
 Interferon-alfa
Combination of chemotherapy regimens
 Cyclosporine
Valproic acid
 Vitamin A
 
ADT androgen deprivation therapy, BC breast cancer, ER estrogen receptor, GnRH gonadotropin-releasing hormone, kg kilograms, lb pounds, LHRH luteinizing hormone-releasing hormone, NSAIDs non-steroidal anti-inflammatory drugs, PC prostate cancer
It is important to differentiate between osteoporosis and fractures, because although they share a number of mechanisms and risk factors, others are more specific to each process, such as falls in the case of fractures.

General risk factors for osteoporosis

Figure 1 summarizes the main risk factors, treatments, and pathogenic mechanisms involved in osteoporosis in cancer patients, particularly in patients with BC and PC.

Non-modifiable risk factors

The main non-modifiable risk factors are shown in Table 1. Age is one of the main risk factors for both osteoporosis and fracture. In general, the risk of fracture at any location is greater the older the individual [6]. Sex and ethnicity are also important risk factors [6, 7]. Genetics is another fundamental determinant that increases the risk of osteoporosis. In fact, 60%–70% of an individual’s bone mass is estimated to be genetically determined. A history of hip fracture in a first-degree relative doubles the risk of hip fracture in women (relative risk [RR]: 2 [95% confidence interval, CI 1.4–2.9]), regardless of their bone mineral density (BMD) [8]. A history of previous fragility or low-impact fractures is another important determinant of new fractures, with a previous vertebral fracture increasing the risk of hip fracture (RR: 2.5 [95%CI 1.9–3.2]), new vertebral fracture (RR: 1.7 [95%CI 1.4–2]), and proximal humerus fracture (RR: 1.9 [95%CI 1.5–2.4]) [912].

Modifiable risk factors

The presence of associated comorbidities and treatment with GC or other osteopenizing drugs are particularly relevant in the case of cancer patients, who are usually polymedicated (Table 1). The higher the cumulative dose of GC, the higher the risk of osteoporosis. In general, maintaining doses of ≥ 7.5 mg/day, or even ≥ 5 mg/day for more than 3 months, is a risk factor.
Fragility fractures are usually triggered by low-energy trauma that would not ordinarily cause a fracture, such as a fall from standing height or less, and are a sign of underlying osteoporosis or low bone quality. In osteoporosis, the risk of fragility fractures is also related to the individual’s BMD and likelihood of falling, since the fall itself is a fundamental precipitating factor, especially of major non-vertebral fractures (radius, femur, and humerus). In general, the lower the BMD and the higher the number of falls, the greater the risk of a fragility fracture [6, 7, 13, 14].

Specific risk factors for osteoporosis

Breast cancer

Osteoporosis associated with BC is mainly linked to estrogen deprivation induced by chemotherapy (CT) and hormone therapy (HT), and more specifically, to the use of non-steroidal aromatase inhibitors (AI) [15, 16]. Estrogens play an essential role in bone homeostasis by binding to estrogen receptors alpha and beta (ERα and ERβ, respectively), which are expressed in both osteoblasts (OB) and osteoclasts (OC), thereby decreasing bone resorption and bone loss [17]. Estrogens promote the proliferation and activity of OBs, decrease the apoptosis of osteocytes (involved in bone formation), and reduce the differentiation and maturation of osteoclastic precursors by increasing the production of osteoprotegerin (OPG) and decreasing the synthesis of osteoclast differentiation and proliferation factor (receptor activator of nuclear factor-κB ligand [RANKL]) [18, 19] (Fig. 1).
Bone loss caused by CT (especially CT involving alkylating agents and/or 5-fluorouracil) or after HT (especially HT using non-steroidal AIs) is rapid, and can reach 6–8% during the first year, especially in trabecular bone. CT-induced ovarian failure has more immediate and difficult-to-reverse effects, while hormone-induced failure can be reversed months after discontinuation, especially in young women. The risk level for osteoporosis in decreasing order is as follows: premenopausal women with CT-induced menopause treated with gonadotropin-releasing hormone (GnRH) agonists; women initially treated with tamoxifen and subsequently treated with AI; and finally, women treated only with AI, particularly those aged < 70 years [14, 20, 21].
Activins and inhibins are other mediators of interest in osteoporosis in cancer patients. Both belong to the transforming growth factor β (TGFβ) superfamily, whose functions are only partially known [18]. Activins are homodimeric peptides secreted by breast tumor cells that inhibit the proliferation of estrogen receptor-positive (ER +) cells. In addition, they stimulate osteoclastogenesis via receptor activator of nuclear factor-κB (RANK), although their effect on bone formation is less well known. Inhibins are heterodimeric proteins also secreted by breast cells that mediate opposite effects in carcinogenesis. At bone level, they cause a bone turnover disorder by inhibiting both osteoblastogenesis and osteoclastogenesis (Fig. 1) [18, 22].

Prostate cancer

As with estrogen in women, androgens are fundamental in maintaining bone homeostasis in men. Androgens have a double/triple effect on bone tissue. In fact, they increase bone formation and decrease resorption through a direct effect mediated by androgen receptors (AR). Moreover, some androgens are transformed into estrogens at the peripheral level and act through ERα, which is an additional benefit. Thus, low androgen levels are associated with elevated RANKL levels and greater bone resorption [18, 23]. Osteoporosis is present at diagnosis in 25–40% of patients with prostate cancer (PC). Treatment based on surgical or pharmacological hormone deprivation (androgen deprivation therapy [ADT], which includes the use of luteinizing hormone-releasing hormone [LHRH] analogues), reduces testosterone levels to 20% below baseline after 2–4 weeks [18]. This results in rapid losses in BMD (already detectable 6–9 months after the start of treatment), ranging from 5% to 10% in the first year, especially in the radius and spine, which increases the risk of fractures [18, 24, 25]. Fragility fractures appear in up to 20% of patients in the first 5 years of ADT and the risk increases with time and number of doses administered [25, 26]. ADT can increase the risk of osteoporosis from 10%–40% to 80% after 10 years of treatment exposure [18, 27], and 35% of patients experience skeletal fractures. Other factors that enhance this effect are age and low body mass.
Androgens also have a positive effect at muscle level, while the use of ADT causes an increase in total body fat at the expense of a decrease in lean mass [28, 29]. Therefore, ADT produces sarcopenia with rapid loss of muscle mass and increased risk of falls [28, 30].
Other factors that increase the risk of osteoporosis in PC patients are CT (e.g. docetaxel), RT, prolonged use of GC, and interventions such as orchiectomy. In patients with PC treated with 10–12 mg/day prednisone or equivalent for more than 3 months, a 7-to-17-fold increase in the risk of vertebral or hip fracture has been shown [31].
PC may induce osteoporosis independently of hormone treatment, due to an increase in the expression of TGFβ and parathyroid hormone-related protein (PTHrP) [32]. PTHrP increases the growth and survival of prostate tumor cells in vitro (Fig. 1) [33].
Another tumor that can cause osteoporosis through the aforementioned mechanisms is testicular cancer. In this case, the appearance of osteoporosis is related to age and time from orchiectomy.

Other tumors

Hematology patients undergoing bone marrow transplantation merit special mention. In many cases, these are young women who have received multiple cycles of CT, immunosuppressant and GC before and after transplantation, with frequent gonadal failure, often permanent [3436]. In patients undergoing allogeneic and autologous transplantation, graft-versus-host-disease and its treatment appear to play an important role in osteoporosis [37]. In general, these patients are at increased risk for osteoporosis, and treatment options often include hormone replacement therapy (HRT) with estrogens.

Diagnosis and monitoring of osteoporosis in cancer patients

Role of bone biomarkers in treatment monitoring and evaluation

It is essential to measure bone mass to estimate fracture risk. The main techniques available for quantifying bone mass include dual-energy X-ray absorptiometry (DXA), quantitative computed tomography, and measurements of bone microarchitecture such as the trabecular bone score (TBS). According to the World Health Organization (WHO), DXA is the gold standard for the study of osteoporosis [38].
Bone turnover markers (BTM) are proteins or enzymes secreted by OBs or OCs during the formation or degradation of matrix protein collagen. They are released into the bloodstream, so their detection is useful for the diagnosis and monitoring of osteoporosis, and for the individual assessment of fracture risk [39, 40]. There are two types of BTMs as follows:
  • Bone formation markers (BFMs), which derive from formation processes and reflect osteoblastic activity: alkaline phosphatase (ALP), bone-specific alkaline phosphatase (BALP), procollagen type I carboxy-(PICP) and amino-terminal propeptides (PINP), and osteocalcin.
  • Bone resorption markers (BRMs), which derive from the resorption processes and reflect osteoclastic activity: cross-linked carboxy- and amino-terminal telopeptides of type I collagen (CTX and NTX), tartrate-resistant acid phosphatase (TRAP), pyridinolines and deoxypyridinolines, hydroxyproline and sialoprotein.
BRMs show variations within the first 3 months of treatment, so they can be used to assess the response to a given antiresorptive therapy. A positive treatment outcome is reflected in a reduction in these markers. At the end of treatment (approximately 3–6 months later), the markers return to their baseline levels. The best BRMs for treatment monitoring are NTX and CTX in urine, and CTX in blood. Response to antiresorptive drugs has been defined as a reduction in BTM levels of more than the least significant change or to a value below the mean levels of the premenopausal reference range [41]. A 25–30% decrease in CTX or PINP has been associated with a reduction in vertebral fracture (nearly a 40%) [42], so this may be a good threshold for use in clinical practice to evaluate the response of patients to antiresorptive therapies. The most widely used BFMs in clinical practice are BALP, osteocalcin and PINP [43].
There are certain considerations with respect to BRMs as follows:
  • BRMs measure functional activity and do not quantify bone mass, so they should not be used for the diagnosis of osteoporosis.
  • Collagen-dependent BRMs may be altered due to non-bone pathologies, such as chronic liver disease of any origin.
  • BALP may be altered depending on the liver ALP values.
  • Chronic renal failure may alter the concentration of BRMs.
  • Bone fractures may alter BRM values for several months.
  • In general, it is preferable to measure BRMs in serum instead of in urine, due to their lower variability.

Diagnostic and monitoring tests: recommendations and frequency

Assessing the risk of low BMD is important in cancer patients, since they often present premature loss of bone mass that contributes to the risk of osteoporosis, even in the absence of menopause. Recommendations for patient assessment are summarized in Table 2.
Table 2
Recommendations for patient diagnostic and monitoring screening
Patient assessment
 
Comments
Medical history
Fracture history
Previous fractures increase the risk of future fractures, regardless of BMD. It is useful to perform a spinal x-ray before starting treatment in order to detect previous asymptomatic fractures. Techniques such as CT, MRI and/or PET can be very useful in determining whether an acute fracture is a bone metastasis
 
Classic risk factors
Family history of osteoporosis should be included. The FRAX® is an easily reproducible diagnostic tool developed by the University of Sheffield from a meta-analysis of a wide variety of risk factors for osteoporotic fractures (https://​www.​sheffield.​ac.​uk/​FRAX/​). It allows the estimation of the 10-year risk of hip fracture and major osteoporotic fracture, with or without concomitant determination of BMD, although it may underestimate the risk in cancer patients. When using the FRAX® tool in cancer patients, cancer can be considered a “secondary osteoporosis”. One limitation is that this tool does not weigh the number, severity, or location of previous fractures, or the total or cumulative GC treatment
 
Medications
Treatment review for potentially osteopenizing drugs
 
Fall risk estimation
Estimation of fall risk
 
Vitamin D
Vitamin D deficiency is an independent risk factor for low bone mass, falls, and fractures [112]. Determination of 25-hydroxyvitamin D levels allows patients to be classified as normal (> 30 ng/ml), insufficient (20–30 ng/ml) or deficient (< 20 ng/ml)
Physical & complementary examinations
Height
Height should be measured at least once a year and whenever there is suspicion of a new vertebral compression fracture
 
BRMs
Variations throughout the day explain why their reproducibility is not a critical factor in the assessment of FR in cancer patients. However, it may be useful to determine BRMs at the beginning of diagnosis or once treatment has started to gain insight into the status of bone metabolism and, above all, to monitor treatment
 
