Bone 48 (2011) 677–692 Contents lists available at ScienceDirect Bone j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / b o n e Review Denosumab and bisphosphonates: Different mechanisms of action and effects Roland Baron a,b,1, Serge Ferrari c,1, R. Graham G. Russell d,e,⁎,1 a Department of Medicine, Harvard Medical School, Endocrine Unit, Massachusetts General Hospital, Boston, MA 02115, USA Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA c Service des Maladies Osseuses, Département de Réhabilitation et Gériatrie, Hôpitaux Universitaires et Faculté de Médecine de Genève, CH-1211 Genève 14, Switzerland d Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, Oxford University Institute of Musculoskeletal Sciences (The Botnar Research Centre), Nuffield Orthopaedic Centre, Oxford, OX3 7LD, UK e The Mellanby Centre for Bone Research, Department of Human Metabolism, Sheffield University Medical School, Beech Hill Road, Sheffield, S10 2RX, UK b a r t i c l e i n f o Article history: Received 29 June 2010 Revised 30 November 2010 Accepted 30 November 2010 Available online 9 December 2010 Edited by: T. Jack Martin Keywords: Bisphosphonates Denosumab Osteoclast Osteoporosis RANK ligand a b s t r a c t To treat systemic bone loss as in osteoporosis and/or focal osteolysis as in rheumatoid arthritis or periodontal disease, most approaches target the osteoclasts, the cells that resorb bone. Bisphosphonates are currently the most widely used antiresorptive therapies. They act by binding the mineral component of bone and interfere with the action of osteoclasts. The nitrogen-containing bisphosphonates, such as alendronate, act as inhibitors of farnesyl-pyrophosphate synthase, which leads to inhibition of the prenylation of many intracellular signaling proteins. The discovery of RANKL and the essential role of RANK signaling in osteoclast differentiation, activity and survival have led to the development of denosumab, a fully human monoclonal antibody. Denosumab acts by binding to and inhibiting RANKL, leading to the loss of osteoclasts from bone surfaces. In phase 3 clinical studies, denosumab was shown to significantly reduce vertebral, nonvertebral and hip fractures compared with placebo and increase areal BMD compared with alendronate. In this review, we suggest that the key pharmacological differences between denosumab and the bisphosphonates reside in the distribution of the drugs within bone and their effects on precursors and mature osteoclasts. This may explain differences in the degree and rapidity of reduction of bone resorption, their potential differential effects on trabecular and cortical bone, and the reversibility of their actions. © 2010 Elsevier Inc. All rights reserved. Contents Introduction . . . . . . . . . . . . . . . . . . . . . Osteoclast differentiation and function . . . . . . . . Osteoclast differentiation . . . . . . . . . . . . . Osteoclast activity . . . . . . . . . . . . . . . . Osteoclast survival and life-span. . . . . . . . . . Osteoblast–osteoclast coupling and communication. Preclinical pharmacology . . . . . . . . . . . . . . . Mechanism of action of bisphosphonates. . . . . . Mechanism of action of denosumab . . . . . . . . Effects on osteoclastogenesis . . . . . . . . . . . Other cell targets . . . . . . . . . . . . . . . . . . Bone cells . . . . . . . . . . . . . . . . . . . . Potential effects on osteoblasts. . . . . . . . . Potential effects on osteocytes . . . . . . . . . Effects on the immune and vascular systems . . . . Immunomodulatory effects of RANKL inhibition. Bisphosphonates and the acute-phase reaction . Effects on the vascular system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 678 678 678 678 679 679 679 679 680 681 681 681 681 681 682 682 682 682 ⁎ Corresponding author. Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, Oxford University Institute of Musculoskeletal Sciences (The Botnar Research Centre), Nuffield Orthopaedic Centre, Oxford, OX3 7LD, UK. E-mail addresses: [email protected] (R. Baron), [email protected] (S. Ferrari), [email protected] (R.G.G. Russell). 1 All authors contributed equally. 8756-3282/$ – see front matter © 2010 Elsevier Inc. All rights reserved. doi:10.1016/j.bone.2010.11.020 678 R. Baron et al. / Bone 48 (2011) 677–692 Pharmacokinetics and skeletal distribution . . . . . . . . . . . . . . . . . . Animal models of bone loss . . . . . . . . . . . . . . . . . . . . . . . . . Osteoprotegerin and bisphosphonates in rodents . . . . . . . . . . . . . Single-dose and short-term effects . . . . . . . . . . . . . . . . . . Long-term suppression of bone remodeling . . . . . . . . . . . . . . Denosumab compared with bisphosphonates in preclinical models of bone loss Human RANKL knock-in mouse model . . . . . . . . . . . . . . . . . . Primate model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Clinical trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of denosumab and bisphosphonates on bone histomorphometry . . Safety and specific adverse events: denosumab vs bisphosphonates . . . . Clinical perspectives and conclusions . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Introduction The development and activation of osteoclasts is an essential process during skeletal growth in bone and mineral homeostasis. These processes involve the removal of mineralized bone by osteoclasts through resorption [1], followed by the formation and subsequent mineralization of new bone matrix through the action of osteoblasts [2,3]. In various situations, including after the menopause and in osteoporosis, as well as in pathological conditions such as osteolytic tumor metastases, rheumatoid arthritis, and periodontal disease, the amount of bone removed during resorption by osteoclasts exceeds that replaced during bone formation by osteoblasts, leading to a net loss of bone mass, and increased skeletal fragility and risk of fracture [4]. Accelerated or high bone remodeling usually causes an earlier loss of trabecular bone compared with cortical bone; in the elderly, or after a large amount of trabecular bone has already been lost, more bone loss may occur in the cortical compartment [5–7]. Increased expression and production of RANK ligand (RANKL), as seen with estrogen deprivation, is a central mechanism of osteoclast activation and increased bone resorption [8,9]. Nearly all current pharmacological treatments of osteoporosis utilize drugs that inhibit bone resorption and these include bisphosphonates, raloxifene and calcitonin. A novel and recent approach is to target the RANK/RANKL system in a specific manner. The indispensable role of the RANK/ RANKL pathway in bone remodeling is illustrated by human monogenic diseases in which absence of functional RANK or RANKL results in osteoclast-poor osteopetrosis [10,11]. In early clinical studies, inhibition of RANKL was achieved using the RANKL decoy receptor, osteoprotegerin (OPG), which functions as the natural inhibitor of/antagonist to RANKL [12,13]. Most recent clinical studies have used the new osteoporosis drug denosumab – a fully human monoclonal antibody – to bind to and inhibit RANKL. This article describes how bisphosphonates and denosumab reduce osteoclast-mediated bone resorption in fundamentally different ways. We review the process of osteoclastogenesis and bone resorption, compare and contrast the pharmacokinetic/pharmacodynamic (PK/PD) profiles and the mechanisms of action of denosumab and bisphosphonates, and summarize the similarities and differences between these drugs based on evidence from preclinical and clinical studies. We also discuss the implications that these differences may have for the treatment of osteoporosis. Osteoclast differentiation and function Osteoclasts are multinucleated cells derived from hematopoietic precursors in the monocyte–macrophage lineage. They are responsible for the degradation of mineralized bone matrix during bone development and growth, and for skeletal homeostasis, repair and bone remodeling throughout life. The overall rate of osteoclastic bone resorption is regulated at least at three levels: 1) the differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683 683 683 683 684 684 684 685 685 686 686 687 688 688 rate of osteoclasts from their monocyte/macrophage precursor pool; 2) the activity of the individual mature osteoclast, through regulation of key functional proteins; and 3) the survival rate of differentiated and mature osteoclasts. Thus, reducing osteoclast numbers via decreasing differentiation from precursors, or survival of mature cells and/or decreasing the bone resorbing activity of osteoclasts constitute three possible, yet not mutually exclusive, therapeutic approaches for the treatment of hyper-resorptive skeletal diseases. Osteoclast differentiation Two cytokines are essential and sufficient to induce osteoclast differentiation: the macrophage colony-stimulating factor (M-CSF) [14,15] and RANKL [16] (Fig. 1). M-CSF binds to c-Fms, a single transmembrane domain receptor of the tyrosine kinase family, and RANKL binds to RANK, a single transmembrane receptor of the tumor necrosis factor (TNF) receptor family, forming trimers upon ligand binding. M-CSF first activates the proliferation and survival of cells of the monocyte–macrophage lineage and the expression of RANK, allowing the action of RANKL which, together with M-CSF, constitutes an absolute requirement for commitment to and progression of early precursors along the osteoclast lineage [17–19]. The differentiation step from osteoclast precursors to multinucleated osteoclasts is then induced by RANKL, again through the M-CSF-dependent expression of RANK at the cell surface of these early precursors. Both M-CSF and RANKL are secreted by bone marrow stromal cells and osteoblasts, whereas RANKL is also secreted by T cells and, at lower levels, by B cells, establishing a link to inflammation-induced osteolysis [17,20]. Importantly, most of the cells producing RANKL also produce a decoyRANK receptor, OPG, which acts as an antagonist to RANK signaling and osteoclastogenesis by scavenging RANKL in the extracellular environment [21]. Ultimately, it is the ratio between RANKL and OPG which determines the level of activation of RANK and, thereby, the degree to which osteoclastogenesis is activated. All these molecules are thought to act in a paracrine manner and are therefore regulating bone resorption locally. The link between these local factors and the endocrine regulation of skeletal homeostasis through bone resorption and remodeling is, however, established by the fact that both RANKL and OPG expression and secretion are regulated by several calciumregulating hormones, such as sex hormones, parathyroid hormone (PTH), and vitamin D3 [22,23]. Deletion and overexpression of RANK/RANKL genes in rodents has clearly established the essential role of this pathway on osteoclastogenesis and regulation of bone mass and strength [24]. Osteoclast activity The functional features defining the activity of an osteoclast include 1) the attachment of the cells to the bone surface 2) the migration of osteoclasts along bone surfaces, 3) the synthesis and R. Baron et al. / Bone 48 (2011) 677–692 679 Fig. 1. Osteoclast intracellular signaling and development. directional secretion of hydrolytic enzymes, 4) the acidification of the subosteoclastic bone-resorbing compartment by vacuolar proton pumps (V-ATPase) and chloride channels and 5) efficient internalization of extracellular bone matrix degradation products. The importance of these functional activities is demonstrated by the fact that genetic alterations of these processes lead to defective bone resorption and osteopetrosis [3]. Whereas M-CSF has been shown to favor the migratory function of osteoclasts while decreasing their resorbing activity [25], RANKL seems overall to contribute to the activation of osteoclast function, i.e. bone resorption [26]. RANKL can activate mature osteoclasts in a dose-dependent manner in vitro, and can rapidly increase the resorption of bone in vivo by activating pre-existing osteoclasts. Osteoclast survival and life-span In vitro, the combination of RANKL and M-CSF is required for optimal osteoclast survival [17]. Indeed, osteoclast numbers decline dramatically in the absence of M-CSF or RANKL, both from an impairment of recruitment of precursors and a rapid apoptosis of existing osteoclasts. In mice, a single dose of OPG led to a N90% loss of osteoclasts within 48 h of exposure due to apoptosis and without affecting osteoclast precursor cells. Therefore, RANKL is essential, but not sufficient, for osteoclast survival and in mice endogenous M-CSF levels are not sufficient to maintain osteoclast viability in the absence of RANKL [27]. And while no factors appear capable of maintaining osteoclasts in the absence of RANK/RANKL signaling, the important role of M-CSF can be replaced by vascular endothelial growth factor (VEGF) in mice that lack functional M-CSF [28,29]. Osteoblast–osteoclast coupling and communication Bone homeostasis and bone mass are ensured by the proper balance between bone resorption and bone formation during the bone remodeling process. This balance between catabolic and anabolic functions within bone is ensured by the coupling process — an array of cross-talk networks between the osteoclasts and osteoblasts [30,31]. Any agent that affects one arm of the remodeling process usually also alters the other arm and antiresorptives tend to also decrease bone formation. This results primarily from inhibition of the activation frequency of basic multicellular units (BMUs) (i.e. the decrease in bone remodeling surfaces upon which secondary bone formation can take place), whereas the intrinsic bone-forming ability of osteoblasts may not be affected [32]. It is, however, well accepted that osteoclasts produce and/or release several osteoblast-stimulating cytokines and growth factors from the bone matrix. For example, insulin-like growth factors (IGF) I and II, transforming growth factor beta (TGF-β) and bone morphogenetic proteins (BMPs) are important regulators of osteoblastic differentiation that are released from the matrix during bone resorption. In addition, osteoclast-derived factors regulate coupling with osteoblasts. Recent findings support the existence of direct coupling between bone cells in the BMU. Both Wnt 10a and sphingosine-1-phosphate (SIP) have been identified as osteoclastderived cytokines capable of activating bone formation [33]. In addition, bidirectional signaling takes place in osteoclasts via the transmembrane ephrinB2 ligand and in osteoblasts by EphB4, the tyrosine kinase receptor of ephrinB2 [34,35]. In this physiological context, RANKL and OPG are both expressed by stromal cells and osteoblasts, and their expression is regulated by major calcium-regulating hormones, as well as by local inflammatory cytokines. They constitute a major regulatory component of the crosstalk between osteoblasts and osteoclasts. Indeed, OPG expression is often regulated in an opposite manner by factors that alter RANKL expression: upregulation of RANKL is associated with downregulation of OPG, or vice versa, altering the ratio of RANKL to OPG [24]. Thus, inhibiting RANKL interrupts the osteoblast-dependent regulation of osteoclastogenesis and osteoclast activity, thereby reducing bone resorption. Due to the coupling process it is therefore expected that in turn the reduction in the number and activity of osteoclasts will affect both osteoclast-derived clastokines and the liberation of matrixderived growth factors during resorption, thereby decreasing bone formation [36]. This phenomenon probably occurs with most antiresorptive drugs, such as bisphosphonates. It is unclear at this point whether the suppression of earlier osteoclast precursors will also contribute to the diminished osteoblast activity [37,38]. Preclinical pharmacology Mechanism of action of bisphosphonates The clinical efficacy of bisphosphonates primarily stems from two key properties: their ability to bind strongly to bone mineral and their 680 R. Baron et al. / Bone 48 (2011) 677–692 inhibitory effects on mature osteoclasts [39]. The strong attachment of bisphosphonates to bone gives bisphosphonates the unique property of selective uptake by their intended target organ. As bisphosphonates selectively bind to bone mineral, they enter and inhibit mature osteoclasts at the sites of bone resorption. There is some evidence that bisphosphonates may also have inhibitory effects on osteoclast precursors [40–42], but this process does not appear to be as complete as seen when RANKL is inhibited, because osteoclasts are not ablated after exposure to bisphosphonates in either animals or humans. Early studies with bisphosphonates in the 1960s showed that these agents have multiple effects on hydroxyapatite (HAP): they inhibit the de novo precipitation of calcium phosphate from solution, delay the transformation of amorphous to crystalline HAP, and inhibit the aggregation and dissolution of HAP crystals [43–45]. Subsequent studies showed that bisphosphonates also inhibited bone resorption in a range of experimental models [46], and led to the clinical use of bisphosphonates to treat various hyper-resorptive bone disorders. The effects of bisphosphonates on bone mineral have also been modeled using carbonated apatite, which mimics natural bone more closely than does HAP. Henneman et al. [47] found that bisphosphonates show concentration-dependent inhibition of carbonated apatite dissolution in vitro, but concluded that the contribution of this inhibitory action to the in vivo effects of bisphosphonates remains to be determined. This inhibitory action on HAP and carbonated apatite dissolution is a potential difference between bisphosphonates and denosumab, as the latter is not expected to intrinsically affect crystal dissolution. The bisphosphonates differ in their mineral binding affinity [48], and this may determine their potency and duration of action [39]. The adsorption of bisphosphonates to HAP bone mineral surfaces brings them into close contact with osteoclasts and other cells in the bone. During bone resorption, the acidic environment created by the osteoclast in the resorption lacuna is thought to allow the dissociation of bisphosphonates from HAP, as the ability of bisphosphonates to bind to HAP decreases as pH is lowered [49]. The bisphosphonate molecule – perhaps complexed with calcium and bone organic matrix proteins – is then taken up by the osteoclast. Studies using fluorescently labeled and radiolabeled bisphosphonates have shown that the most likely mechanism of uptake is fluid-phase endocytosis and that internalized bisphosphonates are initially localized within intracellular endocytic vesicles [50,51]. Acidification of these endocytic vesicles seems to be required to allow the transfer of bisphosphonates from the vesicles into the cytosol [51,52], where they exert their biochemical effect. Once in the cytosol, it is then likely that bisphosphonates are imported into organelles such as the peroxisome [51]. Studies to date suggest that the mechanisms by which bisphosphonates are internalized by osteoclasts are similar for different bisphosphonates. Bisphosphonates have a chemical structure similar to that of the inorganic molecule pyrophosphate. Bisphosphonates, however, differ from pyrophosphate in containing a phosphorus–carbon–phosphorus (P–C–P) bond instead of the phosphorus–oxygen–phosphorus bond that is present in pyrophosphate. Importantly, this P–C–P bond is highly resistant to hydrolysis, thus making bisphosphonates resistant to biological degradation. As well as being bonded to two phosphonate groups, the carbon atom in the P–C–P bond is also bonded to two side chains, known as the R1 and R2 groups. Different bisphosphonates contain different R1 and R2 groups and, as a consequence, vary in their binding affinity and biochemical potency [39,44,49,53]. Nitrogen-containing bisphosphonates, such as alendronate, ibandronate, minodronate, pamidronate, risedronate, neridronate and zoledronate, have a side chain that contains a nitrogen atom, whereas the non-nitrogen-containing bisphosphonates, such as clodronate, tiludronate and etidronate, do not. Once taken up by the osteoclast, all bisphosphonates inhibit bone resorption through intracellular effects. Non-nitrogen-containing bisphosphonates are metabolized in the osteoclast cytosol to adenosine triphosphate (ATP) analogs that block osteoclast function and induce osteoclast apoptosis [54]. In contrast, nitrogen-containing bisphosphonates act principally by inhibiting farnesyl pyrophosphate (FPP) synthase – an enzyme in the mevalonate pathway – thereby preventing the post-translational modification (prenylation) of small guanosine triphosphate (GTP)-binding proteins that are essential for osteoclast function and survival (Fig. 2) [39]. Mechanism of action of denosumab The identification of the RANK/RANKL pathway in the 1990s opened up the possibility of developing novel agents that would reduce osteoclastic bone resorption by inhibiting RANKL. The effect of RANKL inhibition was first evaluated in preclinical and clinical studies using Fc fusion proteins [13,55–59] (Fig. 3). With Fc-OPG and OPG-Fc, Fig. 2. Osteoclast biology, RANK signaling pathway, and mevalonate pathway showing the mechanism of action of nitrogen-containing bisphosphonates (NBPs). FPP: farnesyl pyrophosphate. R. Baron et al. / Bone 48 (2011) 677–692 681 Effects on osteoclastogenesis Fig. 3. History of the development of RANKL antagonists. OPG was fused to the Fc portion of human immunoglobulin G1 (IgG1) [13,56,57]. A third molecule, RANK-Fc was formed by fusing the extracellular domain of RANK (amino acids 22–201) to the Fc portion of IgG1 [59]. Later studies investigated RANKL inhibition using denosumab, a fully human monoclonal antibody that targets RANKL [60]. Denosumab is an immunoglobulin of the IgG2 subclass, and this isotype is relatively inactive, eliciting effector functions [61]. In vitro binding assays have shown high-affinity binding of denosumab and OPG to both soluble and membrane-bound forms of human RANKL [62]. However, denosumab has several advantages over OPG-Fc or FcOPG constructs. First, in terms of its selectivity, denosumab does not bind to TRAIL (TNF-related apoptosis-inducing ligand) or to other TNF family members including TNF-α, TNF-β, and CD40 ligand, whereas TRAIL binding has been observed with OPG [62,63]. Second, due to its molecular mass, its half life is prolonged compared to the Fc constructs. Finally, neutralizing antibodies against OPG-Fc could have neutralizing effects on both the drug and OPG, which would not be expected with denosumab. At present, no neutralizing antibodies have been identified with denosumab [64]. Denosumab prevents RANKL from binding to its receptor, RANK, thereby inhibiting the development, activation, and survival of osteoclasts. This is different from the mechanism of action of bisphosphonates, which bind to bone mineral and probably inhibit osteoclast function mainly by being taken up by osteoclasts at sites of bone resorption (Fig. 4). Fig. 4. Osteoclast inhibition with denosumab vs bisphosphonates (BPs). A major difference between denosumab and bisphosphonates on osteoclasts is that bisphosphonates have to be internalized to act upon cells, whereas denosumab acts in the extracellular milieu. RANKL inhibition prevents the fusion of monocytes–macrophages to become multinucleated osteoclasts, whereas long-term bisphosphonate treatment has been associated with an increase in the number of osteoclasts, including giant, hypernucleated, detached osteoclasts that undergo protracted apoptosis [65]. Inhibition of osteoclast activity by bisphosphonates could trigger a feedback loop that results in increased RANKL in the bone environment, which in turn could help stimulate osteoclast precursors to fuse into multinucleated osteoclasts. This hypothesis is supported by the evidence of increased osteoclast numbers on bone surfaces in ovariectomized huRANKL mice (see below) treated with alendronate, whereas denosumab caused a nearly complete disappearance of osteoclasts in this model [32]. Preliminary results of a study that used a rat arthritis model to compare the effects of RANKL inhibition and bisphosphonate treatment on serum and bone proteins further indicate that RANKL inhibition using OPG treatment significantly reduced RANKL in bone, whereas zoledronic acid treatment caused a two-fold increase in RANKL [66]. Other cell targets Bone cells Potential effects on osteoblasts While data exist that OPG binds to membrane-bound RANKL on osteoblasts, this does not apparently trigger intracellular signaling, but may trigger internalization of the OPG/RANKL complex by a clathrin-mediated mechanism followed by proteasomal degradation [67]. Similarly, OPG exerts no direct effects on osteoblast survival, but prevents TRAIL-induced apoptosis when added together with TRAIL to osteoblast-like cell cultures [68]. Cultured murine bone marrow stromal cells exhibited enhanced osteoblastic differentiation when treated with OPG [69], but the mechanisms of these effects have not been elucidated. It is therefore unlikely that the moderate decrease of osteoblast numbers per bone surface observed for instance in huRANKL mice treated with denosumab [62] and/or the profound decrease in bone forming indices observed with denosumab in rodents and humans (see below) [32] might be explained by the effects of RANKL antagonists directly on osteoblasts, but rather by their marked inhibition of bone remodeling. Very low concentrations of bisphosphonates (as low as 10− 11 mol/L) have been shown to stimulate osteoblasts in vitro to release factors that inhibit bone resorption by osteoclasts [70]. In contrast, others have suggested that continuous exposure of osteoblasts to high-dose bisphosphonates could inhibit osteoblast function and/or survival [71]. However, despite many studies, direct effects of bisphosphonates on osteoblasts, either positive or negative, have been difficult to demonstrate in vivo using clinically relevant doses [39]. Similar to RANKL inhibitors, most of the clinical effects of bisphosphonates on osteoblast function, such as suppression of osteoblast-derived biochemical markers, can be attributed to indirect effects on the remodeling cycle caused by the reduction in bone resorption and consequent reduction in bone formation (see section ‘Osteoblast–osteoclast coupling and communication’ above). Potential effects on osteocytes Osteocytes, which make up over 90–95% of all bone cells in the adult skeleton, are important cells for bone health. [72]. Due to their distribution throughout the bone matrix and extensive interconnectivity, osteocytes are thought to play a major role in sensing mechanical strain and orchestrating signals of resorption and formation [73]. 682 R. Baron et al. / Bone 48 (2011) 677–692 Osteocytes are a major regulator of OPG production locally, since sclerostin – a main product of these osteocytes – inhibits the production of OPG by differentiated osteoblasts [74]. A recent study has shown that suppression of beta-catenin signaling in osteocytes (like in osteoblasts) results in low bone mass mainly due to increased bone resorption, because of diminished OPG levels [75]. Conversely, preliminary data suggest that transgenic rats overexpressing OPG showed a slight but significant increase in the percentage of trabecular lacunae occupied by osteocytes in lumbar vertebrae compared with wild-type controls [76]. A modest but significant increase in the percentage of trabecular lacunae occupied by osteocytes was also observed in the distal femur of orchiectomized rats treated with OPG-Fc, compared with orchiectomized controls [77]. Whether these increases in the occupancy of lacunae by osteocytes resulted from the direct effect of OPG on osteoblast and/or osteocyte survival, or more likely from an overall decrease of bone remodeling in these animals, is unknown. Whether this phenomenon also happens with denosumab treatment is also currently unknown. