Skip to main content
Erschienen in: Journal of Translational Medicine 1/2013

Open Access 01.12.2013 | Review

The clinical utility of bone marker measurements in osteoporosis

verfasst von: Gillian Wheater, Mohsen Elshahaly, Stephen P Tuck, Harish K Datta, Jacob M van Laar

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2013

Abstract

Osteoporosis is characterised by low bone mass and structural deterioration of bone tissue, resulting in increased fragility and susceptibility to fracture. Osteoporotic fractures are a significant cause of morbidity and mortality. Direct medical costs from such fractures in the UK are currently estimated at over two billion pounds per year, resulting in a substantial healthcare burden that is expected to rise exponentially due to increasing life expectancy. Currently bone mineral density is the WHO standard for diagnosis of osteoporosis, but poor sensitivity means that potential fractures will be missed if it is used alone. During the past decade considerable progress has been made in the identification and characterisation of specific biomarkers to aid the management of metabolic bone disease. Technological developments have greatly enhanced assay performance producing reliable, rapid, non-invasive cost effective assays with improved sensitivity and specificity. We now have a greater understanding of the need to regulate pre-analytical sample collection to minimise the effects of biological variation. However, bone turnover markers (BTMs) still have limited clinical utility. It is not routinely recommended to use BTMs to select those at risk of fractures, but baseline measurements of resorption markers are useful before commencement of anti-resorptive treatment and can be checked 3–6 months later to monitor response and adherence to treatment. Similarly, formation markers can be used to monitor bone forming agents. BTMs may also be useful when monitoring patients during treatment holidays and aid in the decision as to when therapy should be recommenced. Recent recommendations by the Bone Marker Standards Working Group propose to standardise research and include a specific marker of bone resorption (CTX) and bone formation (P1NP) in all future studies. It is hoped that improved research in turn will lead to optimised markers for the clinical management of osteoporosis and other bone diseases.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1479-5876-11-201) contains supplementary material, which is available to authorized users.

Competing interests

JMvL has received a research grant, consultancy and speaker fees from Roche. SPT has received speaker fees from Ely Lilly.

Author’ contributions

GW drafted the manuscript. ME revised the manuscript. SPT and HKD revised and critically appraised the manuscript. JMvL revised, critically appraised and provided overall supervision for the project. All authors read and approved the final manuscript.
Abkürzungen
AGEs
Advanced glycation end products
ALP
Alkaline phosphatase
BSAP
Bone specific alkaline phosphatase
βCTX
Beta-isomerised carboxy terminal telopeptide of type I collagen
BMD
Bone mineral density
BTM
Bone turnover marker
CTX
Carboxy-terminal cross-linked telopeptides of type 1 collagen
CVA
Total analytical imprecision
CV1
Within-subject coefficient of variation
DKK-1
Dickkopf-related protein 1
DMP1
Dentin matrix protein 1
DPD
Deoxypyridinoline
DXA
Dual-energy X-ray absorptiometry
EDTA
Ethylenediaminetetraacetic acid
Fz
Frizzled protein
GFR
Glomerular filtration rate
GH
Growth hormone
HELP
Type 1 collagen alpha 1 helicoidal peptide
HRT
Hormone replacement therapy
ICTP
Carboxy-terminal cross-linked telopeptide of type 1 collagen
IDS
Immuno-Diagnostic Systems Ltd
IFCC
International Federation of Clinical Chemistry and Laboratory Medicine
IGF1
Insulin-like growth factor 1
IOF
International Osteoporosis Foundation
LRP
Low-density lipoprotein receptor-related protein
LSC
Least significant change
MEPE
Matrix extracellular phosphoglycoprotein
MMP
Matrix metalloproteinases
MoM
Multiple of the median
MoMF
Multiple of the median formation marker
MoMR
Multiple of the median resorption marker
NBHA
National Bone Health Alliance
NTX
Amino-terminal cross-linked telopeptide of type 1 collagen
OB
osteoblast
OC
osteocalcin
OCY
osteocyte
OPG
osteoprotegerin
OPN
osteopontin
PICP
Procollagen type 1 carboxy-terminal propeptide
PINP
Procollagen type 1 amino-terminal propeptide
PPR
PTH/PTHrP receptor
PTH
Parathyroid hormone
PTHrP
Parathyroid hormone related peptide
PYD
pyridinoline
RA
Rheumatoid arthritis
RANK
Receptor activator of nuclear factor kappa B
RANKL
Receptor activator of nuclear factor kappa B ligand
sRANKL
Soluble receptor activator of nuclear factor kappa B ligand
SCL
sclerostin
SD
Standard deviation
SERMs
Selective estrogen receptor moderators
Sost
SclerOSTeosis gene
TRAP
Tartrate resistant acid phosphatase
TSH
Thyroid stimulating hormone
UK
United Kingdom
WHO
World Health Organisation
Wnt
Wingless and Integration-1.

Introduction

Bone is a specialised connective tissue consisting primarily of glycoproteins and proteoglycans. The fibres of bone are mostly composed of type-I collagen impregnated with mineral in the form of hydroxyapatite. The functional integrity and strength of the skeleton is maintained by this highly cross-linked structure. Several factors may be involved in determining bone quality, including bone density and qualitative determinants of bone strength such as the rate of bone turnover, the extent of trabecular connectivity, cortical and periosteal bone size and skeletal morphometry [1]. Bone is metabolically active and is constantly being repaired and remodelled throughout an individual’s lifetime. Approximately twenty percent of bone tissue is replaced annually varying by site and type [2]. Remodelling begins before birth and continues until death, it is a highly synchronised process contained within basic multicellular units (Figure 1). Recent research has demonstrated the role of receptor activator of nuclear factor kappa B ligand/ receptor activator of nuclear factor kappa B/ osteoprotegerin (RANKL/RANK/OPG) in regulating bone metabolism [3]. Parathyroid hormone (PTH), PTH-related peptide (PTH-rP), 1,25-dihydroxyvitamin D3, prostaglandin E2, and interleukins among others regulate bone turnover through this system [4]. Additionally, bone metabolism is now known to be at least partly regulated by osteocytes, the fully differentiated osteoblasts present in lacunae in the mineralised matrix and osteoid tissue of bone [1]. Osteocytes detect mechanical loads and release signalling molecules (Figure 2) which coordinate the recruitment and activity of osteoblasts and osteoclasts thereby controlling bone turnover [5].
Under normal conditions bone formation and resorption are tightly linked through a variety of regulatory signals. Osteoporosis occurs when bone resorption is the more active resulting in a low bone mass and micro-architectural deterioration of bone tissue, leading to increased bone fragility and consequent increase in fracture risk. Osteoporotic fractures are a significant cause of morbidity and mortality, in the year 2010 there were an estimated 300,000 osteoporotic fractures in the UK and direct medical costs from such fractures were estimated at over two billion pounds [8]. Osteoporosis may be either primary (idiopathic) or secondary to a large number of conditions. These include hypogonadism, hyperthyroidism, skeletal metastases, multiple myeloma, anticonvulsant or oral corticosteroid use and alcohol abuse. Up to 30% of women and 55% of men with symptomatic vertebral crush fractures have an underlying cause of secondary osteoporosis [9]. The prevalence of osteoporosis increases with age, bone loss is reportedly more rapid in females in the first few years post menopause and is influenced by oestrogen deficiency [10], but it is also thought to increase in ageing men [11]. The World Health Organisation (WHO) has defined osteoporosis as a bone mineral density (BMD) measured by dual-energy X-ray absorptiometry (DXA) 2.5 standard deviations (SD) or more below the mean peak bone mass of premenopausal females (T-score ≤ −2.5 SD) [12]. Technical developments in the measurement of BMD have led to its adoption as the standard for diagnosis of osteoporosis, however the relatively poor sensitivity contrasting with high specificity means that many potential fractures will be missed if BMD assessment is used alone [13].
In recent years cellular components of the bone matrix have been identified and categorised as either markers of bone formation or resorption. Reliable, rapid, non-invasive, cost effective assays have been developed with improved sensitivity and specificity. Although these markers have been used in research for a long time they are only now being recognised as tools in the clinical management of bone disease. Technological advances have greatly enhanced the accuracy and reliability of bone marker measurement, although assays still vary significantly. In this review we will summarise the most widely used bone turnover makers (BTMs), briefly look at more novel markers and discuss their strengths, weaknesses and their clinical utility in the management of osteoporosis.

Commonly used markers of bone turnover

Biomarkers of bone turnover can be measured in blood or urine and are used in selective combinations of formation and resorption markers that express the metabolic activity of osteoblasts or osteoclasts respectively, although in most circumstances the bone remodelling processes are coupled and tend to change in parallel. BTMs do not control skeletal metabolism and are not disease specific; they reflect the entire skeleton regardless of the underlying cause. Some markers represent both processes, e.g. osteocalcin (OC). Several of the available markers are non-specific, i.e. they are present in tissues other than bone and may therefore be influenced by non-skeletal processes [14]. Results should therefore always take into consideration the whole clinical picture and an understanding of the nature and source of each marker is essential for a comprehensive interpretation. The major advantages and disadvantages of each marker are included in Table 1.
Table 1
Major sources of variability in biochemical markers of bone turnover
Bone marker (Abbreviation)
Source
Action
Advantages
Disadvantages
Analysis and sample type
Formation markers
Bone Alkaline Phosphatase (BAP)
Enzyme present in osteoblast plasma membranes
Enzymatic degradation of the mineralisation inhibitor pyrophosphate at alkaline pH
Low intra-individual variability <10% [15] Not affected by renal function [15] Food has little effect [16] Long circulatory half-life 1–2 days [17] Sample stability [18] Cheap
Up to 20% cross reactivity with liver isoforms [14] Changes with therapy minimal i.e. less than LSC of 25% [15] 2 peaks at 14:00 and 23:30 hrs Nadir 30% ↓at 06:30 [19] Multiple methodologies, can measure mass or activity [20]
Automated and manual immunoassays Serum, EDTA plasma
Osteocalcin (OC)
Major non-collagen bone Gla protein. Produced by osteoblasts during bone formation and bound to hydroxyapatite
Influences osteoid mineralisation Provides negative feedback during remodelling process
EDTA sample more stable [21] Late marker of osteoblast activity [15]
Intact molecule unstable [15] Large inter-lab variation [20] Released during formation and resorption [17] Short half-life of a few minutes [22, 23] Influenced by Vit K status, renal function and circadian variability [15, 17] OC gene regulated at transcriptional level by 1,25-OH2 Vit D Vit K essential co-factor for γ-carboxylation of OC resulting in ↑ affinity for Ca and hydroxyapatite [14]
Automated and manual immunoassays Multiplex microarray Serum, EDTA plasma
Procollagen type 1 Carboxy-terminal Propeptide (P1CP)
Specific product of proliferating osteoblasts and fibroblasts.
Cleaved from type 1 pro-collagen by proteases during type 1 collagen formation
Quantitative measure of newly formed type 1 collagen Thermostability [14]
Short half-life 6-8mins [14] Cleared in liver by mannose receptor so sensitive to thyroid hormones and IGF-1 [20] Highest levels 01:30 – 04:30, up to 20% higher than nadir 11:00 – 15:00 [19] Lacks sensitivity to changes during menopause [14]
Automated and manual immunoassays Serum, EDTA plasma
*Procollagen type 1 amiNo-terminal Propeptide (P1NP)
Specific product of proliferating osteoblasts and fibroblasts.
Cleaved from type 1 pro-collagen by proteases during type 1 collagen formation
Low intra-individual variability[20] Small circadian rhythm[15] Stable at room temp[21] Good assay precision[20] Superior for PMO monitoring - change from baseline ↑up to 80% with anti-resorptive and ↓up to 200% with PTH medication within 3months[15]
Total assay affected by delayed clearance of monomeric fraction e.g. in renal failure or metastatic bone disease[24] Expensive
Automated and manual immunoassays Multiplex microarray Total or Intact fractions Serum, EDTA plasma
Resorption markers Collagen derived
*Carboxy-Terminal cross-linked telopeptides of type 1 collagen (CTX)
Type 1 collagen mainly bone Isomerisation to β aspartyl occurs in mature collagen
Cleaved from type 1 collagen by cathepsin-K during bone resorption
Variability↓ fasting[25] Sample stability, especially EDTA[18][21] Substantial ↓ post anti-resorptive treatment[26] Blood sample now preferential
Large circadian variation – highest 01:30 – 04:30 approx 2x nadir 11:00–15:00[27]
Automated and manual immunoassays Multiplex microarray Urine, serum, EDTA plasma
Carboxy-Terminal cross-linked telopeptides of type 1 collagen (ICTP or CTX-MMP)
Newly synthesised type 1 collagen predominantly bone
Cleaved from type 1 collagen by MMP during bone resorption
 
Large circadian variation [20] Influenced by renal and liver function [20] Not responsive to usual osteoporotic treatments [20]
Manual immunoassay
amiNo-Terminal cross-linked telopeptides of type 1 collagen (NTX)
Type 1 collagen mainly bone
Cleaved from type 1 collagen by cathepsin-K during bone resorption
Urine sample stable [15] uNTX sig predictor of fracture risk in postmenopausal women [28] Small dietary influence, although fasting blood sample preferred [15]
Large circadian variation Influenced by renal and liver function [20] Units based on manufacturer’s calibrator i.e. bone collagen equivalents [15]
Automated and manual immunoassays Urine, serum, EDTA plasma
Type 1 collagen alpha 1 helicoidal peptide (HELP)
Type 1 collagen Amino acid 620–633 sequence of the α chain
Cleaved from helical region of type 1 collagen by cathepsin-K during bone resorption
High correlation to other markers of collagen degradation [14]
24 hr collection – hard to collect 2nd morning void with creatinine correction – additional analytical variability Clinical validity needs further investigation
Manual immunoassay Urinary marker
Deoxypyridinoline (DPD)
Mature type 1 collagen
Cross link released when mature type 1 collagen breaks down Mechanically stabilise the molecule
Reflect degradation of mature collagen only Specific to bone [14] Independent of dietary sources [20] Less invasive than blood
24 hr collection – hard to collect 2nd morning void with creatinine correction – additional analytical variability Circadian variation [20]
Automated and manual immunoassays Urinary marker
Pyridinoline (PYD)
Mature type 1 and 11 collagen
Cross link released when mature collagen type 1 and 11 breaks down Mechanically stabilise the molecule
Reflect degradation of mature collagen only Independent of dietary sources [20]
Non-specific 24 hr collection – hard to collect 2nd morning void with creatinine correction – additional analytical variability Circadian variation [20] Influenced by liver function [20]
Automated and manual immunoassays Urinary marker
Resorption markers Osteoclastic Enzymes
Tartrate Resistant Acid Phosphatase –isoform 5b (TRAP5b)
Isoform of acid phosphatase, resistant to tartrate, cleaved by proteases into 5b, present in ruffled border of osteoclasts
Cleaves type 1 collagen into fragments
Characteristic of osteoclastic activity [14]
Unstable at room temperature [22, 23] Circadian variability ↑ immediately after exercise [29]
Automated and manual immunoassays Serum
Cathepsin K
Cysteine protease present in ruffled border of actively resorbing osteoclasts
Cleaves telopeptide and helical regions of type 1 collagen
Specific biomarker of osteoclastic activity [14]
Unstable at room temp Clinical validity needs further investigation
Manual immunoassay Serum, EDTA plasma
Osteocyte activity markers
Receptor Activator of Nuclear factor Kappa B Ligand (RANKL)
Produced by osteoblasts, activated by B and T cells
Binds to RANK, which is expressed on osteoclasts and their precursors, stimulating their differentiation and activity
Novel biomarker Provide safety, efficacy and pharmacokinetics data to confirm drug mechanisms and mode of action for future use
Analytical problems Can measure free or OPG-bound [30] Circulating levels may not reflect bone microenvironment [31] Affected by thyroid function [32] Research method only Clinical and analytical validity needs further investigation
Manual research –grade immunoassay Total or soluble forms in serum
Osteoprotegerin (OPG)
Secreted by osteoblasts
Decoy receptor to RANKL reduces bone resorption by binding to RANK and preventing osteoclastogenesis
Novel biomarker Provide safety, efficacy and pharmacokinetics data to confirm drug mechanisms and mode of action for future use
Affected by thyroid function [32] Research method only Clinical and analytical validity needs further investigation
Manual research-grade immunoassay Serum
Dickkopf-related protein 1 (DKK1)
Produced by osteocytes
Inhibition of Wnt signalling pathway through binding to LRP5/6, blocking the Wnt effects on osteoblasts and decreasing bone formation
Key role in regulation of bone turnover
Research method only Clinical and analytical validity needs further investigation
Manual research –grade immunoassay Serum
Sclerostin (SCL)
Secreted by osteocytes
Inhibition of Wnt signalling pathway through binding to LRP5/6, blocking the Wnt effects on osteoblasts and decreasing bone formation
Significant ↓ with PTH therapy [33]
Research method only Affected by immobility [34] ↑ in type 1 and 2 diabetes [35, 36] Clinical and analytical validity needs further investigation
Manual research-grade immunoassaySerum
*P1NP and CTX (highlighted in bold) are the markers of choice, recommended by the IOF, IFCC (2011) and NBHA (2012).
ALP alkaline phosphatase, BSAP bone specific alkaline phosphatase, CTX carboxy-terminal cross-linked telopeptides of type 1 collagen, DKK-1 dickkopf-related protein 1, DPD deoxypyridinoline, EDTA ethylenediaminetetraacetic acid, HELP Type 1 collagen alpha 1 helicoidal peptide, ICTP carboxy-terminal cross-linked telopeptide of type 1 collagen, IGF-1 insulin-like growth factor-1, LRP low-density lipoprotein receptor-related protein, MMP matrix metalloproteinases, NTX amino-terminal cross-linked telopeptide of type 1 collagen, OPG osteoprotegerin, OC osteocalcin, PICP procollagen type 1 carboxy-terminal propeptide, PINP procollagen type 1 amino-terminal propeptide, PYD pyridinoline, RANK receptor activator of nuclear factor kappa B, RANKL receptor activator of nuclear factor kappa B ligand, SCL sclerostin, TRAP tartrate resistant acid phosphatase.

Markers of bone formation

Markers of bone formation are either by-products of active osteoblasts expressed during the various phases of their development or osteoblastic enzymes. The most widely used markers of bone formation are measured in serum or plasma and include: bone specific alkaline phosphatase (BSAP), osteocalcin and the carboxy- and amino-terminal propeptides of type 1 collagen (P1CP, P1NP). P1NP has several functional advantages and has been recommended by the Bone Marker Standards Working Group; it has low inter-individual variability [20] and is relatively stable in serum at room temperature [21]. P1NP is cleared by liver endothelial cells via a macrophage receptor species, the scavenger receptor, that recognises and endocytoses modified proteins [37]. P1NP is released as a trimeric structure, but is rapidly broken down to a monomeric form by thermal degradation [38]. Current immunoassays detect either the trimeric ‘intact’ molecule (automated IDS iSYS assay) or can measure both fractions and are thus called ‘total’ P1NP assays (automated Roche Elecsys assay).

Markers of bone resorption

The majority of bone resorption markers are degradation products of bone collagen, the exception being tartrate-resistant acid phosphatase (TRAP5b). Earlier research into bone metabolism relied primarily on urinary markers such as pyridinoline (PYD) and deoxypyridinoline (DPD), which were time-consuming and cumbersome and relied on complete twenty-four hour urine collections or second morning void/ creatinine ratios, increasing the imprecision of the measurement. However, now that serum/ plasma markers are available these have become the preferred means of measuring resorption. Examples include carboxy-terminal and amino-terminal cross-linked telopeptide of type 1 collagen (CTX and NTX respectively), of which CTX is considered the marker of choice [20]. CTX is generated by cathepsin K activity, the CTX epitope contains an aspartyl-glycine motif that is susceptible to spontaneous isomerisation and racemisation generating four isoforms [17]; the α-aspartic acid converts to the β-form as the bone ages. Two automated immunoassays are available that target βCTX indicative of the breakdown of mature type 1 collagen (IDS iSYS and Roche Elecsys). The major disadvantage of CTX is its large circadian variation necessitating a morning fasting sample for accurate interpretation [25]. The choice of marker in clinical practice needs to be made on pragmatic grounds. Urine NTX may be the preferred marker in the clinic setting as unlike plasma CTX, it is not as sensitive to circadian changes and is not affected by food intake, it also avoids the invasive venepuncture associated with a blood sample and may be preferred by patients [39]. However the various drugs licensed for the treatment of osteoporosis have a differing spectrum of effects on BTMs and not all markers respond by the same amount for a given degree of bone resorption. Amongst the bone resorption markers, plasma CTX tends to change more than urine NTX which tends to change more than TRAP5b [20].

Markers of osteoclastogenesis

Osteoclast regulatory proteins are commonly measured in research, but have yet to find a niche clinically. The discovery of the OPG/RANK/RANKL system has clarified a major component of the bone remodelling cycle. RANKL is expressed in vivo in either membrane-bound or soluble form (sRANKL) and is also present in serum as a free or OPG-bound molecule, as a consequence design differences between immunoassays have created difficulties in comparing research and interpreting clinical data [30]. Furthermore circulating levels may not reflect the bone microenvironment [31]. Research into the relationship between circulating levels of OPG and sRANKL to BMD in postmenopausal osteoporosis are controversial, some studies reporting an inverse relationship [40], while others have found no association [41]. Rigorous testing of commercial assays and identification of the sources of variability are required before they can be adapted to routine clinical practice.

Osteocyte markers

Over the last decade research has focused mainly on the role of osteoclasts and osteoblasts in osteoporosis, more recently however, osteocytes have been found to play a key role in the regulation of bone turnover. Osteocytes are fully differentiated osteoblasts and lie in lacunae in the mineralized matrix and osteoid tissue of bone [42]. Osteocytes are able to detect changes in bone morphology, particularly micro-fractures through their sensitivity to mechanical forces, acting like bone mechanoreceptors [43]. They regulate bone turnover both through direct physical contact with other bone cells and by producing various factors which affect bone formation and can be measured in blood such as, sclerostin (SCL), dickkopf-related protein 1 (DKK1), dentin matrix protein 1 (DMP1) and matrix extracellular phosphoglycoprotein (MEPE).
DKK1 and SCL are secreted osteocyte markers acting as inhibitors to the Wnt signalling pathway through binding to low density lipoprotein receptor-related protein 5 and 6 (LRP5/6) and hence blocking the Wnt effects on osteoblasts decreasing bone formation (Figure 2) [44, 45]. In vivo studies have shown that osteocyte depletion results in profound loss of trabecular bone mass [4648] and suggest a close interaction between osteocytes and other bone cells, highlighting their role in the regulation of both bone formation and resorption.
Although widely used in research, their diagnostic importance remains to be validated due in part to their analytical and biological variability. In healthy adults, SCL levels correlate positively with age, BMI, and bone mineral content and negatively with osteocalcin and calcium [49]. SCL is increased in type 2 diabetes. Moreover, the transcriptional suppression of SCL production by PTH might be impaired in type 1 and type 2 diabetes [35]. SCL levels are significantly lower in osteoporotic compared to non-osteoporotic patients with type 2 diabetes [36]. The Wnt signalling pathway has recently been identified as central to the development of disuse osteoporosis [50]. Mechanical unloading in long-term immobilized patients causes up regulation of SCL and therefore inhibits bone formation via suppressed osteoblast activity and survival [34]. Circulating SCL reflects the severity of bone loss and is a candidate biomarker of osteoporosis severity in chronic spinal cord injury [51]. Higher serum SCL levels are associated with a greater risk of hip fractures in older women. In addition, the risk of hip fracture is amplified when high SCL levels are combined with lower BMD [52]. Serum SCL levels are regulated by both estrogens and PTH in postmenopausal women [53]. Serum SCL is decreased in women with postmenopausal osteoporosis compared with non-osteoporotic early postmenopausal women and positively correlated to lumbar spine BMD. Furthermore, levels are increased after 6 months treatment with risedronate, but remain essentially unchanged after 6 months teriparatide treatment [54]. However, serum or plasma SCL concentrations should be interpreted with caution as current assays produce very different results. Standardization of sclerostin assays is necessary before being introduced into general clinical laboratory use [55].

Variability in markers of bone turnover

An understanding of the source and magnitude of the absolute inter and intra-person variability, including biological, pre-analytic and analytical variation, of each marker is necessary to interpret serial measurements and individualise treatment.

Biological variability

Intra-individual variation
Bone turnover shows a circadian rhythm, this is more obvious in the serum and urinary markers of bone resorption. βCTX for example is highest between 01:30 and 04:30 hours and may be more than twice that at the nadir between 11:00 and 15:00 hours [27], this may be attenuated by several factors such as; age, gender, ethnicity, menopausal status, osteoporotic stage and anti-resorptive agents or calcium supplementation [19], but the disparity is diminished with fasting [25]. All bone markers are significantly lower in the fed state with the exception of BSAP, this may be due to several factors including the clearance rate of individual markers or food composition [16] and may be partly explained by variation in serum insulin [25]. Osteocalcin and P1CP follow the same diurnal pattern but show only twenty percent difference and BSAP has two peaks at 14:00 and 23:30 hours with a nadir thirty percent reduced at 06:30 [19]. Therefore timing of the sample collection and fasting status should be tightly controlled.
The existence of intra-individual low-frequency biological rhythms, imply that biomarkers can also vary between consecutive days, this is more noticeable in the urinary resorption markers [14]. There is a degree of controversy regarding seasonal variation with some researchers suggesting that overall seasonal changes are insignificant [56], whilst others have found a substantial wintertime increase [57], which may be due in part to reduced levels of vitamin D. Physical activity is also significant, TRAP and to a lesser extent BSAP and CTX are reduced immediately after plyometrics, but return to pre concentrations within two hours. Interestingly similar changes were found in PTH [29]. Details of exercise in the previous twenty-four hours should therefore be recorded.
Bone turnover varies with the menstrual cycle, research suggests that osteoblastic activity is higher during the luteal period [58] and bone resorption is increased during the follicular phase [59]. Pregnancy affects all BTMs due in part to the calcium requirements of the foetus, but also to changes in maternal glomerular filtration rate (GFR) affecting renal clearance. However the time change is contentious, one study following ten women at regular intervals reported an increase in urinary resorption markers throughout pregnancy with a significant increase in bone formation in the third trimester [60]. A more recent study measured serum OPG, RANKL, osteocalcin and CTX in twenty six different women at each trimester. The study found increased bone formation in the first trimester and increased resorption in the second which surprisingly decreased again in the third trimester [61]. Postpartum, levels gradually start to decrease but may still be higher than pre-pregnancy levels for up to a year [62].
A comprehensive drug history should also be taken into account when interpreting bone marker results. Anti-resorptive drugs such as bisphosphonates [26] and hormone replacement therapy (HRT) [63] have a major effect on markers of bone resorption and long-term corticosteroid therapy is known to suppress bone formation [64].
Inflammatory conditions are major precipitators for bone loss, especially rheumatoid arthritis (RA) which is further aggravated by decreased functional activity and the use of glucocorticoids [65]. In a prior study, we found that B-cell depletion increases bone formation and decreases bone resorption in RA patients. This may be a direct effect on osteoblasts and osteoclasts respectively and be at least partially explained by the decreased inflammation and disease activity [66]. In diabetes serum osteocalcin is negatively correlated with glucose levels and advanced glycation end products (AGEs) are known to have a negative impact on bone [67]. Thyroid disorders such as thyrotoxicosis are well known to affect bone turnover. Thyroid stimulating hormone (TSH) receptors are present in both osteoblasts and osteoclasts and the low TSH levels observed in thyroidectomised patients on L-thyroxine are associated with an increase in OPG and decrease in RANKL and are significantly correlated with vertebral fractures [32]. Bone markers are cleared through the liver or kidneys and are also influenced by diseases affecting these systems, decreased GFR for example will decrease the urinary excretion of CTX and therefore increase serum levels. They are also affected by any disease states leading to increased periods of bed rest and immobility. Research has shown that microgravity induces significant and progressive bone loss, a consequence of increased bone resorption and retardation of bone formation [68]. Certainly levels of all bone markers increase significantly in the first few weeks after fracture and may remain elevated for up to a year. The rate of increase is dependent on the location, severity and size of the fracture and the age of the patient. BTM’s can be elevated for up to 6 months after minor fractures e.g. forearm fractures but up to one year after a hip fracture and needs to be taken into consideration when measuring them [19, 69]. However, they fall gradually over time and using a reduction of 50% in bone resorption when using anti-resorptives as a good indicator of response would be greater than any reduction that might otherwise occur.
In light of the above evidence it can be seen that to use bone turnover to monitor change can be quite difficult. In order to minimise problems it is best to measure the BTMs in as similar a set of circumstances as possible. Particular attention should be paid to the time of day and hence research studies tend to use early morning fasted samples. One way in which to help overcome within person variability in serial measurements and to monitor therapy is to use the ‘least significant change’ (LSC) model [70]. LSC at a significance level of p=<0.05 is defined as 1.96*√2*√ (CV12+CVA2); where CV1 is the within-subject coefficient of variation and CVA is the total analytical imprecision. LSC identifies the true physiological change in the marker. In general a change of more than twenty percent is considered significant for formation markers [71], similarly between twenty-seven to thirty-six percent is significant for markers of bone resorption [72].
Inter-individual variation
Between person variability is much harder to control, e.g. age, gender, and menopausal status, but is equally important to validate results. Bone metabolism rates are higher in infants up to three years of age, they are relatively stable throughout adolescence but sex-specific increases in bone marker levels are evident during the pubertal growth spurt and are reportedly influenced by pubertal stage rather than age [73]. BTMs are higher in men between twenty and thirty years of age then reach their lowest levels during their fifties [14], whereas in females there is a substantial increase in bone turnover corresponding to oestrogen deficiency during the menopause [28]. We checked our local population and found a trend towards higher bone turnover in males during their third decade whilst reaching peak bone mass, although there seemed to be little evidence of any age related change in women possibly due to the lower numbers of postmenopausal females in our cohort (Figure 3). It is important for each laboratory to investigate the transferability of the quoted reference intervals to its own patient population based on equivalent standardised collection conditions. The widespread availability of automated immunoassays now means that harmonisation of method specific reference ranges is possible and studies from well-characterised populations have reported robust BTM ranges in large well defined cohorts [74]. In contrast to the use of reference ranges some researchers have suggested combining a marker of formation and resorption to gain a direct insight into the changes in the balance and rate of bone turnover in relation to a reference value [75], leading factors in estimating fracture risk and prognosis. Given the large observed differences observed between genders, different ages and developmental stages means that care must be taken when comparing populations and in the design of research studies.

Analytical variability

Technical variation
Over the last decade many of the traditional BTM immunoassays have been automated, improving technical performance and increasing their availability. Nevertheless, analytical aspects such as within and between batch precision, accuracy and standardisation, remain problematic. Inter-laboratory variation is also crucial; a European study in 2001, measuring pooled samples of serum and urine in seventy-three laboratories concluded that even with identical assays results for the majority of the markers were significantly different [76]. Similarly an American study in 2010 comparing six commercial laboratories over an eight month period concluded that reproducibility varied substantially for urine NTX and serum BSAP [77]. Moreover there is an extensive list of bone markers being offered making it very difficult to compare research evidence. Consequently, the International Osteoporosis Foundation (IOF), the International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) [20] and the National Bone Health Alliance (NBHA) [78], have recommended that a marker of bone formation and resorption, namely P1NP and CTX, are used as reference analytes in clinical studies. They go on to stipulate that these markers should be measured by standardised assays to minimise immunochemical heterogeneity and recommend that manufacturers adopt international reference standards and minimise batch to batch variability [20].
Sample stability
Appropriate control of sample collection and preparation is vital for successful BTM measurement. Several BTMs, especially osteocalcin and TRAP5b, are sensitive to thermo degradation and levels can be significantly reduced after only a few hours at room temperature [22, 23]. TRAP5b activity is also reduced during storage, samples must be kept at −70°C or lower and multiple freeze-thaw cycles should be avoided. No significant decrease has been detected in CTX stored at −20°C or lower for up to three years, nevertheless it rapidly decreases in serum at both 4°C and 37°C. The molecular mechanism is unknown but this decrease is minimised by ethylenediaminetetraacetic acid (EDTA) [18]. CTX is reportedly stable in EDTA blood tubes before separation for up to forty-eight hours, likewise osteocalcin becomes stable for up to eight hours at room temperature [21]. Consequently blood should be collected into EDTA tubes and separated as soon as possible, if samples are not analysed immediately they should be stored at −20°C or lower. Both P1NP and BSAP were found to be stable in any sample type [21]. Notably current TRAP5b assays are not affected by haemolysis, but erythrocytes are known to contain proteases which degrade osteocalcin. Grossly haemolysed samples in general should always be avoided.

Clinical usefulness of bone turnover markers in osteoporosis

BTMs are frequently used in clinical trials and provide valuable information on the efficacy of osteoporotic treatments, but their predictive value is limited by their large biological variation and diagnostically they are less often used for individualized patient care. Other routine laboratory investigations are frequently used to identify or exclude secondary causes of osteoporosis such as hyperparathyroidism, vitamin D status, thyrotoxicosis and hypogonadism [79].

Diagnostic

Currently the WHO recommends the use of BMD of the spine and proximal femur measured by DXA as the gold standard to diagnose osteoporosis and its severity [12]. Although BMD has methodological limitations especially in the elderly due in part to degenerative changes in the lumbar spine [80], BTMs alone would not be suitable to estimate bone loss.

Prediction of bone loss

Women generally lose about one to two percent of their bone per year after the menopause, however thirty percent lose bone at a faster rate [28]. Longitudinal studies of post-menopausal women have demonstrated two characteristic groups; high bone turnover and normal or low bone turnover. Serial BTM measurements are effective in identifying those women who lose bone most rapidly, this is important because this group respond more readily to anti-resorptive medication [81]. Furthermore a meta-analysis of eighty-five studies reported a significant correlation between serial levels of BTMs and BMD measurements during bisphosphonate treatment [82], the association becomes stronger with advancing age [83]. However, BTMs should only be used to supplement BMD not measured in isolation.

Prediction of fracture risk

BMD is widely used to predict osteoporotic fractures but approximately thirty to fifty percent of patients with fragility fractures have T-scores above the osteoporotic threshold [84]. There is evidence that high bone turnover, as measured by a single or combination of BTMs, is associated with an increased fracture risk [28], but their use alone to predict fracture has yet to be established. Two clinical risk assessment algorithms have been validated for use in the UK to predict fracture incidence over ten years [85], namely FRAX and QFracture, currently they do not include all risk factors. BTMs have not been included because of their inconsistency in research studies so far. There is a need for studies confirming whether the addition of BTMs to FRAX would result in increased sensitivity and specificity.

Treatment selection and monitoring

BMD and BTMs are independent predictors of fracture risk, recent evidence does not support the use of BTMs to select the optimal treatment, but BTMs can be used to monitor treatment efficacy before BMD changes can be evaluated. Additionally early changes in BTMs can be used to measure the clinical efficacy of an anti-resorptive treatment and to reinforce patient compliance [26]. The effectiveness of osteoporotic therapy can be assessed by serial BMD measurements usually by DXA, but quantifiable changes in bone mass are small and are only apparent after twelve to twenty-four months, furthermore they only measure net balance in a very small portion of the skeleton. DXA reproducibility is also affected by machine and operator error plus patient variability (weight or degenerative changes) [86]. The minimum acceptable precision for an individual technician is 1.9% (LSC 5.3%) at the lumbar spine and 1.8% (LSC 5.0%) at the total hip. Intervals between measurements depend on the patient’s clinical status, but given the need to exceed the LSC and the relatively modest changes in BMD observed with most treatments it is generally going to be a minimum of two years before a significant change can be observed. Indeed, there are trends for a variety of reasons towards less frequent measurement of BMD to three or even five year intervals [87].
Meta-regression analysis has found no evidence of a relationship between BMD changes and reduction in risk of fractures among patients receiving calcium with or without vitamin D supplementation. Calcium and/or Vitamin D may reduce fracture rates through a mechanism independent of bone density [13]. BTMs on the other hand show a substantial and more immediate global effect, they measure both bone formation and resorption rate and can classify patients into low or high remodelling groups. Osteoporosis treatments such as bisphosphonates, strontium ranelate, denosumab, hormone replacement therapy (HRT) and selective estrogen receptor moderators (SERMs) act by reducing BTM levels by forty to sixty percent within three to six months [88]. Thus one use of BTMs is to give an early indication of the success of the treatment. Baseline measurements can be repeated at the next follow up appointment say three to six months later. A significant change in BTMs as assessed by the LSC method can then be used to judge success of the treatment and will hopefully be reflected by an increase in BMD in the fullness of time. In the meantime the change in BTM supplies reassurance to the clinician and can be used to encourage the patient. Unfortunately, as BTMs are highly variable this is at best only an indication.
There has been considerable discussion about how long to treat with bisphosphonates, because these drugs accumulate in the skeleton, leading to a reservoir that continues to be released for months or years after treatment has stopped. These medications also result in a low bone turnover state over time with both resorption and formation reduced. This combined with concerns over microfracture, the possibility that they may prevent bone healing and the association with atypical femoral shaft fractures has led to the belief that it may not be wise to continue these medications indefinitely. It is generally accepted that the need to continue bisphosphonates be reviewed after 5 years and kept under review until ten years of treatment. Depending on the individual circumstances a decision to stop treatment, give a drug holiday or change treatment may be made. If a drug holiday is decided upon then BTMs could be checked at regular intervals, e.g. annually. Once these are rising again and especially on return to pre-treatment levels therapy could be restarted. Such an approach may be particularly useful with longer acting agents such as zoledronic acid [89]. The BTMs should be used in conjunction with the clinical circumstances and with repeated BMD after appropriate time intervals.
More recently anabolic agents such as PTH, e.g. teriparatide, have become available which stimulate osteoblastic activity. Markers of bone formation increase early after the initiation of teriparatide therapy with a delayed, but significant, increase in resorption markers [90]. It has been proposed clinically to measure P1NP at baseline and three months post treatment a positive response is defined as a change of greater than 10 μg/L [91].

Conclusions

During the last decade significant advances have been made in the identification and characterisation of specific BTMs for use in clinical drug trials and to aid in the therapeutic management of osteoporosis. Technological developments have greatly enhanced assay performance producing reliable, rapid, non-invasive cost effective assays with improved sensitivity and specificity. We now have a greater understanding of the need to regulate pre-analytical sample collection to minimise the effects of biological variation. The use of BTMs to select those at risk of fractures is not routinely recommended partly due to their degree of variability. However, baseline measurements of resorption markers are useful before commencement of anti-resorptive treatment e.g. bisphosphonates or denosumab and can be checked 3–6 months later to check response and adherence to treatment. Similarly a formation marker such as P1NP can be used to monitor bone forming agents such as PTH analogues. BTMs may also be useful when monitoring patients during treatment holidays and aid in the decision as to when therapy should be recommenced. The recent recommendations by the Bone Marker Standards Working Group aim to standardise research and include a marker of bone resorption (CTX) and formation (P1NP) in all future studies. They anticipate that manufacturers will calibrate their assays in future using an international reference standard to establish robust reference ranges. It is hoped that improved research in turn will lead to optimise markers for the clinical management of osteoporosis and other bone diseases. The biochemical assessment, utilizing BSAP, is now the mainstay of the diagnosis and management of metabolic bone disease in chronic kidney disease.

Acknowledgements

The authors wish to thank staff from the Biochemistry laboratory at the James Cook University Hospital in Middlesbrough who made the reference range study possible. In particular we acknowledge Cheryl Goodrum for analysing the samples.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

JMvL has received a research grant, consultancy and speaker fees from Roche. SPT has received speaker fees from Ely Lilly.

Author’ contributions

GW drafted the manuscript. ME revised the manuscript. SPT and HKD revised and critically appraised the manuscript. JMvL revised, critically appraised and provided overall supervision for the project. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Datta HK, Ng FW, Walker JA, Tuck SP, Varanasi SS: The cell biology of bone metabolism-a review. J Clin Path. 2008, 61: 577-587. 10.1136/jcp.2007.048868.CrossRefPubMed Datta HK, Ng FW, Walker JA, Tuck SP, Varanasi SS: The cell biology of bone metabolism-a review. J Clin Path. 2008, 61: 577-587. 10.1136/jcp.2007.048868.CrossRefPubMed
2.
Zurück zum Zitat Carey JJ, Licata AA, Delaney MF: Biochemical markers of bone turnover. Clin Rev Bone Miner Metab. 2006, 4 (3): 197-212. 10.1385/BMM:4:3:197.CrossRef Carey JJ, Licata AA, Delaney MF: Biochemical markers of bone turnover. Clin Rev Bone Miner Metab. 2006, 4 (3): 197-212. 10.1385/BMM:4:3:197.CrossRef
3.
Zurück zum Zitat Vega D, Maalouf NM, Sakhaee K: The role of receptor activator of nuclear factor-κB (RANK)/RANK Ligand/Osteoprotegerin: clinical implications. J Clin Endocrinol Metab. 2007, 92 (12): 4514-4521. 10.1210/jc.2007-0646.CrossRefPubMed Vega D, Maalouf NM, Sakhaee K: The role of receptor activator of nuclear factor-κB (RANK)/RANK Ligand/Osteoprotegerin: clinical implications. J Clin Endocrinol Metab. 2007, 92 (12): 4514-4521. 10.1210/jc.2007-0646.CrossRefPubMed
4.
Zurück zum Zitat Leibbrandt A, Penninger JM: RANK/RANKL: regulators of immune responses and bone physiology. Ann N Y Acad Sci. 2008, 1143: 123-150. 10.1196/annals.1443.016.CrossRefPubMed Leibbrandt A, Penninger JM: RANK/RANKL: regulators of immune responses and bone physiology. Ann N Y Acad Sci. 2008, 1143: 123-150. 10.1196/annals.1443.016.CrossRefPubMed
5.
Zurück zum Zitat Klein-Nulend J, Bakker AD, Bacabac RG, Vatsa A, Weinbaum S: Mechanosensation and transduction in osteocytes. Bone. 2013, 54 (2): 182-190. 10.1016/j.bone.2012.10.013.CrossRefPubMed Klein-Nulend J, Bakker AD, Bacabac RG, Vatsa A, Weinbaum S: Mechanosensation and transduction in osteocytes. Bone. 2013, 54 (2): 182-190. 10.1016/j.bone.2012.10.013.CrossRefPubMed
6.
Zurück zum Zitat Kobayashi Y, Maeda K, Takahashi N: Roles of Wnt signalling in bone formation and resorption. Jpn Dent Sci Rev. 2008, 44: 76-82. 10.1016/j.jdsr.2007.11.002.CrossRef Kobayashi Y, Maeda K, Takahashi N: Roles of Wnt signalling in bone formation and resorption. Jpn Dent Sci Rev. 2008, 44: 76-82. 10.1016/j.jdsr.2007.11.002.CrossRef
10.
Zurück zum Zitat Bongartz TA, Scholmerich J, Straub RH: From Osteoporosis in postmenopausal women. Bone disease in rheumatology. Edited by: Maricic M, Gluck OS. 2005, Arizona: Lippincott Williams and Wilkens, 155-156. Bongartz TA, Scholmerich J, Straub RH: From Osteoporosis in postmenopausal women. Bone disease in rheumatology. Edited by: Maricic M, Gluck OS. 2005, Arizona: Lippincott Williams and Wilkens, 155-156.
11.
Zurück zum Zitat Bauer D, Garnero P, Harison SL, Cauley JA, Eastell R, Ensrud KE, Orwoll E: Biochemical markers on bone turnover, hip loss and fracture in older men: the MrOS study. J Bone Miner Res. 2009, 24 (12): 2032-2038. 10.1359/jbmr.090526.PubMedCentralCrossRefPubMed Bauer D, Garnero P, Harison SL, Cauley JA, Eastell R, Ensrud KE, Orwoll E: Biochemical markers on bone turnover, hip loss and fracture in older men: the MrOS study. J Bone Miner Res. 2009, 24 (12): 2032-2038. 10.1359/jbmr.090526.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Kanis JA, McCloskey EV, Johansson H, Oden A, Melton LJ, Khaltaev N: A reference standard for the description of osteoporosis. Bone. 2008, 42: 467-475. 10.1016/j.bone.2007.11.001.CrossRefPubMed Kanis JA, McCloskey EV, Johansson H, Oden A, Melton LJ, Khaltaev N: A reference standard for the description of osteoporosis. Bone. 2008, 42: 467-475. 10.1016/j.bone.2007.11.001.CrossRefPubMed
13.
Zurück zum Zitat Rabinda V, Bruyère O, Reginster JY: Relationship between bone mineral density changes and risk of fractures among patients receiving calcium with or without vitamin D supplementation: a meta-regression. Osteoporos Int. 2011, 22: 893-901. 10.1007/s00198-010-1469-x.CrossRef Rabinda V, Bruyère O, Reginster JY: Relationship between bone mineral density changes and risk of fractures among patients receiving calcium with or without vitamin D supplementation: a meta-regression. Osteoporos Int. 2011, 22: 893-901. 10.1007/s00198-010-1469-x.CrossRef
14.
Zurück zum Zitat Seibel MJ: Biochemical markers of bone turnover: part 1: biochemistry and variability. Clin Biochem Rev. 2005, 26: 97-122.PubMedCentralPubMed Seibel MJ: Biochemical markers of bone turnover: part 1: biochemistry and variability. Clin Biochem Rev. 2005, 26: 97-122.PubMedCentralPubMed
15.
Zurück zum Zitat Brown JP, Albert C, Nassar BA, Adachi JD, Cole D, Davison KS, Dooley KC, Don-Wauchope A, Douville P, Hanley DA, Jamal SA, Josse R, Kaiser S, Krahn J, Krause R, Kremer R, Lepage R, Letendre E, Morin S, Ooi DS, Papaioaonnou A, Ste-Marie L-G: Bone turnover markers in the management of osteoporosis. Clin Biochem. 2009, 42: 929-942. 10.1016/j.clinbiochem.2009.04.001.CrossRefPubMed Brown JP, Albert C, Nassar BA, Adachi JD, Cole D, Davison KS, Dooley KC, Don-Wauchope A, Douville P, Hanley DA, Jamal SA, Josse R, Kaiser S, Krahn J, Krause R, Kremer R, Lepage R, Letendre E, Morin S, Ooi DS, Papaioaonnou A, Ste-Marie L-G: Bone turnover markers in the management of osteoporosis. Clin Biochem. 2009, 42: 929-942. 10.1016/j.clinbiochem.2009.04.001.CrossRefPubMed
16.
Zurück zum Zitat Clowes JA, Hannon RA, Yap TS, Hoyle NR, Blumsohn A, Eastell R: Effect of feeding on bone turnover markers and its impact on biological variability of measurements. Bone. 2002, 30 (6): 886-890. 10.1016/S8756-3282(02)00728-7.CrossRefPubMed Clowes JA, Hannon RA, Yap TS, Hoyle NR, Blumsohn A, Eastell R: Effect of feeding on bone turnover markers and its impact on biological variability of measurements. Bone. 2002, 30 (6): 886-890. 10.1016/S8756-3282(02)00728-7.CrossRefPubMed
17.
Zurück zum Zitat Swaminathan R: Biochemical markers of bone turnover. Clin Chim Acta. 2001, 313: 95-105. 10.1016/S0009-8981(01)00656-8.CrossRefPubMed Swaminathan R: Biochemical markers of bone turnover. Clin Chim Acta. 2001, 313: 95-105. 10.1016/S0009-8981(01)00656-8.CrossRefPubMed
18.
Zurück zum Zitat Qvist P, Munk M, Hoyle N, Christiansen C: Serum and plasma fragments of C-telopeptides of type I collagen (CTX) are stable during storage at low temperatures for 3 years. Clin Chim Acta. 2004, 350 (1–2): 167-173.CrossRefPubMed Qvist P, Munk M, Hoyle N, Christiansen C: Serum and plasma fragments of C-telopeptides of type I collagen (CTX) are stable during storage at low temperatures for 3 years. Clin Chim Acta. 2004, 350 (1–2): 167-173.CrossRefPubMed
19.
Zurück zum Zitat Hannon R, Eastell R: Preanalytical variability of biochemical markers of bone turnover. Osteoporos Int. 2000, 11 (Suppl 6): S30-44.CrossRefPubMed Hannon R, Eastell R: Preanalytical variability of biochemical markers of bone turnover. Osteoporos Int. 2000, 11 (Suppl 6): S30-44.CrossRefPubMed
20.
Zurück zum Zitat Vasikaran S, Eastell R, Bruyère O, Foldes AJ, Garnero P, Griesmacher A, McClung M, Morris HA, Silverman S, Trenti T, Wahl DA, Cooper C, Kanis JA, for the IOF-IFCC Bone Marker Standards Working Group: 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: 391-420. 10.1007/s00198-010-1501-1.CrossRefPubMed Vasikaran S, Eastell R, Bruyère O, Foldes AJ, Garnero P, Griesmacher A, McClung M, Morris HA, Silverman S, Trenti T, Wahl DA, Cooper C, Kanis JA, for the IOF-IFCC Bone Marker Standards Working Group: 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: 391-420. 10.1007/s00198-010-1501-1.CrossRefPubMed
21.
Zurück zum Zitat Stokes FJ, Ivanov P, Bailey LM, Fraser WD: The effects of sampling procedures and storage conditions on short-term stability of blood-based biochemical markers of bone metabolism. Clin Chem. 2011, 57 (1): 138-140. 10.1373/clinchem.2010.157289.CrossRefPubMed Stokes FJ, Ivanov P, Bailey LM, Fraser WD: The effects of sampling procedures and storage conditions on short-term stability of blood-based biochemical markers of bone metabolism. Clin Chem. 2011, 57 (1): 138-140. 10.1373/clinchem.2010.157289.CrossRefPubMed
22.
Zurück zum Zitat Blumsohn A, Hannon RA, Eastell R: Apparent instability of osteocalcin in serum as measured with different commercially available immunoassays. Clin Chem. 1995, 41: 318-319.PubMed Blumsohn A, Hannon RA, Eastell R: Apparent instability of osteocalcin in serum as measured with different commercially available immunoassays. Clin Chem. 1995, 41: 318-319.PubMed
23.
Zurück zum Zitat Halleen JM, Alatalo SL, Suominen H, Cheng S, Janckila AJ, Väänänen HK: Tartrate-resistant acid phosphatase 5b: a novel serum marker of bone resorption. J Bone Miner Res. 2000, 15 (7): 1337-1345. 10.1359/jbmr.2000.15.7.1337.CrossRefPubMed Halleen JM, Alatalo SL, Suominen H, Cheng S, Janckila AJ, Väänänen HK: Tartrate-resistant acid phosphatase 5b: a novel serum marker of bone resorption. J Bone Miner Res. 2000, 15 (7): 1337-1345. 10.1359/jbmr.2000.15.7.1337.CrossRefPubMed
24.
Zurück zum Zitat Marin L, Koivula M-K, Jukkola-Vuorinen A, Leino A, Risteli J: Comparison of total and intact aminoterminal propeptide of type 1 procollagen assays in patients with breast cancer with or without bone metastases. Ann Clin Biochem. 2011, 48: 447-451. 10.1258/acb.2011.011040.CrossRefPubMed Marin L, Koivula M-K, Jukkola-Vuorinen A, Leino A, Risteli J: Comparison of total and intact aminoterminal propeptide of type 1 procollagen assays in patients with breast cancer with or without bone metastases. Ann Clin Biochem. 2011, 48: 447-451. 10.1258/acb.2011.011040.CrossRefPubMed
25.
Zurück zum Zitat Bjarnason NH, Henriksen EEG, Alexandersen P, Christgau S, Henriksen DB, Christiansen C: Mechanism of circadian variation in bone resorption. Bone. 2002, 30: 307-313. 10.1016/S8756-3282(01)00662-7.CrossRefPubMed Bjarnason NH, Henriksen EEG, Alexandersen P, Christgau S, Henriksen DB, Christiansen C: Mechanism of circadian variation in bone resorption. Bone. 2002, 30: 307-313. 10.1016/S8756-3282(01)00662-7.CrossRefPubMed
26.
Zurück zum Zitat Bergmann P, Body JJ, Boonen S, Boutsen Y, Devogelaer JP, Goemaere S, Kaufman JM, Reginster JY, Gangji V, Members of the Advisory Board on Bone Markers: Evidence-based guidelines for the use of biochemical markers of bone turnover in the selection and monitoring of bisphosphonate treatment in osteoporosis: a consensus document of the Belgian bone club. Int J Clin Pract. 2009, 63 (1): 19-26. 10.1111/j.1742-1241.2008.01911.x.PubMedCentralCrossRefPubMed Bergmann P, Body JJ, Boonen S, Boutsen Y, Devogelaer JP, Goemaere S, Kaufman JM, Reginster JY, Gangji V, Members of the Advisory Board on Bone Markers: Evidence-based guidelines for the use of biochemical markers of bone turnover in the selection and monitoring of bisphosphonate treatment in osteoporosis: a consensus document of the Belgian bone club. Int J Clin Pract. 2009, 63 (1): 19-26. 10.1111/j.1742-1241.2008.01911.x.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Wichers M, Schmidt E, Bidlingmaier F, Klingmüller D: Diurnal rhythm of cross laps in human serum. Clin Chem. 1999, 45: 1858-1860.PubMed Wichers M, Schmidt E, Bidlingmaier F, Klingmüller D: Diurnal rhythm of cross laps in human serum. Clin Chem. 1999, 45: 1858-1860.PubMed
28.
Zurück zum Zitat Garnero P, Sornay-Rendu E, Claustrat B, Delmas PD: Biochemical markers of bone turnover, endogenous hormones and the risk of fractures in postmenopausal women: the OFELY study. J Bone Miner Res. 2000, 15: 1526-1536. 10.1359/jbmr.2000.15.8.1526.CrossRefPubMed Garnero P, Sornay-Rendu E, Claustrat B, Delmas PD: Biochemical markers of bone turnover, endogenous hormones and the risk of fractures in postmenopausal women: the OFELY study. J Bone Miner Res. 2000, 15: 1526-1536. 10.1359/jbmr.2000.15.8.1526.CrossRefPubMed
29.
Zurück zum Zitat Rogers RS, Dawson AW, Wang Z, Thyfault JP, Hinton PS: Acute response of plasma markers of bone turnover to a single bout of resistance training or plyometrics. J Appl Physiol. 2011, 111: 1353-1360. 10.1152/japplphysiol.00333.2011.CrossRefPubMed Rogers RS, Dawson AW, Wang Z, Thyfault JP, Hinton PS: Acute response of plasma markers of bone turnover to a single bout of resistance training or plyometrics. J Appl Physiol. 2011, 111: 1353-1360. 10.1152/japplphysiol.00333.2011.CrossRefPubMed
30.
Zurück zum Zitat Bowsher RR, Sailstad JM: Insights in the application of research-grade diagnostic kits for biomarker assessments in support of clinical drug development: Bioanalysis of circulating concentrations of soluble receptor activator of nuclear factor κB ligand. J Pharm Biomed Anal. 2008, 48 (5): 1282-1289. 10.1016/j.jpba.2008.09.026.CrossRefPubMed Bowsher RR, Sailstad JM: Insights in the application of research-grade diagnostic kits for biomarker assessments in support of clinical drug development: Bioanalysis of circulating concentrations of soluble receptor activator of nuclear factor κB ligand. J Pharm Biomed Anal. 2008, 48 (5): 1282-1289. 10.1016/j.jpba.2008.09.026.CrossRefPubMed
31.
Zurück zum Zitat Kearns AE, Khosla S, Kostenuik PJ: Receptor activator of nuclear factor κB ligand and osteoprotegerin regulation of bone remodelling in health and disease. Endocr Rev. 2008, 29 (2): 155-192.PubMedCentralCrossRefPubMed Kearns AE, Khosla S, Kostenuik PJ: Receptor activator of nuclear factor κB ligand and osteoprotegerin regulation of bone remodelling in health and disease. Endocr Rev. 2008, 29 (2): 155-192.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Nicholls JJ, Brassill MJ, Williams GR, Duncan Bassett JH: The skeletal consequences of thyrotoxicosis. J Endocrinol. 2012, 213 (3): 209-221. 10.1530/JOE-12-0059.CrossRefPubMed Nicholls JJ, Brassill MJ, Williams GR, Duncan Bassett JH: The skeletal consequences of thyrotoxicosis. J Endocrinol. 2012, 213 (3): 209-221. 10.1530/JOE-12-0059.CrossRefPubMed
33.
Zurück zum Zitat Drake MT, Srinivasan B, Mödder UI, Peterson JM, McCready LK, Riggs BL, Dwyer D, Stolina M, Kostenuik P, Khosla S: Effects of parathyroid hormone treatment on circulating sclerostin levels in postmenopausal women. J Clin Endocrinol Metab. 2010, 95 (11): 5056-5062. 10.1210/jc.2010-0720.PubMedCentralCrossRefPubMed Drake MT, Srinivasan B, Mödder UI, Peterson JM, McCready LK, Riggs BL, Dwyer D, Stolina M, Kostenuik P, Khosla S: Effects of parathyroid hormone treatment on circulating sclerostin levels in postmenopausal women. J Clin Endocrinol Metab. 2010, 95 (11): 5056-5062. 10.1210/jc.2010-0720.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Gaudio A, Pennisi P, Bratengeier C, Torrisi V, Lindner B, Mangiafico RA, Pulvirenti I, Hawa G, Tringali G, Fiore CE: Increased sclerostin serum levels associated with bone formation and resorption markers in patients with immobilization-induced bone loss. J Clin Endocrinol Metab. 2010, 95 (5): 2248-2253. 10.1210/jc.2010-0067.CrossRefPubMed Gaudio A, Pennisi P, Bratengeier C, Torrisi V, Lindner B, Mangiafico RA, Pulvirenti I, Hawa G, Tringali G, Fiore CE: Increased sclerostin serum levels associated with bone formation and resorption markers in patients with immobilization-induced bone loss. J Clin Endocrinol Metab. 2010, 95 (5): 2248-2253. 10.1210/jc.2010-0067.CrossRefPubMed
35.
Zurück zum Zitat Gennari L, Merlotti D, Valenti R, Ceccarelli E, Ruvio M, Pietrini MG, Capodarca C, Franci MB, Campagna MS, Calabrò A, Cataldo D, Stolakis K, Dotta F, Nuti R: Circulating sclerostin levels and bone turnover in type 1 and type 2 diabetes. J Clin Endocrinol Metab. 2012, 97 (5): 1737-1744. 10.1210/jc.2011-2958.CrossRefPubMed Gennari L, Merlotti D, Valenti R, Ceccarelli E, Ruvio M, Pietrini MG, Capodarca C, Franci MB, Campagna MS, Calabrò A, Cataldo D, Stolakis K, Dotta F, Nuti R: Circulating sclerostin levels and bone turnover in type 1 and type 2 diabetes. J Clin Endocrinol Metab. 2012, 97 (5): 1737-1744. 10.1210/jc.2011-2958.CrossRefPubMed
36.
Zurück zum Zitat Garcia-Martin A, Rozas-Moreno P, Reyes-Garcia R, Morales-Santana S, Garcia-Fontana B, Garcia-salcedo JA, Muñoz-Torres M: Circulating levels of sclerostin are increased in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2012, 97 (1): 234-241. 10.1210/jc.2011-2186.CrossRefPubMed Garcia-Martin A, Rozas-Moreno P, Reyes-Garcia R, Morales-Santana S, Garcia-Fontana B, Garcia-salcedo JA, Muñoz-Torres M: Circulating levels of sclerostin are increased in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2012, 97 (1): 234-241. 10.1210/jc.2011-2186.CrossRefPubMed
37.
Zurück zum Zitat Melkko J, Hellevik T, Risteli L, Risteli J, Smedsrød S: Clearance of NH2-terminal propeptides of types I and III Procollagen is a physiological function of the scavenger receptor in liver endothelial cells. J Exp Med. 1994, 179: 405-412. 10.1084/jem.179.2.405.CrossRefPubMed Melkko J, Hellevik T, Risteli L, Risteli J, Smedsrød S: Clearance of NH2-terminal propeptides of types I and III Procollagen is a physiological function of the scavenger receptor in liver endothelial cells. J Exp Med. 1994, 179: 405-412. 10.1084/jem.179.2.405.CrossRefPubMed
38.
Zurück zum Zitat Brandt J, Krogh TN, Jensen CH, Frederiksen JK, Teisner B: Thermal instability of the trimeric structure of the N-terminal propeptide of human Procollagen type I in relation to assay technology. Clin Chem. 1999, 45 (1): 47-53.PubMed Brandt J, Krogh TN, Jensen CH, Frederiksen JK, Teisner B: Thermal instability of the trimeric structure of the N-terminal propeptide of human Procollagen type I in relation to assay technology. Clin Chem. 1999, 45 (1): 47-53.PubMed
39.
Zurück zum Zitat Baxter I, Rogers A, Eastell R, Peel N: Evaluation of urinary N-telopeptide of type I collagen measurements in the management of osteoporosis in clinical practice. Osteoporos Int. 2013, 24: 941-947. 10.1007/s00198-012-2097-4.CrossRefPubMed Baxter I, Rogers A, Eastell R, Peel N: Evaluation of urinary N-telopeptide of type I collagen measurements in the management of osteoporosis in clinical practice. Osteoporos Int. 2013, 24: 941-947. 10.1007/s00198-012-2097-4.CrossRefPubMed
40.
Zurück zum Zitat Jabbar S, Drury J, Fordham JN, Datta HK, Francis RM, Tuck SP: Osteoprotegerin, RANKL and bone turnover in postmenopausal osteoporosis. J Clin Pathol. 2011, 64: 354-357. 10.1136/jcp.2010.086595.CrossRefPubMed Jabbar S, Drury J, Fordham JN, Datta HK, Francis RM, Tuck SP: Osteoprotegerin, RANKL and bone turnover in postmenopausal osteoporosis. J Clin Pathol. 2011, 64: 354-357. 10.1136/jcp.2010.086595.CrossRefPubMed
41.
Zurück zum Zitat Liu JM, Zhao HY, Ning G, Zhao YJ, Chen Y, Zhang Z, Sun LH, Xu M-Y, Chen JL: Relationships between the changes of serum levels of OPG and RANKL with age, menopause, bone biochemical markers and bone mineral density in Chinese women aged 20–75. Calcif Tissue Int. 2005, 76 (1): 1-6. 10.1007/s00223-004-0007-2.CrossRefPubMed Liu JM, Zhao HY, Ning G, Zhao YJ, Chen Y, Zhang Z, Sun LH, Xu M-Y, Chen JL: Relationships between the changes of serum levels of OPG and RANKL with age, menopause, bone biochemical markers and bone mineral density in Chinese women aged 20–75. Calcif Tissue Int. 2005, 76 (1): 1-6. 10.1007/s00223-004-0007-2.CrossRefPubMed
42.
Zurück zum Zitat Noble BS: The osteocyte lineage. Arch Biochem Biophys. 2008, 473 (2): 106-11. 10.1016/j.abb.2008.04.009.CrossRefPubMed Noble BS: The osteocyte lineage. Arch Biochem Biophys. 2008, 473 (2): 106-11. 10.1016/j.abb.2008.04.009.CrossRefPubMed
43.
Zurück zum Zitat Bonewald LF: Osteocytes: a proposed multifunctional bone cell. J Musculoskelet Neuronal Interact. 2002, 2 (3): 239-41.PubMed Bonewald LF: Osteocytes: a proposed multifunctional bone cell. J Musculoskelet Neuronal Interact. 2002, 2 (3): 239-41.PubMed
44.
Zurück zum Zitat Zhang Y, Wang Y, Li X, Zhang J, Mao J, Li Z, Zhang J, Li L, Harris S, Wu D: The LRP5 high-bone-mass G171V mutation disrupts LRP5 interaction with Mesd. Mol Cell Biol. 2004, 24 (11): 4677-4684. 10.1128/MCB.24.11.4677-4684.2004.PubMedCentralCrossRefPubMed Zhang Y, Wang Y, Li X, Zhang J, Mao J, Li Z, Zhang J, Li L, Harris S, Wu D: The LRP5 high-bone-mass G171V mutation disrupts LRP5 interaction with Mesd. Mol Cell Biol. 2004, 24 (11): 4677-4684. 10.1128/MCB.24.11.4677-4684.2004.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D: Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signalling. J Biol Chem. 2005, 280: 19883-19887. 10.1074/jbc.M413274200.CrossRefPubMed Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D: Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signalling. J Biol Chem. 2005, 280: 19883-19887. 10.1074/jbc.M413274200.CrossRefPubMed
46.
Zurück zum Zitat You L, Temiyasathit S, Lee P, Kim CH, Tummala P, Yao W, Kingery W, Malone AM, Kwon RY, Jacobs CR: Osteocytes as mechanosensors in the inhibition of bone resorption due to mechanical loading. Bone. 2008, 42 (1): 172-179. 10.1016/j.bone.2007.09.047.PubMedCentralCrossRefPubMed You L, Temiyasathit S, Lee P, Kim CH, Tummala P, Yao W, Kingery W, Malone AM, Kwon RY, Jacobs CR: Osteocytes as mechanosensors in the inhibition of bone resorption due to mechanical loading. Bone. 2008, 42 (1): 172-179. 10.1016/j.bone.2007.09.047.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Gross TS, King KA, Rabaia NA, Pathare P, Srinivasan S: Upregulation of osteopontin by osteocytes deprived of mechanical loading or oxygen. J Bone Miner Res. 2005, 20 (2): 250-256.PubMedCentralCrossRefPubMed Gross TS, King KA, Rabaia NA, Pathare P, Srinivasan S: Upregulation of osteopontin by osteocytes deprived of mechanical loading or oxygen. J Bone Miner Res. 2005, 20 (2): 250-256.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Noble BS, Peet N, Stevens HY, Brabbs A, Mosley JR, Reilly GC, Reave J, Skerry TM, Lanyon LE: Mechanical loading: biphasic osteocyte survival and targeting of osteoclasts for bone destruction in rat cortical bone. Am J Physiol Cell Physiol. 2003, 284: C934-C943. 10.1152/ajpcell.00234.2002.CrossRefPubMed Noble BS, Peet N, Stevens HY, Brabbs A, Mosley JR, Reilly GC, Reave J, Skerry TM, Lanyon LE: Mechanical loading: biphasic osteocyte survival and targeting of osteoclasts for bone destruction in rat cortical bone. Am J Physiol Cell Physiol. 2003, 284: C934-C943. 10.1152/ajpcell.00234.2002.CrossRefPubMed
49.
Zurück zum Zitat Amrein K, Amrein S, Drexler C, Dimai HP, Dobnig H, Pfeifer K, Tomaschitz A, Pieber TR, Fahrleitner-Pammer A: Sclerostin and its association with physical activity, age, gender, body composition and bone mineral content in healthy adults. J Clin Endocrinol Metab. 2012, 97 (1): 148-154. 10.1210/jc.2011-2152.CrossRefPubMed Amrein K, Amrein S, Drexler C, Dimai HP, Dobnig H, Pfeifer K, Tomaschitz A, Pieber TR, Fahrleitner-Pammer A: Sclerostin and its association with physical activity, age, gender, body composition and bone mineral content in healthy adults. J Clin Endocrinol Metab. 2012, 97 (1): 148-154. 10.1210/jc.2011-2152.CrossRefPubMed
50.
Zurück zum Zitat Lin C, Jiang X, Dai Z, Guo X, Weng T, Wang J, Li Y, Feng G, Gao X, He L: Sclerostin mediates bone response to mechanical unloading through antagonizing Wnt/β-Catenin signalling. J Bone Miner Res. 2009, 24 (10): 1651-1661. 10.1359/jbmr.090411.CrossRefPubMed Lin C, Jiang X, Dai Z, Guo X, Weng T, Wang J, Li Y, Feng G, Gao X, He L: Sclerostin mediates bone response to mechanical unloading through antagonizing Wnt/β-Catenin signalling. J Bone Miner Res. 2009, 24 (10): 1651-1661. 10.1359/jbmr.090411.CrossRefPubMed
51.
Zurück zum Zitat Morse LR, Sudhakar S, Lazzari AA, Tun C, Garshick E, Zafonte R, Battaglino RA: Sclerostin: a candidate biomarker of SCI-induced osteoporosis. Osteoporos Int. 2013, 24: 961-968. 10.1007/s00198-012-2072-0.PubMedCentralCrossRefPubMed Morse LR, Sudhakar S, Lazzari AA, Tun C, Garshick E, Zafonte R, Battaglino RA: Sclerostin: a candidate biomarker of SCI-induced osteoporosis. Osteoporos Int. 2013, 24: 961-968. 10.1007/s00198-012-2072-0.PubMedCentralCrossRefPubMed
52.
Zurück zum Zitat Arasu A, Cawthon PM, Lui L-Y, Do TP, Arora PS, Cauley JA, Ensrud KE, Cummings SR: Serum sclerostin and risk of hip fracture in older Caucasian women. J Clin Endocrinol Metab. 2012, 97 (6): 2027-2032. 10.1210/jc.2011-3419.PubMedCentralCrossRefPubMed Arasu A, Cawthon PM, Lui L-Y, Do TP, Arora PS, Cauley JA, Ensrud KE, Cummings SR: Serum sclerostin and risk of hip fracture in older Caucasian women. J Clin Endocrinol Metab. 2012, 97 (6): 2027-2032. 10.1210/jc.2011-3419.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Mirza FS, Padhi ID, Raisz LG, Lorenzo JA: Serum sclerostin levels negatively correlate with parathyroid hormone levels and free estrogen index in postmenopausal women. J Clin Endocrinol Metab. 2010, 95 (4): 1991-1997. 10.1210/jc.2009-2283.PubMedCentralCrossRefPubMed Mirza FS, Padhi ID, Raisz LG, Lorenzo JA: Serum sclerostin levels negatively correlate with parathyroid hormone levels and free estrogen index in postmenopausal women. J Clin Endocrinol Metab. 2010, 95 (4): 1991-1997. 10.1210/jc.2009-2283.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Polyzos SA, Anastasilakis AD, Bratengeier C, Woloszczuk W, Papatheodorou A, Terpos E: Serum sclerostin levels positively correlate with lumbar spinal bone mineral density in postmenopausal women – the six-month effect of risedronate and teriparatide. Osteoporos Int. 2012, 23: 1171-1176. 10.1007/s00198-010-1525-6.CrossRefPubMed Polyzos SA, Anastasilakis AD, Bratengeier C, Woloszczuk W, Papatheodorou A, Terpos E: Serum sclerostin levels positively correlate with lumbar spinal bone mineral density in postmenopausal women – the six-month effect of risedronate and teriparatide. Osteoporos Int. 2012, 23: 1171-1176. 10.1007/s00198-010-1525-6.CrossRefPubMed
55.
Zurück zum Zitat McNulty M, Singh RJ, Li X, Bergstralh EJ, Kumar R: Determination of serum and plasma sclerostin concentrations by enzyme-linked immunoassays. J Clin Endocrinol Metab. 2011, 96 (7): E1159-E1162. 10.1210/jc.2011-0254.PubMedCentralCrossRefPubMed McNulty M, Singh RJ, Li X, Bergstralh EJ, Kumar R: Determination of serum and plasma sclerostin concentrations by enzyme-linked immunoassays. J Clin Endocrinol Metab. 2011, 96 (7): E1159-E1162. 10.1210/jc.2011-0254.PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Blumsohn A, Naylor KE, Timm W, Eagleton AC, Hannon RA, Eastell R: Absence of marked seasonal change in bone turnover: a longitudinal and multicentre cross-sectional study. J Bone Miner Res. 2003, 18: 1274-1281. 10.1359/jbmr.2003.18.7.1274.CrossRefPubMed Blumsohn A, Naylor KE, Timm W, Eagleton AC, Hannon RA, Eastell R: Absence of marked seasonal change in bone turnover: a longitudinal and multicentre cross-sectional study. J Bone Miner Res. 2003, 18: 1274-1281. 10.1359/jbmr.2003.18.7.1274.CrossRefPubMed
57.
Zurück zum Zitat Woitge HW, Scheidt-Nave C, Kissling C, Leidig-Bruckner G, Meyer K, Grauer A, Scharla SH, Ziegler R, Seibel MJ: Seasonal variation of biochemical indices of bone turnover: results of a population-based study. J Clin Endocrinol Metab. 1998, 83: 68-75. 10.1210/jc.83.1.68.PubMed Woitge HW, Scheidt-Nave C, Kissling C, Leidig-Bruckner G, Meyer K, Grauer A, Scharla SH, Ziegler R, Seibel MJ: Seasonal variation of biochemical indices of bone turnover: results of a population-based study. J Clin Endocrinol Metab. 1998, 83: 68-75. 10.1210/jc.83.1.68.PubMed
58.
Zurück zum Zitat Nielsen HK, Brixen K, Bouillon R, Mosekilde L: Changes in biochemical markers of osteoblastic activity during the menstrual cycle. J Clin Endocrinol Metab. 1990, 70: 1431-1437. 10.1210/jcem-70-5-1431.CrossRefPubMed Nielsen HK, Brixen K, Bouillon R, Mosekilde L: Changes in biochemical markers of osteoblastic activity during the menstrual cycle. J Clin Endocrinol Metab. 1990, 70: 1431-1437. 10.1210/jcem-70-5-1431.CrossRefPubMed
59.
Zurück zum Zitat Chiu KM, Ju J, Mayes D, Bacchetti P, Weitz S, Arnaud CD: Changes in bone resorption during the menstrual cycle. J Bone Miner Res. 1999, 14 (4): 609-615. 10.1359/jbmr.1999.14.4.609.CrossRefPubMed Chiu KM, Ju J, Mayes D, Bacchetti P, Weitz S, Arnaud CD: Changes in bone resorption during the menstrual cycle. J Bone Miner Res. 1999, 14 (4): 609-615. 10.1359/jbmr.1999.14.4.609.CrossRefPubMed
60.
Zurück zum Zitat Black AJ, Topping J, Durham B, Farquharson RG, Fraser WD: A detailed assessment of alterations in bone turnover, calcium homeostasis, and bone density in normal pregnancy. J Bone Miner Res. 2000, 15 (3): 557-563.CrossRefPubMed Black AJ, Topping J, Durham B, Farquharson RG, Fraser WD: A detailed assessment of alterations in bone turnover, calcium homeostasis, and bone density in normal pregnancy. J Bone Miner Res. 2000, 15 (3): 557-563.CrossRefPubMed
61.
Zurück zum Zitat Dorota D-K, Bogdan KG, Mieczyslaw G, Bozena L-G, Jan O: The concentrations of markers of bone turnover in normal pregnancy and pre-eclampsia. Hypertens Pregnancy. 2012, 31: 166-176. 10.3109/10641955.2010.484084.CrossRefPubMed Dorota D-K, Bogdan KG, Mieczyslaw G, Bozena L-G, Jan O: The concentrations of markers of bone turnover in normal pregnancy and pre-eclampsia. Hypertens Pregnancy. 2012, 31: 166-176. 10.3109/10641955.2010.484084.CrossRefPubMed
62.
Zurück zum Zitat Naylor KE, Iqbal P, Fledelius C, Fraser RB, Eastell R: The effect of pregnancy on bone density and turnover. J Bone Miner Res. 2000, 15: 129-137. 10.1359/jbmr.2000.15.1.129.CrossRefPubMed Naylor KE, Iqbal P, Fledelius C, Fraser RB, Eastell R: The effect of pregnancy on bone density and turnover. J Bone Miner Res. 2000, 15: 129-137. 10.1359/jbmr.2000.15.1.129.CrossRefPubMed
63.
Zurück zum Zitat Hannon R, Blumsohn A, Naylor K, Eastell R: Response of biochemical markers of bone turnover to hormone replacement therapy: impact of biological variability. J Bone Miner Res. 1998, 13 (7): 1124-33. 10.1359/jbmr.1998.13.7.1124.CrossRefPubMed Hannon R, Blumsohn A, Naylor K, Eastell R: Response of biochemical markers of bone turnover to hormone replacement therapy: impact of biological variability. J Bone Miner Res. 1998, 13 (7): 1124-33. 10.1359/jbmr.1998.13.7.1124.CrossRefPubMed
64.
Zurück zum Zitat van Staa TP, Leufkens HG, Cooper C: The epidemiology of corticosteroid-induced osteoporosis: a meta-analysis. Osteoporos Int. 2002, 13: 777-787. 10.1007/s001980200108.CrossRefPubMed van Staa TP, Leufkens HG, Cooper C: The epidemiology of corticosteroid-induced osteoporosis: a meta-analysis. Osteoporos Int. 2002, 13: 777-787. 10.1007/s001980200108.CrossRefPubMed
66.
Zurück zum Zitat Wheater G, Hogan VE, Teng YKO, Tekstra J, Tuck SP, Lafeber FP, Huizinga TWJ, Bijlsma JWJ, Francis RM, Datta HK, van Laar JM: Suppression of bone turnover by B-cell depletion in patients with rheumatoid arthritis. Osteoporos Int. 2011, 12: 3067-3072.CrossRef Wheater G, Hogan VE, Teng YKO, Tekstra J, Tuck SP, Lafeber FP, Huizinga TWJ, Bijlsma JWJ, Francis RM, Datta HK, van Laar JM: Suppression of bone turnover by B-cell depletion in patients with rheumatoid arthritis. Osteoporos Int. 2011, 12: 3067-3072.CrossRef
67.
Zurück zum Zitat Yamaguchi T, Sugimoto T: Bone metabolism and fracture risk in type 2 diabetes mellitus. Endocr J. 2011, 58 (8): 613-624. 10.1507/endocrj.EJ11-0063.CrossRefPubMed Yamaguchi T, Sugimoto T: Bone metabolism and fracture risk in type 2 diabetes mellitus. Endocr J. 2011, 58 (8): 613-624. 10.1507/endocrj.EJ11-0063.CrossRefPubMed
68.
Zurück zum Zitat Inque M, Tanaka H, Moriwake T, Oka M, Sekiguchi C, Seino Y: Altered biochemical markers of bone turnover in humans during 120 days of bed rest. Bone. 2000, 26 (3): 281-286. 10.1016/S8756-3282(99)00282-3.CrossRef Inque M, Tanaka H, Moriwake T, Oka M, Sekiguchi C, Seino Y: Altered biochemical markers of bone turnover in humans during 120 days of bed rest. Bone. 2000, 26 (3): 281-286. 10.1016/S8756-3282(99)00282-3.CrossRef
69.
Zurück zum Zitat Veitch SW, Findlay SC, Hamer AJ, Blumsohn A, Eastell R, Ingle BM: Changes in bone mass and bone turnover following tibial shaft fracture. Osteoporos Int. 2006, 17: 364-372. 10.1007/s00198-005-2025-y.CrossRefPubMed Veitch SW, Findlay SC, Hamer AJ, Blumsohn A, Eastell R, Ingle BM: Changes in bone mass and bone turnover following tibial shaft fracture. Osteoporos Int. 2006, 17: 364-372. 10.1007/s00198-005-2025-y.CrossRefPubMed
70.
Zurück zum Zitat Vesper H, Cosman F, Endres DB, Garnero P, Hoyle NR, Kleerekoper MK, Mallinak NJS: Application of biochemical markers of bone turnover in the assessment and monitoring of bone diseases; approved guideline. NCCLS document. 2004, 24 (22): 1-37. Vesper H, Cosman F, Endres DB, Garnero P, Hoyle NR, Kleerekoper MK, Mallinak NJS: Application of biochemical markers of bone turnover in the assessment and monitoring of bone diseases; approved guideline. NCCLS document. 2004, 24 (22): 1-37.
71.
Zurück zum Zitat Garnero P, Vergnaud P, Hoyle N: Evaluation of a fully automated serum assay for total N-terminal propeptide of type I collagen in postmenopausal osteoporosis. Clin Chem. 2008, 54 (1): 188-196.CrossRefPubMed Garnero P, Vergnaud P, Hoyle N: Evaluation of a fully automated serum assay for total N-terminal propeptide of type I collagen in postmenopausal osteoporosis. Clin Chem. 2008, 54 (1): 188-196.CrossRefPubMed
72.
Zurück zum Zitat Chubb SAP: Measurement of C-terminal telopeptide of type I collagen (CTX) in serum. Clin Biochem. 2012, 45 (12): 928-935. 10.1016/j.clinbiochem.2012.03.035.CrossRefPubMed Chubb SAP: Measurement of C-terminal telopeptide of type I collagen (CTX) in serum. Clin Biochem. 2012, 45 (12): 928-935. 10.1016/j.clinbiochem.2012.03.035.CrossRefPubMed
73.
Zurück zum Zitat Mora S, Pitukcheewanont P, Kaufman FR, Nelson JC, Gilsanz V: Biochemical markers of bone turnover and the volume and the density of bone in children at different stages of sexual development. J Bone Miner Res. 1999, 14: 1664-1671. 10.1359/jbmr.1999.14.10.1664.CrossRefPubMed Mora S, Pitukcheewanont P, Kaufman FR, Nelson JC, Gilsanz V: Biochemical markers of bone turnover and the volume and the density of bone in children at different stages of sexual development. J Bone Miner Res. 1999, 14: 1664-1671. 10.1359/jbmr.1999.14.10.1664.CrossRefPubMed
74.
Zurück zum Zitat Eastell R, Garnero P, Audebert C, Cahall DL: Reference intervals of bone turnover markers in healthy premenopausal women: results from a cross-sectional European study. Bone. 2012, 50 (5): 1141-1147. 10.1016/j.bone.2012.02.003.CrossRefPubMed Eastell R, Garnero P, Audebert C, Cahall DL: Reference intervals of bone turnover markers in healthy premenopausal women: results from a cross-sectional European study. Bone. 2012, 50 (5): 1141-1147. 10.1016/j.bone.2012.02.003.CrossRefPubMed
75.
Zurück zum Zitat Bieglmayer C, Kudlacek S: The bone marker plot: an innovative method to assess bone turnover in women. Eur J Clin Invest. 2009, 39: 230-238. 10.1111/j.1365-2362.2009.02087.x.CrossRefPubMed Bieglmayer C, Kudlacek S: The bone marker plot: an innovative method to assess bone turnover in women. Eur J Clin Invest. 2009, 39: 230-238. 10.1111/j.1365-2362.2009.02087.x.CrossRefPubMed
76.
Zurück zum Zitat Seibel MJ, Lang M, Geilenkeuser W-J: Interlaboratory variation of biochemical markers of bone turnover. Clin Chem. 2001, 47 (8): 1443-1450.PubMed Seibel MJ, Lang M, Geilenkeuser W-J: Interlaboratory variation of biochemical markers of bone turnover. Clin Chem. 2001, 47 (8): 1443-1450.PubMed
77.
Zurück zum Zitat Schafer AL, Vittinghoff E, Ramachandran R, Mahmoudi N, Bauer DC: Laboratory reproducibility of biochemical markers of bone turnover in clinical practice. Osteoporos Int. 2010, 21: 439-445. 10.1007/s00198-009-0974-2.PubMedCentralCrossRefPubMed Schafer AL, Vittinghoff E, Ramachandran R, Mahmoudi N, Bauer DC: Laboratory reproducibility of biochemical markers of bone turnover in clinical practice. Osteoporos Int. 2010, 21: 439-445. 10.1007/s00198-009-0974-2.PubMedCentralCrossRefPubMed
78.
Zurück zum Zitat Bauer D, Krege J, Lane N, Leary E, Libanati C, Miller P, Myers G, Silverman S, Vesper HW, Lee D, Payette M, Randall S: National bone health alliance bone turnover marker project: current practices and the need for US harmonization, standardization, and common reference ranges. Osteoporos Int. 2012, 23: 2425-2433. 10.1007/s00198-012-2049-z.PubMedCentralCrossRefPubMed Bauer D, Krege J, Lane N, Leary E, Libanati C, Miller P, Myers G, Silverman S, Vesper HW, Lee D, Payette M, Randall S: National bone health alliance bone turnover marker project: current practices and the need for US harmonization, standardization, and common reference ranges. Osteoporos Int. 2012, 23: 2425-2433. 10.1007/s00198-012-2049-z.PubMedCentralCrossRefPubMed
79.
Zurück zum Zitat Lee J, Vasikaran S: Current recommendations for laboratory testing and use of bone turnover markers in management of osteoporosis. Ann Lab Med. 2012, 32: 105-112. 10.3343/alm.2012.32.2.105.PubMedCentralCrossRefPubMed Lee J, Vasikaran S: Current recommendations for laboratory testing and use of bone turnover markers in management of osteoporosis. Ann Lab Med. 2012, 32: 105-112. 10.3343/alm.2012.32.2.105.PubMedCentralCrossRefPubMed
80.
Zurück zum Zitat Steiger P, Cummings SR, Black DM, Spencer NE, Genant HK: Age related decrements in bone mineral density in women over 65. J Bone Miner Res. 1992, 7: 625-632.CrossRefPubMed Steiger P, Cummings SR, Black DM, Spencer NE, Genant HK: Age related decrements in bone mineral density in women over 65. J Bone Miner Res. 1992, 7: 625-632.CrossRefPubMed
81.
Zurück zum Zitat Ross PD, Knowlton W: Rapid bone loss is associated with increased levels of biochemical markers. J Bone Miner Res. 1998, 13 (2): 297-302. 10.1359/jbmr.1998.13.2.297.CrossRefPubMed Ross PD, Knowlton W: Rapid bone loss is associated with increased levels of biochemical markers. J Bone Miner Res. 1998, 13 (2): 297-302. 10.1359/jbmr.1998.13.2.297.CrossRefPubMed
82.
Zurück zum Zitat Crane M, Davis T, Kaldale R, Black C, Davies R, Devas V, Williams W: Relating increases in bone mineral density and fracture risk reduction with early suppression in biomarkers of bone turnover: a literature-based meta-analysis of bisphosphonates treatments. J Bone Miner Res. 2005, 20 (Suppl 1): S95- Crane M, Davis T, Kaldale R, Black C, Davies R, Devas V, Williams W: Relating increases in bone mineral density and fracture risk reduction with early suppression in biomarkers of bone turnover: a literature-based meta-analysis of bisphosphonates treatments. J Bone Miner Res. 2005, 20 (Suppl 1): S95-
83.
Zurück zum Zitat Delmas PD, Eastell R, Garnero P, Seibel MJ, Stepan J: The use of biochemical markers of bone turnover in osteoporosis. Committee of Scientific Advisors of the International Osteoporosis Foundation. Osteoporos Int. 2000, 11 (Suppl 6): S2-S17.PubMed Delmas PD, Eastell R, Garnero P, Seibel MJ, Stepan J: The use of biochemical markers of bone turnover in osteoporosis. Committee of Scientific Advisors of the International Osteoporosis Foundation. Osteoporos Int. 2000, 11 (Suppl 6): S2-S17.PubMed
84.
Zurück zum Zitat Schuit SC, van der Klift M, Weel AE, de Laet CE, Burger H, Seeman E, Hofman A, Uitterlinden AG, van Leeuwen JP, Pols HA: Fracture incidence and association with bone mineral density in elderly men and women: The rotterdam study. Bone. 2004, 34: 195-202. 10.1016/j.bone.2003.10.001.CrossRefPubMed Schuit SC, van der Klift M, Weel AE, de Laet CE, Burger H, Seeman E, Hofman A, Uitterlinden AG, van Leeuwen JP, Pols HA: Fracture incidence and association with bone mineral density in elderly men and women: The rotterdam study. Bone. 2004, 34: 195-202. 10.1016/j.bone.2003.10.001.CrossRefPubMed
86.
Zurück zum Zitat Blank RD, Malone DG, Christian RC, Vallarta-Ast NL, Krueger DC, Drezner MK, Binkley NC, Hansen KE: Patient variables impact lumbar spine dual energy x-ray absorptiometry precision. Osteoporos Int. 2006, 17: 768-774. 10.1007/s00198-005-0050-5.CrossRefPubMed Blank RD, Malone DG, Christian RC, Vallarta-Ast NL, Krueger DC, Drezner MK, Binkley NC, Hansen KE: Patient variables impact lumbar spine dual energy x-ray absorptiometry precision. Osteoporos Int. 2006, 17: 768-774. 10.1007/s00198-005-0050-5.CrossRefPubMed
88.
Zurück zum Zitat Jordan N, Barry M, Murphy E: Comparative effects of antiresorptive agents on bone mineral density and bone turnover in postmenopausal women. Clin Interv Aging. 2006, 1 (4): 377-387. 10.2147/ciia.2006.1.4.377.PubMedCentralCrossRefPubMed Jordan N, Barry M, Murphy E: Comparative effects of antiresorptive agents on bone mineral density and bone turnover in postmenopausal women. Clin Interv Aging. 2006, 1 (4): 377-387. 10.2147/ciia.2006.1.4.377.PubMedCentralCrossRefPubMed
89.
Zurück zum Zitat Watts NB, Diab DL: Long-term use of bispohosphonates in osteoporosis. J Clin Endocrinol Metab. 2010, 95 (4): 1555-1565. 10.1210/jc.2009-1947.CrossRefPubMed Watts NB, Diab DL: Long-term use of bispohosphonates in osteoporosis. J Clin Endocrinol Metab. 2010, 95 (4): 1555-1565. 10.1210/jc.2009-1947.CrossRefPubMed
90.
Zurück zum Zitat Finkelstein JS, Wyland JJ, Lee H, Neer RM: Effects of teriparatide, alendronate, or both in women with postmenopausal osteoporosis. J Clin Endocrinol Metab. 2010, 95 (4): 1838-1845. 10.1210/jc.2009-1703.PubMedCentralCrossRefPubMed Finkelstein JS, Wyland JJ, Lee H, Neer RM: Effects of teriparatide, alendronate, or both in women with postmenopausal osteoporosis. J Clin Endocrinol Metab. 2010, 95 (4): 1838-1845. 10.1210/jc.2009-1703.PubMedCentralCrossRefPubMed
91.
Zurück zum Zitat Meier C, Seibel MJ, Kraenzlin ME: From Biochemical markers of bone turnover – clinical aspects. Contemporary Endocrinology: Osteoporosis: Pathophysiology and Clinical Management. Edited by: Adler RA. 2010, 131-155. Humana Press Meier C, Seibel MJ, Kraenzlin ME: From Biochemical markers of bone turnover – clinical aspects. Contemporary Endocrinology: Osteoporosis: Pathophysiology and Clinical Management. Edited by: Adler RA. 2010, 131-155. Humana Press
Metadaten
Titel
The clinical utility of bone marker measurements in osteoporosis
verfasst von
Gillian Wheater
Mohsen Elshahaly
Stephen P Tuck
Harish K Datta
Jacob M van Laar
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2013
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/1479-5876-11-201

Weitere Artikel der Ausgabe 1/2013

Journal of Translational Medicine 1/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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