BMD
DXA is recommended to measure and compare BMD with previous DXA to assess the progression of osteoporosis. The WHO recommends performing these measurements every 2 years from menopause. The standardized recommendation for menopausal women treated with AI was an annual BMD assessment for the duration of treatment, especially if there is baseline osteopenia or osteoporosis [113]. The ASCO recommends increasing the frequency of DXA follow-up screening if deemed medically necessary based on the results of BMD testing and expected bone loss [84] Fig. 2
AI aromatase inhibitors, ASCO American Society of Clinical Oncology, BMD bone mineral density, BRMs bone resorption markers, CT computed tomography, DXA dual energy X-ray absorptiometry, FRAX Fracture Risk Assessment Tool, GC glucocorticoid; FR fracture risk, MRI magnetic resonance imaging, PET positron emission tomography, PMW postmenopausal women, PrMW premenopausal women, WHO World Health Organization

Prevention and treatment of osteoporosis in cancer patients

This section will discuss in detail the main pharmacological and non-pharmacological measures to prevent and treat osteoporosis (Table 3).
Table 3
Pharmacological and non-pharmacological measures for the prevention and treatment of osteoporosis in patients with cancer
Non-pharmacological measures
Pharmacological measures
Smoking cessation
Hormone replacement therapies
Avoid excess alcohol intake
Antiresorptive agents
Avoid excess caffeine intake
Selective estrogen receptor modulators (SERMs)
Avoid sedentary lifestyle
Calcitonin
Prevent falls
Bisphosphonates (alendronate, risedronate, ibandronate, zoledronate)
Balanced diet
Denosumab (anti-RANKL biologic)
Adequate intake of:
 
Trace minerals
 
Proteins
 
Vitamin D
 
Calcium
Combination of calcium and vitamin D
 
Physical therapy (improve muscle strength and balance)
 
RANKL receptor activator of NF-κB ligand, SERMs selective estrogen receptor modulators

Non-pharmacological measures

Dietary supplements: amino acids, calcium and vitamin D

Age and cancer treatments can impair protein synthesis with decreased response to amino acids and insulin resistance. This is often compounded by the physical or psychological difficulty of achieving adequate intake, a result of the disease itself or the therapeutic sequelae. Adequate protein, calcium, and vitamin D levels are essential for proper bone homeostasis, so it is recommended that they be added to the diet, preferably as dietary supplements because of their greater efficacy and tolerance [44, 45]. Vitamin D plays a fundamental role in calcium and phosphorus homeostasis and is essential to maintain skeletal muscle health, muscle mass and strength, and balance [46]. Vitamin D levels higher than 50 nmol/L (20 ng/mL) are recommended in the general population. Levels above 100 nmol/L (40 ng/mL) have been shown to reduce AI-associated arthralgias [47, 48]. As regards calcium, the necessary dietary intake is estimated to be 1200–1500 mg/day.
All patients starting treatment for osteoporosis should have normal calcium and vitamin D levels at the start of treatment. The National Comprehensive Cancer Network (NCCN) guidelines recommend calcium (1200 mg daily) and vitamin D (800–1000 IU daily) supplements for young women at risk of losing BMD and for women over 50 years of age. For men with PC, calcium (1000 mg daily) and vitamin D (800–1000 IU daily) supplements are recommended from the age of 50 [49, 50]. The European Association of Urology (EAU) mentions the protective role of calcium supplements in patients with PC, of whom 71% receive ADT [51]. Several studies and reviews have confirmed the role of dietary supplements in alleviating the adverse events of ADT on BMD [52]. Monitoring by bone densitometry every 1–2 years (or lengthening the scan interval in the case of stabilization) is also recommended in these patients. BRMs such as serum CTX or urine NTX, or BFMs such as serum PINP every 3–6 months from the start of treatment may be considered.
Obesity and vitamin D deficiency are global health issues. Although evidence from meta-analyses has consistently supported an association between body weight and fat mass and low levels of vitamin D, the underlying pathophysiological mechanisms are complex and many potential confounding factors should be taken into account. Patients with obesity frequently present decreased levels of vitamin D in serum, as this parameter correlates inversely with BMI, body weight or abdominal fat, irrespective of gender, likely leading to an increased risk of osteoporosis [53]. The potential benefit of vitamin D supplementation in obese people has been reported in several studies [54, 55], so the assessment of vitamin D levels in patients with osteoporosis and high BMI is advisable in order to prescribe vitamin D supplementation when necessary.

Physical exercise

Exercise has been shown to improve a wide spectrum of cancer-related adverse events. Several clinical practice guidelines currently recommend exercise as a key element in the management of cancer patients. The SEOM recently published a position statement on physical exercise and cancer due to its possible impact on the prevention and reduction of complications and relapses [56]. However, there are no specific guidelines for a particular type of exercise, and little is known about the extent to which physical exercise can prevent bone loss.
The results of different randomized trials on the effect of exercise on BMD in cancer survivors have not shown a significant overall improvement in lumbar spine or femoral neck BMD (exercise versus placebo) [57, 58].
In women with BC on treatment, the results of programs combining moderate intensity strength and resistance exercises are inconclusive, largely due to the limitations and heterogeneity of the patient sample and poor adherence to exercise programs [59]. Although not all types of exercise are equally osteogenic, moderate-intensity aerobic exercise has beneficial effects on the lumbar spine BMD in women with BC, and it is also important to encourage adherence to training programs [57, 6062].
There is still no evidence on the effect of exercise in preventing fractures [63] in men with PC undergoing ADT, although positive effects have been observed on other aspects such as anxiety, bone loss and sexual dysfunction [64].

Fatty acids

Omega-3 fatty acids have anti-inflammatory potential and are related to PC. It has been suggested that daily use of omega-3 and omega-6 combined with calcium has a positive effect on bone health [65]. However, the results of the various studies analyzed were inconclusive, so larger-scale research is needed to determine the role and effect of these nutrients in the prevention of events, especially fractures.

Pharmacological measures

Table 3 lists the most used drugs, while the most relevant efficacy and safety clinical trials in patients with BC and PC are presented in Online Resource 1.

Bisphosphonates: breast cancer

Several randomized clinical trials have shown that bisphosphonates prevent or reduce bone mineral loss in women with BC treated with AI, although they do not significantly reduce the overall incidence of fractures. A systematic review of six studies confirmed with moderate-quality evidence that the RR of skeletal events was not significantly reduced in patients treated with bisphosphonates compared to the placebo or the no bisphosphonate group [66].
The studies with the largest number of patients are the Z-FAST and the ZO-FAST trials (Zometa-Femara Adjuvant Synergy Trials). Both evaluated the efficacy of intravenous zoledronic acid (ZOL) in preventing AI-induced bone loss. All patients received calcium (500 mg) and vitamin D (400–800 IU) supplements. In the Z-FAST study, 602 postmenopausal women with early BC who received adjuvant letrozole were randomized to receive upfront or delayed-start intravenous ZOL treatment. At 12 months, BMD was higher in the group that received ZOL up front compared to the delayed-start group, and a significant reduction in NTX and BALP was observed in the first group [67] (Online Resource 1). In the 5-year extension study, a progressive increase in BMD was observed in the upfront treatment group, with significant differences between both groups and no significant differences in the incidence of fractures [68]. One quarter of patients (25%) in the delayed treatment group required ZOL treatment. This suggests that not all women need antiresorptive treatment, and patients should preferably be pre-selected individually on the basis of their fracture risk identified from the BMD and clinical risk factors. In the similarly designed ZO-FAST study, the lumbar spine BMD increased at 36 months in the upfront treatment group, while loss of BMD was observed in the delayed-start group. Twenty-one percent of patients in the delayed group required ZOL treatment during the study [69] (Online Resource 1). Very similar results were obtained in the N03CC trial [70, 71]. It should be noted that the regimen commonly used in non-BC-related menopausal osteoporosis is 5 mg intravenous ZOL annually, which is likely to also be effective in the treatment of AI-induced osteoporosis, although this has not been studied.
With regard to oral bisphosphonates, various trials have assessed their effect on the prevention of AI-induced osteoporosis. The doses studied are the same as those recommended in postmenopausal osteoporosis. Of these, the most extensively studied is risedronate (35 mg/week). In the SABRE trial, 154 postmenopausal patients with BC treated with the AI anastrozole as adjuvant therapy and who had moderate risk of osteoporotic fracture were randomized to receive risedronate or placebo for 2 years. In the risedronate group, the lumbar spine and total hip BMD increased significantly, but decreased in the placebo group [72]. In a more recent placebo-controlled trial that recruited 109 women with low BMD treated with different AIs (anastrozole, letrozole, or exemestane), risedronate achieved greater increases in spine and hip BMD at 24 months compared with placebo [73]. All women received supplemental calcium and vitamin D. Greater reduction in CTX and PINP correlated with a better response in spinal BMD. Risedronate also achieved better conservation of lumbar bone microarchitecture estimated using the TBS [74] (Online Resource 1).
A recent large observational cohort study to evaluate the efficacy of oral bisphosphonates under clinical practice conditions in 36 472 women diagnosed with BC and treated with tamoxifen and/or AI showed that in the subgroup of women treated with AI who had high fracture risk, treatment with oral bisphosphonates reduced the risk of fractures by 30% compared to the group that did not receive bisphosphonates. This is the first real-world study to confirm positive data from previous clinical trials with bisphosphonates, and to report a positive effect on fractures [47].

Bisphosphonates: prostate cancer

A recent systematic review and meta-analysis of 14 clinical trials has shown the protective effects of bisphosphonates on BMD loss in men with non-metastatic PC receiving ADT, resulting in a significant increase in lumbar, femoral neck and total hip BMD after 12 months of treatment [75] (Table 3). Most of the trials analyzed included administration of intravenous ZOL, alendronate, and to a lesser extent, risedronate. Despite this, it has not been possible to demonstrate a reduction in the fracture risk with oral or intravenous bisphosphonates in these patients, since the number of fractures recorded in the trials was extremely low [76]. For this reason, ASCO clinical guidelines consider oral or intravenous bisphosphonates to be a reasonable option to reduce the fracture risk in patients with non-metastatic PC and ADT [77].

Adverse events of bisphosphonates

Bisphosphonates are generally well tolerated. In prolonged treatment with intravenous ZOL, the appearance of flu-like symptoms, renal failure, hypocalcemia and osteonecrosis of the jaw (ONJ) should be monitored, among others. ONJ is a very rare problem that is more common in cancer patients with bone metastases who receive monthly doses of intravenous ZOL for long periods. In early BC, the incidence of ONJ is 0.25%. The Z-FAST trial reported two unconfirmed cases of ONJ, and generalized bone pain was most commonly seen in the group randomized to upfront intravenous ZOL versus the delayed-start group [62].

Denosumab

Denosumab is a human monoclonal antibody directed against RANKL, thus inhibiting the differentiation, proliferation and activity of OCs and, therefore, reducing bone resorption. In clinical trials in postmenopausal women with osteoporosis, denosumab treatment for up to 10 years increased lumbar spine and femoral neck BMD, and reduced the risk of osteoporotic vertebral, hip and non-vertebral fractures [78, 79]. The most common adverse events associated with denosumab are hypocalcemia, diarrhea, eczema and skin infections; the occurrence of hypophosphatemia and ONJ, among other effects, has also been described, albeit relatively less frequently [80].

Denosumab: breast cancer

In a placebo-controlled clinical trial in women with non-metastatic BC treated with AI and low BMD, the use of denosumab (according to the recommended regimen for postmenopausal osteoporosis) led to a significant increase in lumbar spine, hip and femoral neck BMD and a reduction in BTMs. This effect was independent of the duration of AI therapy. There was no significant effect on the number of fractures [81]. The ABCSG-18 (Adjuvant Denosumab in Breast Cancer Trial) study, which evaluated the protective effect of denosumab against fractures in women with non-metastatic BC on AI, found a 50% reduction in the risk of clinical fracture in women treated with denosumab compared to placebo. In addition, a decrease in the risk of new vertebral fractures and worsening of existing fractures, and a significant increase in lumbar spine, total hip and femoral neck BMD were confirmed. The benefits of denosumab were independent of baseline BMD and age [82] (Table 3).

Denosumab: prostate cancer

In a trial in patients with PC (> 70 years, or low bone mass with T-score < -1) undergoing surgical castration or ADT with GnRH agonists, a reduction in the risk of new vertebral fractures was observed after 12 months of denosumab treatment (compared to placebo). In addition, a significant and progressive increase in BMD at the lumbar spine, total hip, femoral neck and distal third of the radius was confirmed from the first month of treatment. The incidence of adverse events was similar in both groups, with no cases of ONJ or atypical fractures reported. Cataracts were more frequent in the denosumab group, while the occurrence of bone metastases was more frequent in the placebo group [83] (Online Resource 1).
Different clinical guidelines recommend denosumab as the drug of choice for the prevention of bone loss in patients with non-metastatic PC on ADT [84].

Other treatments

Bone-forming agents

Osteoanabolic agents stimulate the differentiation, function and survival of OBs. These include teriparatide (recombinant form of parathyroid hormone [PTH]), abaloparatide (PTHrP analogue), and romosozumab. After a certain period of time, the conventional treatment for postmenopausal osteoporosis applied in the general population should be administered according to the usual risk factors.
Chronic exposure to PTH or PTHrP analogues causes bone resorption, although intermittent administration has been shown to stimulate bone formation more than resorption in postmenopausal women [85, 86]. Their use in cancer patients on ADT is usually restricted or contraindicated. However, they can be used in patients with osteoporosis with high fracture risk and in cases of ONJ or atypical femoral fracture, both antiresorptive-related complications, as they can facilitate their rapid resolution [87]. Bone-forming treatments are contraindicated in patients with primary or secondary hyperparathyroidism, hypercalcemia, or patients at increased risk of osteosarcoma (such as patients with Paget’s disease of bone and patients who have received RT). A single course of treatment with these drugs is generally recommended for up to 2 years. Although they are usually well tolerated and no associated complications have been described, they can cause hypercalcemia and hypercalciuria to a marginal extent.
Romosozumab (anti-sclerostin monoclonal antibody) is a new anabolic agent approved by the Food and Drug Administration (FDA) in 2019 and the European Medicines Agency (EMA) in 2020, after having demonstrated a reduction in vertebral and non-vertebral fractures compared to placebo and alendronate. Romosozumab is approved for the treatment of severe osteoporosis in postmenopausal women with high fracture risk [88, 89]. There is no formal contraindication for the romosozumab use in cancer patients, despite one of the criteria for patient exclusion in the pivotal clinical trial was a previous history of cancer [90].

Selective estrogen receptor modulators and calcitonin

Selective estrogen receptor modulators (SERMs) are a group of drugs that have either proestrogenic (mainly cardiovascular, liver, and bone) or antiestrogenic activities (breast, uterus) depending on the target tissue on which they act.
Raloxifene has estrogenic activity in the bone and no proestrogenic activity in the endometrium, unlike other SERMs. It has, therefore, been approved for the prevention and treatment of osteoporosis in postmenopausal women. Tamoxifen, in contrast, has agonist activity at the endometrial level, and has been associated with an increased risk of endometrial cancer. Although not used as a treatment for osteoporosis, tamoxifen contributes to the improvement of bone health in postmenopausal patients who receive it as treatment or prophylaxis against BC [91].
All these drugs carry a slight increase in the risk of thromboembolic events due to their estrogenic agonist activity at the cardiovascular level, as well as climacteric symptoms.
New agents have been added to classic SERMs, such as bazedoxifene, a third-generation SERM that has been approved for the treatment of postmenopausal osteoporosis with increased risk of fractures. The use of these drugs is associated with a lower risk of BC, and they are approved for chemoprevention treatment in patients at high risk of BC.
Calcitonin is a hormone that acts on OCs and inhibits bone resorption. In Spain, it is marketed as salmon calcitonin or eel calcitonin [92]. The former is the most frequently used and has a high affinity for the calcitonin receptor (up to 40 times higher than human calcitonin). Intranasal, oral and parenteral formulations have been developed [93, 94]. Calcitonin has shown a benefit in increasing bone mass in the axial skeleton and in reducing the risk of fracture, although to a lesser extent compared with other agents such as bisphosphonates. Analysis of data from different studies evaluating the safety of prolonged use of calcitonin in the treatment of osteoporosis identified an increase in the incidence of cancer in patients receiving the drug compared to the placebo group (with incidence rates of 0.7–2.4%). Accordingly, EMA issued a statement explaining that the benefits of calcitonin as a treatment for osteoporosis did not outweigh the risk identified in the safety analyses and recommended limiting its indication to acute periods of the disease. The main adverse events associated with administration of this treatment are nausea, vomiting, and hot flushes. Calcitonin is not used as a first-line therapy due to the existence of other drugs that are more effective in preventing bone loss and reducing fracture risk. It is mainly indicated in patients with recent osteoporotic fracture, but should only be administered for 2–5 weeks at the lowest effective dose for the patient, or until resolution of pain. After the acute episode, it is recommended to switch calcitonin to other more effective osteoporosis medications.

Sequential treatment

Oral bisphosphonates are recommended as first-line treatment of osteoporosis due to their efficacy, safety, accessibility and cost-effectiveness. In patients contraindicated for oral or even intravenous bisphosphonates (as is the case in patients with gastrointestinal disturbances or renal failure) or patients with high fracture risk who have new fracture events despite bisphosphonate treatment (which should be switched to an anabolic agent), other alternatives may be offered based on the fracture risk, efficacy, safety and patient preference. One of these alternatives is denosumab.
The denosumab discontinuation effect has been the subject of recent attention, given the risk of fracture following discontinuation due to a rebound effect on bone resorption observed in clinical series, although this has not been confirmed with high-level evidence. In this respect, a recent systematic review by the European Calcified Tissue Society (ECTS) working group suggests that patients with high fracture risk can maintain denosumab beyond 5 years and even continue treatment for up to 10 years [95]. In case of discontinuation, close patient follow-up or switching to bisphosphonates is recommended, although there is no high-level evidence to date to support the regimen to be followed and its duration.
Finally, romosozumab, a more potent bone-forming agent than PTH, has been shown to be more effective than teriparatide after prolonged treatment with oral bisphosphonates in postmenopausal women, according to the STRUCTURE study [88], although there is still no experience with this drug in cancer patients.

Osteoporotic fractures in cancer patients: prevention and treatment

The burden of osteoporotic fractures has been growing despite the development of bone-protecting medication, mainly due to population aging. Furthermore, as long-term survival of oncologic disease increases, more patients with osteoporotic fractures will be likely to have a history of cancer, particularly breast cancer [96].
Surgical management of fractures follows a series of principles published by the AO Society (Arbeitsgemeinschaft für Ostheosynthesefragen) [97]: (1) restoration of anatomical relationships; (2) fixation providing stability as required by the type of fracture, patient and injury; (3) preservation of blood supply to tissues and bones; and (4) early and safe mobilization of the injured part and the patient as a whole. Several technological innovations in recent decades have helped stabilize osteoporotic fractures while fulfilling these principles, and can be summarized as angular-stable implants, augmentation, and minimally invasive techniques that allow optimal application of biomechanical principles to protect the entire bone [98].

Surgical management

Implants and devices

Angular-stable implants have an additional fixation point between the screw and the implant itself to increase resistance to shearing in fragile or comminuted bone. This added point of fixation is achieved in plates by using locking screw heads that lock into the plate itself through threaded screw heads or locknuts (Fig. 3); in nails, the holes for the locking bolts are threaded or lined with polyethylene or a similar material to lock the screws in place. Locking plates do not rely on bone-to-plate contact for stability, acting as “internal fixators” without excessive periosteal stripping or soft tissue dissection. This allows for indirect reduction and plating using minimally invasive surgery and plate constructs with long working distances that are less stiff and distribute loads across the bone. The trend is towards using longer implants that protect the entire bone (i.e., long cephalomedullary nails), especially in cases with a possibility of metastatic disease [99, 100].
A recent survey of French physicians showed a willingness to use implantable devices to prevent contralateral hip fractures, particularly in oncologic indications [101]. Several techniques have been developed, but experience is limited to small case series and preliminary trials [102], and the cost-effectiveness of these interventions has not been evaluated yet [103].

Augmentation techniques

Augmentation is the injection of bone cement or bone substitutes in the area of the screws to increase purchase in poor bone and assist load transfer through fenestrations in specially designed cannulated screws (Fig. 2A and B). This technique is also used in vertebral kyphoplasty [104], where bone cement is injected into a cavity in the vertebral body created by balloon expansion (Fig. 2C) to reduce micromotion at the fracture site and increase trabecular bone resistance to compression. Osteoplasty is the application of this technique to bones other than spine and has been used percutaneously in combination with internal fixation for traumatic injuries [105] as well as lytic bone lesions [106].

Rehabilitation

The fracture fixation construct should be sufficiently stable to enable early mobilization and weight bearing. There is evidence that frail elderly patients are unable to comply with partial weight-bearing [107]; furthermore, early weight-bearing reduces morbidity and mortality [108, 109]. Early intervention has been shown to improve physical function following fracture, particularly hip fractures, though it remains unclear which types of exercise are superior [110].
The effect of exercise interventions seems less marked in patients who have already experienced fractures, although benefits were observed for measures of balance and mobility, fall risk, physical activity, mood, and community outings [111].

Conclusions

Steady improvements in the effectiveness of cancer treatments have not been accompanied by equally optimal management of skeletal health, which is badly affected by the disease and the treatments themselves. Efforts are needed to raise awareness among physicians and specialists in the care of cancer patients about the importance of monitoring bone health. Many sectors of the healthcare system still remain oblivious to the tremendous impact on quality of life and functional status caused by bone loss from mild fractures and vertebral compression. The healthcare system also plays an essential role in improving understanding among patients of the available treatments, the risks of fracture and bone weakness associated with some therapies, and the diet and lifestyle changes that are most effective in preventing bone loss and fractures. There is still limited knowledge about the risk of osteoporosis associated with a wide range of medical and surgical treatments for cancer. More research is needed to increase the effectiveness and number of available antiresorptive and bone-forming therapies, and more evidence on the effectiveness of combined bone resorption and formation treatments is needed. Other areas, such as the management of severe osteoporosis, early identification of patients with increased or imminent risk of fracture after recent fractures and patient adherence to long-term treatments, also need to be improved. The emergence of new bone-forming therapies and the application of more personalized precision medicine may represent an important advance in the management of bone health in patients with cancer.
Until these issues have been resolved, efforts should be focused on promoting the identification of cancer patients at risk of morbidity due to bone loss and their proper follow-up. In this context, BMD is currently one of the most important tools in the diagnosis and monitoring of these patients.

Acknowledgements

The authors would like to thank the collaboration of FAES FARMA, S.A. and the scientific support and medical writing services provided by Susana Cañón-Sánchez from Medical Statistics Consulting S.L. (Valencia). We also would like to acknowledge all the scientific societies involved in this work, especially the support received from the Spanish Society of Medical Oncology (SEOM).

Declarations

Conflict of interest

Santos Castañeda has received personal fees from Amgen, Gebro Pharma, Gedeon-Richter Spain, Grünenthal, Lilly, MSD, Pfizer, Roche, Sanofi, Stada, SOBI and UCB, and he is assistant professor of the UAM-Roche EPID-Future chair, Universidad Autónoma de Madrid (UAM), Madrid, Spain. Ana Casas has received consultant or speaker’s fees from Novartis, Roche, Astra Zeneca and Pierre Fabre. Aránzazu González-del-Alba has received research funding from Astellas, travel grants from Astellas, Jansen, Sanofi, BMS, Roche, Pfizer, MSD and Ipsen and honoraria for speaking engagements, membership of advisory boards and continuing medical education from Janssen, Astellas, Sanofi, Bayer, Roche, Ipsen, BMS, MSD, Pfizer, Eusa Pharma, Eisai and Astra Zeneca. Guillermo Martínez-Díaz-Guerra has received research funding from Lilly and Amgen; travel grants from Lilly, UCB, and Amgen; speaker’s fees from Lilly, Amgen, Kyowa Kirin, Italfármaco, and Gebro Pharma; and honoraria for membership of advisory boards and continuing medical education from Lilly, UCB, Amgen, Kyowa Kirin, Takeda, and Alexion. Xavier Nogués has received consultant or speaker’s fees from Amgen, Lilly, Faes Farma and UCB Pharma. Cristina Ojeda Thies has recieved honoraria for presentations and continuing medical education from AMGEN, UCB Pharma, Grünenthal and MBA Surgical Empowerment. Óscar Torregrosa Suau declares no conflicts of interest. Álvaro Rodríguez-Lescure has received consultant or advisor fees from Roche, Pfizer, Novartis, Lilly, MSD, AstraZeneca, and Daiichi-Sankyo, and research funding for his institution from Roche, Novartis, Pfizer, Lilly, AstraZeneca, Amgen, MSD, BMS, Zymeworks, and GEICAM; he has also received speaker’s fees from Roche, Pfizer, Novartis, Lilly, AstraZeneca, MSD, Merck, Pierre-Fabré, and Ipsen.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Peck WA. Consensus development conference : diagnosis, prophylaxis, and treatment of osteoporosis. Am J Med. 1993;94(6):646–50. Peck WA. Consensus development conference : diagnosis, prophylaxis, and treatment of osteoporosis. Am J Med. 1993;94(6):646–50.
2.
Zurück zum Zitat D’Oronzo S, Stucci S, Tucci M, Silvestris F. Cancer treatment-induced bone loss (CTIBL): pathogenesis and clinical implications. Cancer Treat Rev. 2015;41(9):798–808.PubMed D’Oronzo S, Stucci S, Tucci M, Silvestris F. Cancer treatment-induced bone loss (CTIBL): pathogenesis and clinical implications. Cancer Treat Rev. 2015;41(9):798–808.PubMed
3.
Zurück zum Zitat Carmona-Bayonas A, Jiménez-Fonseca P, Castañón E, Ramchandani-Vaswani A, Sánchez-Bayona R, Custodio A, Calvo-Temprano D, Virizuela JA. Chronic opioid therapy in long-term cancer survivors. Clin Transl Oncol. 2017;19(2):236–50.PubMed Carmona-Bayonas A, Jiménez-Fonseca P, Castañón E, Ramchandani-Vaswani A, Sánchez-Bayona R, Custodio A, Calvo-Temprano D, Virizuela JA. Chronic opioid therapy in long-term cancer survivors. Clin Transl Oncol. 2017;19(2):236–50.PubMed
4.
Zurück zum Zitat Imanishi Y. Secondary osteoporosis Cancer treatment-induced bone loss. Clin Calcium. 2018;28(12):1665–70.PubMed Imanishi Y. Secondary osteoporosis Cancer treatment-induced bone loss. Clin Calcium. 2018;28(12):1665–70.PubMed
5.
Zurück zum Zitat Winters-Stone KM, Torgrimson B, Horak F, Eisner A, Nail L, Leo MC, Chui S, Luoh SW. Identifying factors associated with falls in postmenopausal breast cancer survivors: a multi-disciplinary approach. Arch Phys Med Rehabil. 2011;92(4):646–52.PubMedPubMedCentral Winters-Stone KM, Torgrimson B, Horak F, Eisner A, Nail L, Leo MC, Chui S, Luoh SW. Identifying factors associated with falls in postmenopausal breast cancer survivors: a multi-disciplinary approach. Arch Phys Med Rehabil. 2011;92(4):646–52.PubMedPubMedCentral
6.
Zurück zum Zitat Kanis JA, Johnell O, Oden A, Dawson A, De Laet C, Jonsson B, Ten,. year probabilities of osteoporotic fractures according to BMD and diagnostic thresholds. Osteoporos Int. 2001;12(12):989–95.PubMed Kanis JA, Johnell O, Oden A, Dawson A, De Laet C, Jonsson B, Ten,. year probabilities of osteoporotic fractures according to BMD and diagnostic thresholds. Osteoporos Int. 2001;12(12):989–95.PubMed
7.
Zurück zum Zitat Compston JE, McClung MR, Leslie WD. Osteoporosis. Lancet. 2019;393(10169):364–76.PubMed Compston JE, McClung MR, Leslie WD. Osteoporosis. Lancet. 2019;393(10169):364–76.PubMed
8.
Zurück zum Zitat Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud KE, Cauley J, Black D, Vogt TM. Risk factors for hip fracture in white women. Study of Osteoporotic Fractures Research Group. N Engl J Med. 1995;332(12):767–73.PubMed Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud KE, Cauley J, Black D, Vogt TM. Risk factors for hip fracture in white women. Study of Osteoporotic Fractures Research Group. N Engl J Med. 1995;332(12):767–73.PubMed
9.
Zurück zum Zitat Cauley JA, Hochberg MC, Lui LY, Palermo L, Ensrud KE, Hillier TA, Nevitt MC, Cummings SR. Long-term risk of incident vertebral fractures. JAMA. 2007;298(23):2761–7.PubMed Cauley JA, Hochberg MC, Lui LY, Palermo L, Ensrud KE, Hillier TA, Nevitt MC, Cummings SR. Long-term risk of incident vertebral fractures. JAMA. 2007;298(23):2761–7.PubMed
10.
Zurück zum Zitat Center JR, Bliuc D, Nguyen TV, Eisman JA. Risk of subsequent fracture after low-trauma fracture in men and women. JAMA. 2007;297(4):387–94.PubMed Center JR, Bliuc D, Nguyen TV, Eisman JA. Risk of subsequent fracture after low-trauma fracture in men and women. JAMA. 2007;297(4):387–94.PubMed
11.
Zurück zum Zitat Gehlbach S, Saag KG, Adachi JD, Hooven FH, Flahive J, Boonen S, Chapurlat RD, Compston JE, Cooper C, Diez-Perez A, Greenspan SL, LaCroix AZ, Netelenbos JC, Pfeilschifter J, Rossini M, Roux C, Sambrook PN, Silverman S, Siris ES, Watts NB, Lindsay R. Previous fractures at multiple sites increase the risk for subsequent fractures: the Global Longitudinal Study of Osteoporosis in Women. J Bone Miner Res. 2012;27(3):645–53.PubMed Gehlbach S, Saag KG, Adachi JD, Hooven FH, Flahive J, Boonen S, Chapurlat RD, Compston JE, Cooper C, Diez-Perez A, Greenspan SL, LaCroix AZ, Netelenbos JC, Pfeilschifter J, Rossini M, Roux C, Sambrook PN, Silverman S, Siris ES, Watts NB, Lindsay R. Previous fractures at multiple sites increase the risk for subsequent fractures: the Global Longitudinal Study of Osteoporosis in Women. J Bone Miner Res. 2012;27(3):645–53.PubMed
12.
Zurück zum Zitat Kanis JA, Johnell O, De Laet C, Johansson H, Oden A, Delmas P, Eisman J, Fujiwara S, Garnero P, Kroger H, McCloskey EV, Mellstrom D, Melton LJ, Pols H, Reeve J, Silman A, Tenenhouse A. A meta-analysis of previous fracture and subsequent fracture risk. Bone. 2004;35(2):375–82.PubMed Kanis JA, Johnell O, De Laet C, Johansson H, Oden A, Delmas P, Eisman J, Fujiwara S, Garnero P, Kroger H, McCloskey EV, Mellstrom D, Melton LJ, Pols H, Reeve J, Silman A, Tenenhouse A. A meta-analysis of previous fracture and subsequent fracture risk. Bone. 2004;35(2):375–82.PubMed
13.
Zurück zum Zitat Kanis JA, Borgstrom F, De Laet C, Johansson H, Johnell O, Jonsson B, Oden A, Zethraeus N, Pfleger B, Khaltaev N. Assessment of fracture risk. Osteoporos Int. 2005;16(6):581–9.PubMed Kanis JA, Borgstrom F, De Laet C, Johansson H, Johnell O, Jonsson B, Oden A, Zethraeus N, Pfleger B, Khaltaev N. Assessment of fracture risk. Osteoporos Int. 2005;16(6):581–9.PubMed
14.
Zurück zum Zitat Rossini M, Adami S, Bertoldo F, Diacinti D, Gatti D, Giannini S, Giusti A, Malavolta N, Minisola S, Osella G, Pedrazzoni M, Sinigaglia L, Viapiana O, Isaia GC. Guidelines for the diagnosis, prevention and management of osteoporosis. Reumatismo. 2016;68(1):1–39.PubMed Rossini M, Adami S, Bertoldo F, Diacinti D, Gatti D, Giannini S, Giusti A, Malavolta N, Minisola S, Osella G, Pedrazzoni M, Sinigaglia L, Viapiana O, Isaia GC. Guidelines for the diagnosis, prevention and management of osteoporosis. Reumatismo. 2016;68(1):1–39.PubMed
15.
Zurück zum Zitat Cheung AM, Heisey R, Srighanthan J. Breast cancer and osteoporosis. Curr Opin Endocrinol Diabetes Obes. 2013;20(6):532–8.PubMed Cheung AM, Heisey R, Srighanthan J. Breast cancer and osteoporosis. Curr Opin Endocrinol Diabetes Obes. 2013;20(6):532–8.PubMed
16.
Zurück zum Zitat Goss PE, Hadji P, Subar M, Abreu P, Thomsen T, Banke-Bochita J. Effects of steroidal and nonsteroidal aromatase inhibitors on markers of bone turnover in healthy postmenopausal women. Breast Cancer Res. 2007;9(4):R52.PubMedPubMedCentral Goss PE, Hadji P, Subar M, Abreu P, Thomsen T, Banke-Bochita J. Effects of steroidal and nonsteroidal aromatase inhibitors on markers of bone turnover in healthy postmenopausal women. Breast Cancer Res. 2007;9(4):R52.PubMedPubMedCentral
17.
Zurück zum Zitat Roman-Blas JA, Castaneda S, Largo R, Herrero-Beaumont G. Osteoarthritis associated with estrogen deficiency. Arthritis Res Ther. 2009;11(5):241.PubMedPubMedCentral Roman-Blas JA, Castaneda S, Largo R, Herrero-Beaumont G. Osteoarthritis associated with estrogen deficiency. Arthritis Res Ther. 2009;11(5):241.PubMedPubMedCentral
18.
Zurück zum Zitat Handforth C, D’Oronzo S, Coleman R, Brown J. Cancer Treatment and Bone Health. Calcif Tissue Int. 2018;102(2):251–64.PubMedPubMedCentral Handforth C, D’Oronzo S, Coleman R, Brown J. Cancer Treatment and Bone Health. Calcif Tissue Int. 2018;102(2):251–64.PubMedPubMedCentral
19.
Zurück zum Zitat Rachner TD, Coleman R, Hadji P, Hofbauer LC. Bone health during endocrine therapy for cancer. Lancet Diabetes Endocrinol. 2018;6(11):901–10.PubMed Rachner TD, Coleman R, Hadji P, Hofbauer LC. Bone health during endocrine therapy for cancer. Lancet Diabetes Endocrinol. 2018;6(11):901–10.PubMed
20.
Zurück zum Zitat Hadji P, Gnant M, Body JJ, Bundred NJ, Brufsky A, Coleman RE, Guise TA, Lipton A, Aapro MS. Cancer treatment-induced bone loss in premenopausal women: a need for therapeutic intervention? Cancer Treat Rev. 2012;38(6):798–806.PubMed Hadji P, Gnant M, Body JJ, Bundred NJ, Brufsky A, Coleman RE, Guise TA, Lipton A, Aapro MS. Cancer treatment-induced bone loss in premenopausal women: a need for therapeutic intervention? Cancer Treat Rev. 2012;38(6):798–806.PubMed
21.
Zurück zum Zitat Zaman K, Thurlimann B, Huober J, Schonenberger A, Pagani O, Luthi J, Simcock M, Giobbie-Hurder A, Berthod G, Genton C, Brauchli P, Aebi S, Swiss Group for Clinical Cancer. Bone mineral density in breast cancer patients treated with adjuvant letrozole, tamoxifen, or sequences of letrozole and tamoxifen in the BIG 1–98 study (SAKK 21/07). Ann Oncol. 2012;23(6):1474–81.PubMed Zaman K, Thurlimann B, Huober J, Schonenberger A, Pagani O, Luthi J, Simcock M, Giobbie-Hurder A, Berthod G, Genton C, Brauchli P, Aebi S, Swiss Group for Clinical Cancer. Bone mineral density in breast cancer patients treated with adjuvant letrozole, tamoxifen, or sequences of letrozole and tamoxifen in the BIG 1–98 study (SAKK 21/07). Ann Oncol. 2012;23(6):1474–81.PubMed
22.
Zurück zum Zitat Nicks KM, Perrien DS, Akel NS, Suva LJ, Gaddy D. Regulation of osteoblastogenesis and osteoclastogenesis by the other reproductive hormones. Activin Inhibin Mol Cell Endocrinol. 2009;310(1–2):11–20.PubMed Nicks KM, Perrien DS, Akel NS, Suva LJ, Gaddy D. Regulation of osteoblastogenesis and osteoclastogenesis by the other reproductive hormones. Activin Inhibin Mol Cell Endocrinol. 2009;310(1–2):11–20.PubMed
23.
Zurück zum Zitat Mirza F, Canalis E. Management of endocrine disease: Secondary osteoporosis: pathophysiology and management. Eur J Endocrinol. 2015;173(3):R131–51.PubMedPubMedCentral Mirza F, Canalis E. Management of endocrine disease: Secondary osteoporosis: pathophysiology and management. Eur J Endocrinol. 2015;173(3):R131–51.PubMedPubMedCentral
24.
Zurück zum Zitat Greenspan SL, Coates P, Sereika SM, Nelson JB, Trump DL, Resnick NM. Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J Clin Endocrinol Metab. 2005;90(12):6410–7.PubMed Greenspan SL, Coates P, Sereika SM, Nelson JB, Trump DL, Resnick NM. Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J Clin Endocrinol Metab. 2005;90(12):6410–7.PubMed
25.
Zurück zum Zitat Smith MR, Lee WC, Brandman J, Wang Q, Botteman M, Pashos CL. Gonadotropin-releasing hormone agonists and fracture risk: a claims-based cohort study of men with nonmetastatic prostate cancer. J Clin Oncol. 2005;23(31):7897–903.PubMed Smith MR, Lee WC, Brandman J, Wang Q, Botteman M, Pashos CL. Gonadotropin-releasing hormone agonists and fracture risk: a claims-based cohort study of men with nonmetastatic prostate cancer. J Clin Oncol. 2005;23(31):7897–903.PubMed
26.
Zurück zum Zitat Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med. 2005;352(2):154–64.PubMed Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med. 2005;352(2):154–64.PubMed
27.
Zurück zum Zitat Morote J, Morin JP, Orsola A, Abascal JM, Salvador C, Trilla E, Raventos CX, Cecchini L, Encabo G, Reventos J. Prevalence of osteoporosis during long-term androgen deprivation therapy in patients with prostate cancer. Urology. 2007;69(3):500–4.PubMed Morote J, Morin JP, Orsola A, Abascal JM, Salvador C, Trilla E, Raventos CX, Cecchini L, Encabo G, Reventos J. Prevalence of osteoporosis during long-term androgen deprivation therapy in patients with prostate cancer. Urology. 2007;69(3):500–4.PubMed
28.
Zurück zum Zitat Bao T, Peppone LJ, Robinson P. Addressing the unintentional consequences of cancer therapy with novel integrative therapeutics. Am Soc Clin Oncol Educ Book. 2018;38:822–8.PubMed Bao T, Peppone LJ, Robinson P. Addressing the unintentional consequences of cancer therapy with novel integrative therapeutics. Am Soc Clin Oncol Educ Book. 2018;38:822–8.PubMed
29.
Zurück zum Zitat Galvao DA, Spry NA, Taaffe DR, Newton RU, Stanley J, Shannon T, Rowling C, Prince R. Changes in muscle, fat and bone mass after 36 weeks of maximal androgen blockade for prostate cancer. BJU Int. 2008;102(1):44–7.PubMed Galvao DA, Spry NA, Taaffe DR, Newton RU, Stanley J, Shannon T, Rowling C, Prince R. Changes in muscle, fat and bone mass after 36 weeks of maximal androgen blockade for prostate cancer. BJU Int. 2008;102(1):44–7.PubMed
30.
Zurück zum Zitat Bylow K, Hemmerich J, Mohile SG, Stadler WM, Sajid S, Dale W. Obese frailty, physical performance deficits, and falls in older men with biochemical recurrence of prostate cancer on androgen deprivation therapy: a case-control study. Urology. 2011;77(4):934–40.PubMed Bylow K, Hemmerich J, Mohile SG, Stadler WM, Sajid S, Dale W. Obese frailty, physical performance deficits, and falls in older men with biochemical recurrence of prostate cancer on androgen deprivation therapy: a case-control study. Urology. 2011;77(4):934–40.PubMed
31.
Zurück zum Zitat Buehring B, Viswanathan R, Binkley N, Busse W. Glucocorticoid-induced osteoporosis: an update on effects and management. J Allergy Clin Immunol. 2013;132(5):1019–30.PubMed Buehring B, Viswanathan R, Binkley N, Busse W. Glucocorticoid-induced osteoporosis: an update on effects and management. J Allergy Clin Immunol. 2013;132(5):1019–30.PubMed
32.
Zurück zum Zitat Tuck SP, Hanusch B, Walker J, Datta HK. Prostate cancer and osteoporosis. Curr Osteoporos Rep. 2013;11(1):11–20.PubMed Tuck SP, Hanusch B, Walker J, Datta HK. Prostate cancer and osteoporosis. Curr Osteoporos Rep. 2013;11(1):11–20.PubMed
33.
Zurück zum Zitat Iwamura M, Abrahamsson PA, Foss KA, Wu G, Cockett AT, Deftos LJ. Parathyroid hormone-related protein: a potential autocrine growth regulator in human prostate cancer cell lines. Urology. 1994;43(5):675–9.PubMed Iwamura M, Abrahamsson PA, Foss KA, Wu G, Cockett AT, Deftos LJ. Parathyroid hormone-related protein: a potential autocrine growth regulator in human prostate cancer cell lines. Urology. 1994;43(5):675–9.PubMed
34.
Zurück zum Zitat Castaneda S, Carmona L, Carvajal I, Arranz R, Diaz A, Garcia-Vadillo A. Reduction of bone mass in women after bone marrow transplantation. Calcif Tissue Int. 1997;60(4):343–7.PubMed Castaneda S, Carmona L, Carvajal I, Arranz R, Diaz A, Garcia-Vadillo A. Reduction of bone mass in women after bone marrow transplantation. Calcif Tissue Int. 1997;60(4):343–7.PubMed
35.
Zurück zum Zitat Kendler DL, Body JJ, Brandi ML, Broady R, Cannata-Andia J, Cannata-Ortiz MJ, El Maghraoui A, Guglielmi G, Hadji P, Pierroz DD, de Villiers TJ, Rizzoli R, Ebeling PR, International Osteoporosis Foundation Committee of Scientific Advisors Working Group on, C. Bone management in hematologic stem cell transplant recipients. Osteoporos Int. 2018;29(12):2597–610.PubMed Kendler DL, Body JJ, Brandi ML, Broady R, Cannata-Andia J, Cannata-Ortiz MJ, El Maghraoui A, Guglielmi G, Hadji P, Pierroz DD, de Villiers TJ, Rizzoli R, Ebeling PR, International Osteoporosis Foundation Committee of Scientific Advisors Working Group on, C. Bone management in hematologic stem cell transplant recipients. Osteoporos Int. 2018;29(12):2597–610.PubMed
36.
Zurück zum Zitat Yao S, Smiley SL, West K, Lamonica D, Battiwalla M, McCarthy PL Jr, Hahn T. Accelerated bone mineral density loss occurs with similar incidence and severity, but with different risk factors, after autologous versus allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2010;16(8):1130–7.PubMed Yao S, Smiley SL, West K, Lamonica D, Battiwalla M, McCarthy PL Jr, Hahn T. Accelerated bone mineral density loss occurs with similar incidence and severity, but with different risk factors, after autologous versus allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2010;16(8):1130–7.PubMed
37.
Zurück zum Zitat Ebeling PR, Thomas DM, Erbas B, Hopper JL, Szer J, Grigg AP. Mechanisms of bone loss following allogeneic and autologous hemopoietic stem cell transplantation. J Bone Miner Res. 1999;14(3):342–50.PubMed Ebeling PR, Thomas DM, Erbas B, Hopper JL, Szer J, Grigg AP. Mechanisms of bone loss following allogeneic and autologous hemopoietic stem cell transplantation. J Bone Miner Res. 1999;14(3):342–50.PubMed
38.
Zurück zum Zitat Kanis JA, Melton LJ 3rd, Christiansen C, Johnston CC, Khaltaev N. The diagnosis of osteoporosis. J Bone Miner Res. 1994;9(8):1137–41.PubMed Kanis JA, Melton LJ 3rd, Christiansen C, Johnston CC, Khaltaev N. The diagnosis of osteoporosis. J Bone Miner Res. 1994;9(8):1137–41.PubMed
39.
Zurück zum Zitat Vasikaran S, Eastell R, Bruyere O, Foldes AJ, Garnero P, Griesmacher A, McClung M, Morris HA, Silverman S, Trenti T, Wahl DA, Cooper C, Kanis JA, Group, I.-I.B.M.S.W. Markers of bone turnover for the prediction of fracture risk and monitoring of osteoporosis treatment: a need for international reference standards. Osteoporos Int. 2011;22(2):391–420.PubMed Vasikaran S, Eastell R, Bruyere O, Foldes AJ, Garnero P, Griesmacher A, McClung M, Morris HA, Silverman S, Trenti T, Wahl DA, Cooper C, Kanis JA, Group, I.-I.B.M.S.W. Markers of bone turnover for the prediction of fracture risk and monitoring of osteoporosis treatment: a need for international reference standards. Osteoporos Int. 2011;22(2):391–420.PubMed
40.
Zurück zum Zitat Kuo T-R, Chen C-H. Bone biomarker for the clinical assessment of osteoporosis: recent developments and future perspectives. Biomarker Res. 2017;5(1):18. Kuo T-R, Chen C-H. Bone biomarker for the clinical assessment of osteoporosis: recent developments and future perspectives. Biomarker Res. 2017;5(1):18.
41.
Zurück zum Zitat Borgen TT, Solberg LB, Lauritzen T, Apalset EM, Bjørnerem Å, Eriksen EF. Target values and daytime variation of bone turnover markers in monitoring osteoporosis treatment after fractures. JBMR Plus, 2022;n/a(n/a): e10633. Borgen TT, Solberg LB, Lauritzen T, Apalset EM, Bjørnerem Å, Eriksen EF. Target values and daytime variation of bone turnover markers in monitoring osteoporosis treatment after fractures. JBMR Plus, 2022;n/a(n/a): e10633.
42.
Zurück zum Zitat Eastell R, Black DM, Lui L-Y, Chines A, Marin F, Khosla S, de Papp AE, Cauley JA, Mitlak B, McCulloch CE, Vittinghoff E, Bauer DC, for the Foundation for the National Institutes of Health Bone Quality, P. Treatment-Related Changes in Bone Turnover and Fracture Risk Reduction in Clinical Trials of Antiresorptive Drugs: Proportion of Treatment Effect Explained. J Bone Miner Res. 2021;36(2):236–43.PubMed Eastell R, Black DM, Lui L-Y, Chines A, Marin F, Khosla S, de Papp AE, Cauley JA, Mitlak B, McCulloch CE, Vittinghoff E, Bauer DC, for the Foundation for the National Institutes of Health Bone Quality, P. Treatment-Related Changes in Bone Turnover and Fracture Risk Reduction in Clinical Trials of Antiresorptive Drugs: Proportion of Treatment Effect Explained. J Bone Miner Res. 2021;36(2):236–43.PubMed
43.
Zurück zum Zitat Singer FR, Eyre DR. Using biochemical markers of bone turnover in clinical practice. Cleve Clin J Med. 2008;75(10):739–50.PubMed Singer FR, Eyre DR. Using biochemical markers of bone turnover in clinical practice. Cleve Clin J Med. 2008;75(10):739–50.PubMed
44.
Zurück zum Zitat Agostini D, Zeppa Donati S, Lucertini F, Annibalini G, Gervasi M, Ferri Marini C, Piccoli G, Stocchi V, Barbieri E, Sestili P. Muscle and bone health in postmenopausal women: role of protein and vitamin d supplementation combined with exercise training. Nutrients. 2018;10(8):1103.PubMedCentral Agostini D, Zeppa Donati S, Lucertini F, Annibalini G, Gervasi M, Ferri Marini C, Piccoli G, Stocchi V, Barbieri E, Sestili P. Muscle and bone health in postmenopausal women: role of protein and vitamin d supplementation combined with exercise training. Nutrients. 2018;10(8):1103.PubMedCentral
45.
Zurück zum Zitat de Jong N, Chin APMJ, de Groot LC, Hiddink GJ, van Staveren WA. Dietary supplements and physical exercise affecting bone and body composition in frail elderly persons. Am J Public Health. 2000;90(6):947–54.PubMedPubMedCentral de Jong N, Chin APMJ, de Groot LC, Hiddink GJ, van Staveren WA. Dietary supplements and physical exercise affecting bone and body composition in frail elderly persons. Am J Public Health. 2000;90(6):947–54.PubMedPubMedCentral
46.
Zurück zum Zitat Bischoff HA, Stahelin HB, Dick W, Akos R, Knecht M, Salis C, Nebiker M, Theiler R, Pfeifer M, Begerow B, Lew RA, Conzelmann M. Effects of vitamin D and calcium supplementation on falls: a randomized controlled trial. J Bone Miner Res. 2003;18(2):343–51.PubMed Bischoff HA, Stahelin HB, Dick W, Akos R, Knecht M, Salis C, Nebiker M, Theiler R, Pfeifer M, Begerow B, Lew RA, Conzelmann M. Effects of vitamin D and calcium supplementation on falls: a randomized controlled trial. J Bone Miner Res. 2003;18(2):343–51.PubMed
47.
Zurück zum Zitat Pineda-Moncusi M, Garcia-Giralt N, Diez-Perez A, Servitja S, Tusquets I, Prieto-Alhambra D, Nogues X. Increased fracture risk in women treated with aromatase inhibitors versus tamoxifen: beneficial effect of bisphosphonates. J Bone Miner Res. 2020;35(2):291–7.PubMed Pineda-Moncusi M, Garcia-Giralt N, Diez-Perez A, Servitja S, Tusquets I, Prieto-Alhambra D, Nogues X. Increased fracture risk in women treated with aromatase inhibitors versus tamoxifen: beneficial effect of bisphosphonates. J Bone Miner Res. 2020;35(2):291–7.PubMed
48.
Zurück zum Zitat Prieto-Alhambra D, Javaid MK, Servitja S, Arden NK, Martinez-García M, Diez-Perez A, Albanell J, Tusquets I, Nogues X. Vitamin D threshold to prevent aromatase inhibitor-induced arthralgia: a prospective cohort study. Breast Cancer Res Treat. 2011;125(3):869–78.PubMed Prieto-Alhambra D, Javaid MK, Servitja S, Arden NK, Martinez-García M, Diez-Perez A, Albanell J, Tusquets I, Nogues X. Vitamin D threshold to prevent aromatase inhibitor-induced arthralgia: a prospective cohort study. Breast Cancer Res Treat. 2011;125(3):869–78.PubMed
51.
Zurück zum Zitat Planas J, Morote J, Orsola A, Salvador C, Trilla E, Cecchini L, Raventos CX. The relationship between daily calcium intake and bone mineral density in men with prostate cancer. BJU Int. 2007;99(4):812–5.PubMed Planas J, Morote J, Orsola A, Salvador C, Trilla E, Cecchini L, Raventos CX. The relationship between daily calcium intake and bone mineral density in men with prostate cancer. BJU Int. 2007;99(4):812–5.PubMed
52.
Zurück zum Zitat Dueregger A, Heidegger I, Ofer P, Perktold B, Ramoner R, Klocker H, Eder IE. The use of dietary supplements to alleviate androgen deprivation therapy side effects during prostate cancer treatment. Nutrients. 2014;6(10):4491–519.PubMedPubMedCentral Dueregger A, Heidegger I, Ofer P, Perktold B, Ramoner R, Klocker H, Eder IE. The use of dietary supplements to alleviate androgen deprivation therapy side effects during prostate cancer treatment. Nutrients. 2014;6(10):4491–519.PubMedPubMedCentral
53.
Zurück zum Zitat Karampela I, Sakelliou A, Vallianou N, Christodoulatos GS, Magkos F, Dalamaga M. Vitamin D and obesity: current evidence and controversies. Curr Obes Rep. 2021;10(2):162–80.PubMed Karampela I, Sakelliou A, Vallianou N, Christodoulatos GS, Magkos F, Dalamaga M. Vitamin D and obesity: current evidence and controversies. Curr Obes Rep. 2021;10(2):162–80.PubMed
54.
Zurück zum Zitat Abdullah Thani NSI, Khairudin R, Ho JJ, Muhamad NA, Ismail H. Vitamin D supplementation for overweight or obese adults. Cochrane Database Syst. Rev. 2019(5): CD011629. Abdullah Thani NSI, Khairudin R, Ho JJ, Muhamad NA, Ismail H. Vitamin D supplementation for overweight or obese adults. Cochrane Database Syst. Rev. 2019(5): CD011629.
55.
Zurück zum Zitat Migliaccio S, Di Nisio A, Mele C, Scappaticcio L, Savastano S, Colao A. Obesity programs of nutrition, E.R., and assessment, G., obesity and hypovitaminosis D: causality or casualty? International journal of obesity supplements, 2019. 9(1): 20–31. Migliaccio S, Di Nisio A, Mele C, Scappaticcio L, Savastano S, Colao A. Obesity programs of nutrition, E.R., and assessment, G., obesity and hypovitaminosis D: causality or casualty? International journal of obesity supplements, 2019. 9(1): 20–31.
56.
Zurück zum Zitat Pollan M, Casla-Barrio S, Alfaro J, Esteban C, Segui-Palmer MA, Lucia A, Martin M. Exercise and cancer: a position statement from the Spanish Society of Medical Oncology. Clin Transl Oncol. 2020;22:1710.PubMedPubMedCentral Pollan M, Casla-Barrio S, Alfaro J, Esteban C, Segui-Palmer MA, Lucia A, Martin M. Exercise and cancer: a position statement from the Spanish Society of Medical Oncology. Clin Transl Oncol. 2020;22:1710.PubMedPubMedCentral
57.
Zurück zum Zitat Dalla Via J, Daly RM, Fraser SF. The effect of exercise on bone mineral density in adult cancer survivors: a systematic review and meta-analysis. Osteoporos Int. 2018;29(2):287–303.PubMed Dalla Via J, Daly RM, Fraser SF. The effect of exercise on bone mineral density in adult cancer survivors: a systematic review and meta-analysis. Osteoporos Int. 2018;29(2):287–303.PubMed
58.
Zurück zum Zitat Joseph JS, Lam V, Patel MI. Preventing osteoporosis in men taking androgen deprivation therapy for prostate cancer: a systematic review and meta-analysis. Eur Urol Oncol. 2019;2(5):551–61.PubMed Joseph JS, Lam V, Patel MI. Preventing osteoporosis in men taking androgen deprivation therapy for prostate cancer: a systematic review and meta-analysis. Eur Urol Oncol. 2019;2(5):551–61.PubMed
59.
Zurück zum Zitat Fornusek CP, Kilbreath SL. Exercise for improving bone health in women treated for stages I-III breast cancer: a systematic review and meta-analyses. J Cancer Surviv. 2017;11(5):525–41.PubMed Fornusek CP, Kilbreath SL. Exercise for improving bone health in women treated for stages I-III breast cancer: a systematic review and meta-analyses. J Cancer Surviv. 2017;11(5):525–41.PubMed
60.
Zurück zum Zitat Casla S, Hojman P, Márquez-Rodas I, López-Tarruella S, Jerez Y, Barakat R, Martín M. Running away from side effects: physical exercise as a complementary intervention for breast cancer patients. Clin Transl Oncol. 2015;17(3):180–96.PubMed Casla S, Hojman P, Márquez-Rodas I, López-Tarruella S, Jerez Y, Barakat R, Martín M. Running away from side effects: physical exercise as a complementary intervention for breast cancer patients. Clin Transl Oncol. 2015;17(3):180–96.PubMed
61.
Zurück zum Zitat Varahra A, Rodrigues IB, MacDermid JC, Bryant D, Birmingham T. Exercise to improve functional outcomes in persons with osteoporosis: a systematic review and meta-analysis. Osteoporos Int. 2018;29(2):265–86.PubMed Varahra A, Rodrigues IB, MacDermid JC, Bryant D, Birmingham T. Exercise to improve functional outcomes in persons with osteoporosis: a systematic review and meta-analysis. Osteoporos Int. 2018;29(2):265–86.PubMed
62.
Zurück zum Zitat Coughlin SS, Caplan L, Stone R, Stewart J. A review of home-based physical activity interventions for breast cancer survivors. Curr Cancer Rep. 2019;1(1):6–12.PubMedPubMedCentral Coughlin SS, Caplan L, Stone R, Stewart J. A review of home-based physical activity interventions for breast cancer survivors. Curr Cancer Rep. 2019;1(1):6–12.PubMedPubMedCentral
64.
Zurück zum Zitat Yunfeng G, Weiyang H, Xueyang H, Yilong H, Xin G. Exercise overcome adverse effects among prostate cancer patients receiving androgen deprivation therapy: An update meta-analysis. Medicine (Baltimore). 2017;96(27): e7368. Yunfeng G, Weiyang H, Xueyang H, Yilong H, Xin G. Exercise overcome adverse effects among prostate cancer patients receiving androgen deprivation therapy: An update meta-analysis. Medicine (Baltimore). 2017;96(27): e7368.
65.
Zurück zum Zitat Orchard TS, Pan X, Cheek F, Ing SW, Jackson RD. A systematic review of omega-3 fatty acids and osteoporosis. Br J Nutr. 2012;107(Suppl 2):S253–60.PubMedPubMedCentral Orchard TS, Pan X, Cheek F, Ing SW, Jackson RD. A systematic review of omega-3 fatty acids and osteoporosis. Br J Nutr. 2012;107(Suppl 2):S253–60.PubMedPubMedCentral
66.
Zurück zum Zitat O'Carrigan B, Wong MH, Willson ML, Stockler MR, Pavlakis N, Goodwin A. Bisphosphonates and other bone agents for breast cancer. Cochrane Database Syst Rev, 2017. 10: CD003474. O'Carrigan B, Wong MH, Willson ML, Stockler MR, Pavlakis N, Goodwin A. Bisphosphonates and other bone agents for breast cancer. Cochrane Database Syst Rev, 2017. 10: CD003474.
67.
Zurück zum Zitat Brufsky A, Harker WG, Beck JT, Carroll R, Tan-Chiu E, Seidler C, Hohneker J, Lacerna L, Petrone S, Perez EA. Zoledronic acid inhibits adjuvant letrozole-induced bone loss in postmenopausal women with early breast cancer. J Clin Oncol. 2007;25(7):829–36.PubMed Brufsky A, Harker WG, Beck JT, Carroll R, Tan-Chiu E, Seidler C, Hohneker J, Lacerna L, Petrone S, Perez EA. Zoledronic acid inhibits adjuvant letrozole-induced bone loss in postmenopausal women with early breast cancer. J Clin Oncol. 2007;25(7):829–36.PubMed
68.
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. Final 5-year results of Z-FAST trial: adjuvant zoledronic acid maintains bone mass in postmenopausal breast cancer patients receiving letrozole. Cancer. 2012;118(5):1192–201.PubMed 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. Final 5-year results of Z-FAST trial: adjuvant zoledronic acid maintains bone mass in postmenopausal breast cancer patients receiving letrozole. Cancer. 2012;118(5):1192–201.PubMed
69.
Zurück zum Zitat Eidtmann H, de Boer R, Bundred N, Llombart-Cussac A, Davidson N, Neven P, von Minckwitz G, Miller J, Schenk N, Coleman R. Efficacy of zoledronic acid in postmenopausal women with early breast cancer receiving adjuvant letrozole: 36-month results of the ZO-FAST Study. Ann Oncol. 2010;21(11):2188–94.PubMed Eidtmann H, de Boer R, Bundred N, Llombart-Cussac A, Davidson N, Neven P, von Minckwitz G, Miller J, Schenk N, Coleman R. Efficacy of zoledronic acid in postmenopausal women with early breast cancer receiving adjuvant letrozole: 36-month results of the ZO-FAST Study. Ann Oncol. 2010;21(11):2188–94.PubMed
70.
Zurück zum Zitat Hines SL, Mincey B, Dentchev T, Sloan JA, Perez EA, Johnson DB, Schaefer PL, Alberts S, Liu H, Kahanic S, Mazurczak MA, Nikcevich DA, Loprinzi CL. Immediate versus delayed zoledronic acid for prevention of bone loss in postmenopausal women with breast cancer starting letrozole after tamoxifen-N03CC. Breast Cancer Res Treat. 2009;117(3):603–9.PubMedPubMedCentral Hines SL, Mincey B, Dentchev T, Sloan JA, Perez EA, Johnson DB, Schaefer PL, Alberts S, Liu H, Kahanic S, Mazurczak MA, Nikcevich DA, Loprinzi CL. Immediate versus delayed zoledronic acid for prevention of bone loss in postmenopausal women with breast cancer starting letrozole after tamoxifen-N03CC. Breast Cancer Res Treat. 2009;117(3):603–9.PubMedPubMedCentral
71.
Zurück zum Zitat Wagner-Johnston ND, Sloan JA, Liu H, Kearns AE, Hines SL, Puttabasavaiah S, Dakhil SR, Lafky JM, Perez EA, Loprinzi CL. 5-year follow-up of a randomized controlled trial of immediate versus delayed zoledronic acid for the prevention of bone loss in postmenopausal women with breast cancer starting letrozole after tamoxifen: N03CC (Alliance) trial. Cancer. 2015;121(15):2537–43.PubMed Wagner-Johnston ND, Sloan JA, Liu H, Kearns AE, Hines SL, Puttabasavaiah S, Dakhil SR, Lafky JM, Perez EA, Loprinzi CL. 5-year follow-up of a randomized controlled trial of immediate versus delayed zoledronic acid for the prevention of bone loss in postmenopausal women with breast cancer starting letrozole after tamoxifen: N03CC (Alliance) trial. Cancer. 2015;121(15):2537–43.PubMed
72.
Zurück zum Zitat Van Poznak C, Hannon RA, Mackey JR, Campone M, Apffelstaedt JP, Clack G, Barlow D, Makris A, Eastell R. Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol. 2010;28(6):967–75.PubMed Van Poznak C, Hannon RA, Mackey JR, Campone M, Apffelstaedt JP, Clack G, Barlow D, Makris A, Eastell R. Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol. 2010;28(6):967–75.PubMed
73.
Zurück zum Zitat Greenspan SL, Vujevich KT, Brufsky A, Lembersky BC, van Londen GJ, Jankowitz RC, Puhalla SL, Rastogi P, Perera S. Prevention of bone loss with risedronate in breast cancer survivors: a randomized, controlled clinical trial. Osteoporos Int. 2015;26(6):1857–64.PubMedPubMedCentral Greenspan SL, Vujevich KT, Brufsky A, Lembersky BC, van Londen GJ, Jankowitz RC, Puhalla SL, Rastogi P, Perera S. Prevention of bone loss with risedronate in breast cancer survivors: a randomized, controlled clinical trial. Osteoporos Int. 2015;26(6):1857–64.PubMedPubMedCentral
74.
Zurück zum Zitat Prasad C, Greenspan SL, Vujevich KT, Brufsky A, Lembersky BC, van Londen GJ, Jankowitz RC, Puhalla SL, Rastogi P, Perera S. Risedronate may preserve bone microarchitecture in breast cancer survivors on aromatase inhibitors: A randomized, controlled clinical trial. Bone. 2016;90:123–6.PubMed Prasad C, Greenspan SL, Vujevich KT, Brufsky A, Lembersky BC, van Londen GJ, Jankowitz RC, Puhalla SL, Rastogi P, Perera S. Risedronate may preserve bone microarchitecture in breast cancer survivors on aromatase inhibitors: A randomized, controlled clinical trial. Bone. 2016;90:123–6.PubMed
75.
Zurück zum Zitat Alibhai SMH, Zukotynski K, Walker-Dilks C, Emmenegger U, Finelli A, Morgan SC, Hotte SJ, Tomlinson GA, Winquist E. Bone Health and bone-targeted therapies for nonmetastatic prostate cancer: a systematic review and meta-analysis. Ann Intern Med. 2017;167(5):341–50.PubMed Alibhai SMH, Zukotynski K, Walker-Dilks C, Emmenegger U, Finelli A, Morgan SC, Hotte SJ, Tomlinson GA, Winquist E. Bone Health and bone-targeted therapies for nonmetastatic prostate cancer: a systematic review and meta-analysis. Ann Intern Med. 2017;167(5):341–50.PubMed
76.
Zurück zum Zitat Ding H, Yang L, Du W, Teng Y, Fu SJ, Tao Y, Lu JZ, Wang ZP. Bisphosphonates for osteoporosis in nonmetastatic prostate cancer patients receiving androgen-deprivation therapy: a systematic review and meta-analysis. Asian Pac J Cancer Prev. 2013;14(5):3337–43.PubMed Ding H, Yang L, Du W, Teng Y, Fu SJ, Tao Y, Lu JZ, Wang ZP. Bisphosphonates for osteoporosis in nonmetastatic prostate cancer patients receiving androgen-deprivation therapy: a systematic review and meta-analysis. Asian Pac J Cancer Prev. 2013;14(5):3337–43.PubMed
77.
Zurück zum Zitat Saylor PJ, Rumble RB, Tagawa S, Eastham JA, Finelli A, Reddy PS, Kungel TM, Nissenberg MG, Michalski JM. Bone health and bone-targeted therapies for prostate cancer: ASCO endorsement of a cancer care ontario guideline. J Clin Oncol. 2020;38(15):1736–43.PubMed Saylor PJ, Rumble RB, Tagawa S, Eastham JA, Finelli A, Reddy PS, Kungel TM, Nissenberg MG, Michalski JM. Bone health and bone-targeted therapies for prostate cancer: ASCO endorsement of a cancer care ontario guideline. J Clin Oncol. 2020;38(15):1736–43.PubMed
78.
Zurück zum Zitat Bone HG, Wagman RB, Brandi ML, Brown JP, Chapurlat R, Cummings SR, Czerwinski E, Fahrleitner-Pammer A, Kendler DL, Lippuner K, Reginster JY, Roux C, Malouf J, Bradley MN, Daizadeh NS, Wang A, Dakin P, Pannacciulli N, Dempster DW, Papapoulos S. 10 years of denosumab treatment in postmenopausal women with osteoporosis: results from the phase 3 randomised FREEDOM trial and open-label extension. Lancet Diabetes Endocrinol. 2017;5(7):513–23.PubMed Bone HG, Wagman RB, Brandi ML, Brown JP, Chapurlat R, Cummings SR, Czerwinski E, Fahrleitner-Pammer A, Kendler DL, Lippuner K, Reginster JY, Roux C, Malouf J, Bradley MN, Daizadeh NS, Wang A, Dakin P, Pannacciulli N, Dempster DW, Papapoulos S. 10 years of denosumab treatment in postmenopausal women with osteoporosis: results from the phase 3 randomised FREEDOM trial and open-label extension. Lancet Diabetes Endocrinol. 2017;5(7):513–23.PubMed
79.
Zurück zum Zitat Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, Christiansen C, Trial F. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361(8):756–65.PubMed Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, Christiansen C, Trial F. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361(8):756–65.PubMed
80.
Zurück zum Zitat Zhang N, Zhang Z-K, Yu Y, Zhuo Z, Zhang G, Zhang B-T. Pros and Cons of Denosumab treatment for osteoporosis and implication for RANKL aptamer therapy. Front Cell Develop Biol. 2020;8:325. Zhang N, Zhang Z-K, Yu Y, Zhuo Z, Zhang G, Zhang B-T. Pros and Cons of Denosumab treatment for osteoporosis and implication for RANKL aptamer therapy. Front Cell Develop Biol. 2020;8:325.
81.
Zurück zum Zitat Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Smith J, Fan M, Jun S. Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancer. J Clin Oncol. 2008;26(30):4875–82.PubMed Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Smith J, Fan M, Jun S. Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancer. J Clin Oncol. 2008;26(30):4875–82.PubMed
82.
Zurück zum Zitat Gnant M, Pfeiler G, 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, Austrian B, Colorectal Cancer Study G. Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet, 2015:386(9992); p. 433–43. Gnant M, Pfeiler G, 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, Austrian B, Colorectal Cancer Study G. Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet, 2015:386(9992); p. 433–43.
83.
Zurück zum Zitat Smith MR, Egerdie B, Hernandez Toriz N, Feldman R, Tammela TL, Saad F, Heracek J, Szwedowski M, Ke C, Kupic A, Leder BZ, Goessl C, Denosumab HPCSG. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 2009. 361(8): 745–55. Smith MR, Egerdie B, Hernandez Toriz N, Feldman R, Tammela TL, Saad F, Heracek J, Szwedowski M, Ke C, Kupic A, Leder BZ, Goessl C, Denosumab HPCSG. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 2009. 361(8): 745–55.
84.
Zurück zum Zitat Shapiro CL, Van Poznak C, Lacchetti C, Kirshner J, Eastell R, Gagel R, Smith S, Edwards BJ, Frank E, Lyman GH, Smith MR, Mhaskar R, Henderson T, Neuner J. Management of osteoporosis in survivors of adult cancers with nonmetastatic disease: ASCO clinical practice guideline. J Clin Oncol. 2019;37(31):2916–46.PubMed Shapiro CL, Van Poznak C, Lacchetti C, Kirshner J, Eastell R, Gagel R, Smith S, Edwards BJ, Frank E, Lyman GH, Smith MR, Mhaskar R, Henderson T, Neuner J. Management of osteoporosis in survivors of adult cancers with nonmetastatic disease: ASCO clinical practice guideline. J Clin Oncol. 2019;37(31):2916–46.PubMed
85.
Zurück zum Zitat Reginster J, Bianic F, Campbell R, Martin M, Williams SA, Fitzpatrick LA. Abaloparatide for risk reduction of nonvertebral and vertebral fractures in postmenopausal women with osteoporosis: a network meta-analysis. Osteoporos Int. 2019;30(7):1465–73.PubMedCentral Reginster J, Bianic F, Campbell R, Martin M, Williams SA, Fitzpatrick LA. Abaloparatide for risk reduction of nonvertebral and vertebral fractures in postmenopausal women with osteoporosis: a network meta-analysis. Osteoporos Int. 2019;30(7):1465–73.PubMedCentral
86.
Zurück zum Zitat Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, Wang O, Mitlak BH. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001;344(19):1434–41.PubMed Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, Wang O, Mitlak BH. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001;344(19):1434–41.PubMed
87.
Zurück zum Zitat Riancho JA, Peris P, González-Macías J, Pérez-Castrillón JL, Guías de Práctica Clínica en la Osteoporosis Posmenopáusica, Glicocorticoidea y del varón (actualización 2021). Sociedad Española de Investigación Ósea y del Metabolismo Mineral (SEIOMM). Rev Clin Esp, 2022 (in press). Riancho JA, Peris P, González-Macías J, Pérez-Castrillón JL, Guías de Práctica Clínica en la Osteoporosis Posmenopáusica, Glicocorticoidea y del varón (actualización 2021). Sociedad Española de Investigación Ósea y del Metabolismo Mineral (SEIOMM). Rev Clin Esp, 2022 (in press).
88.
Zurück zum Zitat Langdahl BL, Libanati C, Crittenden DB, Bolognese MA, Brown JP, Daizadeh NS, Dokoupilova E, Engelke K, Finkelstein JS, Genant HK, Goemaere S, Hyldstrup L, Jodar-Gimeno E, Keaveny TM, Kendler D, Lakatos P, Maddox J, Malouf J, Massari FE, Molina JF, Ulla MR, Grauer A. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial. Lancet. 2017;390(10102):1585–94.PubMed Langdahl BL, Libanati C, Crittenden DB, Bolognese MA, Brown JP, Daizadeh NS, Dokoupilova E, Engelke K, Finkelstein JS, Genant HK, Goemaere S, Hyldstrup L, Jodar-Gimeno E, Keaveny TM, Kendler D, Lakatos P, Maddox J, Malouf J, Massari FE, Molina JF, Ulla MR, Grauer A. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial. Lancet. 2017;390(10102):1585–94.PubMed
89.
Zurück zum Zitat McClung MR, Brown JP, Diez-Perez A, Resch H, Caminis J, Meisner P, Bolognese MA, Goemaere S, Bone HG, Zanchetta JR, Maddox J, Bray S, Grauer A. Effects of 24 months of treatment with romosozumab followed by 12 Months of denosumab or placebo in postmenopausal women with low bone mineral density: a randomized, double-blind, phase 2, parallel group study. J Bone Miner Res. 2018;33(8):1397–406.PubMed McClung MR, Brown JP, Diez-Perez A, Resch H, Caminis J, Meisner P, Bolognese MA, Goemaere S, Bone HG, Zanchetta JR, Maddox J, Bray S, Grauer A. Effects of 24 months of treatment with romosozumab followed by 12 Months of denosumab or placebo in postmenopausal women with low bone mineral density: a randomized, double-blind, phase 2, parallel group study. J Bone Miner Res. 2018;33(8):1397–406.PubMed
90.
Zurück zum Zitat Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, Zerbini CAF, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375(16):1532–43.PubMed Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, Zerbini CAF, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375(16):1532–43.PubMed
91.
Zurück zum Zitat Cuzick J, Sestak I, Cawthorn S, Hamed H, Holli K, Howell A, Forbes JF, Investigators I-I. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol. 2015;16(1):67–75.PubMedPubMedCentral Cuzick J, Sestak I, Cawthorn S, Hamed H, Holli K, Howell A, Forbes JF, Investigators I-I. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol. 2015;16(1):67–75.PubMedPubMedCentral
92.
Zurück zum Zitat Rico H, Revilla M, Hernandez ER, Villa LF, Alvarez de Buergo M. Total and regional bone mineral content and fracture rate in postmenopausal osteoporosis treated with salmon calcitonin: a prospective study. Calcif Tissue Int, 1995; 56(3): 181–5. Rico H, Revilla M, Hernandez ER, Villa LF, Alvarez de Buergo M. Total and regional bone mineral content and fracture rate in postmenopausal osteoporosis treated with salmon calcitonin: a prospective study. Calcif Tissue Int, 1995; 56(3): 181–5.
93.
Zurück zum Zitat Carstens JH, Feinblatt JD. Future horizons for calcitonin: a U.S. perspective. Calcif Tissue Int, 1991; 49 Suppl 2: p. S2–6. Carstens JH, Feinblatt JD. Future horizons for calcitonin: a U.S. perspective. Calcif Tissue Int, 1991; 49 Suppl 2: p. S2–6.
94.
Zurück zum Zitat Overgaard K, Riis BJ, Christiansen C, Hansen MA. Effect of salcatonin given intranasally on early postmenopausal bone loss. BMJ. 1989;299(6697):477–9.PubMedPubMedCentral Overgaard K, Riis BJ, Christiansen C, Hansen MA. Effect of salcatonin given intranasally on early postmenopausal bone loss. BMJ. 1989;299(6697):477–9.PubMedPubMedCentral
95.
Zurück zum Zitat Tsourdi E, Zillikens MC, Meier C, Body J-J, Gonzalez Rodriguez E, Anastasilakis AD, Abrahamsen B, McCloskey E, Hofbauer LC, Guañabens N, Obermayer-Pietsch B, Ralston SH, Eastell R, Pepe J, Palermo A, Langdahl B. Fracture risk and management of discontinuation of denosumab therapy: a systematic review and position statement by ECTS. J Clin Endocrinol Metab. 2021;106(1):264–81. Tsourdi E, Zillikens MC, Meier C, Body J-J, Gonzalez Rodriguez E, Anastasilakis AD, Abrahamsen B, McCloskey E, Hofbauer LC, Guañabens N, Obermayer-Pietsch B, Ralston SH, Eastell R, Pepe J, Palermo A, Langdahl B. Fracture risk and management of discontinuation of denosumab therapy: a systematic review and position statement by ECTS. J Clin Endocrinol Metab. 2021;106(1):264–81.
96.
Zurück zum Zitat Meyer AC, Drefahl S, Ahlbom A, Lambe M, Modig K. Trends in life expectancy: did the gap between the healthy and the ill widen or close? BMC Med. 2020;18(1):41.PubMedPubMedCentral Meyer AC, Drefahl S, Ahlbom A, Lambe M, Modig K. Trends in life expectancy: did the gap between the healthy and the ill widen or close? BMC Med. 2020;18(1):41.PubMedPubMedCentral
97.
Zurück zum Zitat Buckley RE, Moran CG, Apivatthakakul T, AO Principles of Fracture Management: Vol. 1: Principles, Vol 2: Specific Fractures. 2017: Thieme Medical Publishers Buckley RE, Moran CG, Apivatthakakul T, AO Principles of Fracture Management: Vol. 1: Principles, Vol 2: Specific Fractures. 2017: Thieme Medical Publishers
98.
Zurück zum Zitat Curtis R, Goldhahn J, Schwyn R, Regazzoni P, Suhm N. Fixation principles in metaphyseal bone–a patent based review. Osteoporos Int. 2005;16(Suppl 2):S54-64.PubMed Curtis R, Goldhahn J, Schwyn R, Regazzoni P, Suhm N. Fixation principles in metaphyseal bone–a patent based review. Osteoporos Int. 2005;16(Suppl 2):S54-64.PubMed
99.
Zurück zum Zitat Johnson CN, Gurich RW Jr, Pavey GJ, Thompson MJ. Contemporary management of appendicular skeletal metastasis by primary tumor type. J Am Acad Orthop Surg. 2019;27(10):345–55.PubMed Johnson CN, Gurich RW Jr, Pavey GJ, Thompson MJ. Contemporary management of appendicular skeletal metastasis by primary tumor type. J Am Acad Orthop Surg. 2019;27(10):345–55.PubMed
100.
Zurück zum Zitat Willeumier JJ, van der Linden YM, van de Sande MAJ, Dijkstra PDS. Treatment of pathological fractures of the long bones. EFORT Open Rev. 2016;1(5):136–45.PubMedPubMedCentral Willeumier JJ, van der Linden YM, van de Sande MAJ, Dijkstra PDS. Treatment of pathological fractures of the long bones. EFORT Open Rev. 2016;1(5):136–45.PubMedPubMedCentral
101.
Zurück zum Zitat Rodrigues L, Cornelis FH, Chevret S. Hip fracture prevention in osteoporotic elderly and cancer patients: an on-line french survey evaluating current needs. Medicina (Kaunas). 2020;56(8):397. Rodrigues L, Cornelis FH, Chevret S. Hip fracture prevention in osteoporotic elderly and cancer patients: an on-line french survey evaluating current needs. Medicina (Kaunas). 2020;56(8):397.
102.
Zurück zum Zitat Giannini S, Chiarello E, Mazzotti A, Tedesco G, Faldini C. Surgical prevention of femoral neck fractures in elderly osteoporotic patients: a randomised controlled study on the prevention nail system device. Hip Int, 2018. 28(2_suppl): 78–83. Giannini S, Chiarello E, Mazzotti A, Tedesco G, Faldini C. Surgical prevention of femoral neck fractures in elderly osteoporotic patients: a randomised controlled study on the prevention nail system device. Hip Int, 2018. 28(2_suppl): 78–83.
103.
Zurück zum Zitat Chiarello E, Tedesco G, Cadossi M, Capra P, Terrando S, Miti A, Giannini S. Surgical prevention of femoral neck fractures in elderly osteoporotic patients. A literature review. Clin Cases Miner Bone Metab. 2016;13(1):42–5.PubMedPubMedCentral Chiarello E, Tedesco G, Cadossi M, Capra P, Terrando S, Miti A, Giannini S. Surgical prevention of femoral neck fractures in elderly osteoporotic patients. A literature review. Clin Cases Miner Bone Metab. 2016;13(1):42–5.PubMedPubMedCentral
104.
Zurück zum Zitat Aghayev K, Papanastassiou ID, Vrionis F. Role of vertebral augmentation procedures in the management of vertebral compression fractures in cancer patients. Curr Opin Support Palliat Care. 2011;5(3):222–6.PubMed Aghayev K, Papanastassiou ID, Vrionis F. Role of vertebral augmentation procedures in the management of vertebral compression fractures in cancer patients. Curr Opin Support Palliat Care. 2011;5(3):222–6.PubMed
105.
Zurück zum Zitat Ollivier M, Turati M, Munier M, Lunebourg A, Argenson JN, Parratte S. Balloon tibioplasty for reduction of depressed tibial plateau fractures: Preliminary radiographic and clinical results. Int Orthop. 2016;40(9):1961–6.PubMed Ollivier M, Turati M, Munier M, Lunebourg A, Argenson JN, Parratte S. Balloon tibioplasty for reduction of depressed tibial plateau fractures: Preliminary radiographic and clinical results. Int Orthop. 2016;40(9):1961–6.PubMed
106.
Zurück zum Zitat Lee FY, Latich I, Toombs C, Mungur A, Conway D, Alder K, Ibe I, Lindskog D, Friedlaender G. Minimally invasive image-guided ablation, osteoplasty, reinforcement, and internal fixation (AORIF) for osteolytic lesions in the pelvis and periarticular regions of weight-bearing bones. J Vasc Interv Radiol. 2020;31(4):649-658e1.PubMed Lee FY, Latich I, Toombs C, Mungur A, Conway D, Alder K, Ibe I, Lindskog D, Friedlaender G. Minimally invasive image-guided ablation, osteoplasty, reinforcement, and internal fixation (AORIF) for osteolytic lesions in the pelvis and periarticular regions of weight-bearing bones. J Vasc Interv Radiol. 2020;31(4):649-658e1.PubMed
107.
Zurück zum Zitat Kammerlander C, Pfeufer D, Lisitano LA, Mehaffey S, Bocker W, Neuerburg C. Inability of older adult patients with hip fracture to maintain postoperative weight-bearing restrictions. J Bone Joint Surg Am. 2018;100(11):936–41.PubMed Kammerlander C, Pfeufer D, Lisitano LA, Mehaffey S, Bocker W, Neuerburg C. Inability of older adult patients with hip fracture to maintain postoperative weight-bearing restrictions. J Bone Joint Surg Am. 2018;100(11):936–41.PubMed
108.
Zurück zum Zitat Ottesen TD, McLynn RP, Galivanche AR, Bagi PS, Zogg CK, Rubin LE, Grauer JN. Increased complications in geriatric patients with a fracture of the hip whose postoperative weight-bearing is restricted: an analysis of 4918 patients. Bone Joint J. 2018;100B(10):1377–84. Ottesen TD, McLynn RP, Galivanche AR, Bagi PS, Zogg CK, Rubin LE, Grauer JN. Increased complications in geriatric patients with a fracture of the hip whose postoperative weight-bearing is restricted: an analysis of 4918 patients. Bone Joint J. 2018;100B(10):1377–84.
109.
Zurück zum Zitat Warren J, Sundaram K, Anis H, McLaughlin J, Patterson B, Higuera CA, Piuzzi NS. The association between weight-bearing status and early complications in hip fractures. Eur J Orthop Surg Traumatol. 2019;29(7):1419–27.PubMed Warren J, Sundaram K, Anis H, McLaughlin J, Patterson B, Higuera CA, Piuzzi NS. The association between weight-bearing status and early complications in hip fractures. Eur J Orthop Surg Traumatol. 2019;29(7):1419–27.PubMed
110.
Zurück zum Zitat Beckmann M, Bruun-Olsen V, Pripp AH, Bergland A, Smith T, Heiberg KE. Effect of exercise interventions in the early phase to improve physical function after hip fracture - A systematic review and meta-analysis. Physiotherapy. 2020;108:90–7.PubMed Beckmann M, Bruun-Olsen V, Pripp AH, Bergland A, Smith T, Heiberg KE. Effect of exercise interventions in the early phase to improve physical function after hip fracture - A systematic review and meta-analysis. Physiotherapy. 2020;108:90–7.PubMed
111.
Zurück zum Zitat Sherrington C, Fairhall N, Kirkham C, Clemson L, Tiedemann A, Vogler C, Close JCT, O’Rourke S, Moseley AM, Cameron ID, Mak JCS, Lord SR. Exercise to reduce mobility disability and prevent falls after fall-related leg or pelvic fracture: RESTORE randomized controlled trial. J Gen Intern Med. 2020;35(10):2907–16.PubMedPubMedCentral Sherrington C, Fairhall N, Kirkham C, Clemson L, Tiedemann A, Vogler C, Close JCT, O’Rourke S, Moseley AM, Cameron ID, Mak JCS, Lord SR. Exercise to reduce mobility disability and prevent falls after fall-related leg or pelvic fracture: RESTORE randomized controlled trial. J Gen Intern Med. 2020;35(10):2907–16.PubMedPubMedCentral
112.
Zurück zum Zitat Pludowski P, Holick MF, Pilz S, Wagner CL, Hollis BW, Grant WB, Shoenfeld Y, Lerchbaum E, Llewellyn DJ, Kienreich K, Soni M. Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality-a review of recent evidence. Autoimmun Rev. 2013;12(10):976–89.PubMed Pludowski P, Holick MF, Pilz S, Wagner CL, Hollis BW, Grant WB, Shoenfeld Y, Lerchbaum E, Llewellyn DJ, Kienreich K, Soni M. Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality-a review of recent evidence. Autoimmun Rev. 2013;12(10):976–89.PubMed
113.
Zurück zum Zitat Hillner BE, Ingle JN, Chlebowski RT, Gralow J, Yee GC, Janjan NA, Cauley JA, Blumenstein BA, Albain KS, Lipton A, Brown S. American society of clinical oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol. 2003;21(21):4042–57.PubMed Hillner BE, Ingle JN, Chlebowski RT, Gralow J, Yee GC, Janjan NA, Cauley JA, Blumenstein BA, Albain KS, Lipton A, Brown S. American society of clinical oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol. 2003;21(21):4042–57.PubMed
Metadaten
Titel
Bone loss induced by cancer treatments in breast and prostate cancer patients
verfasst von
Santos Castañeda
Ana Casas
Aránzazu González-del-Alba
Guillermo Martínez-Díaz-Guerra
Xavier Nogués
Cristina Ojeda Thies
Óscar Torregrosa Suau
Álvaro Rodríguez-Lescure
Publikationsdatum
02.07.2022
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 11/2022
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-022-02872-1

Weitere Artikel der Ausgabe 11/2022

Clinical and Translational Oncology 11/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

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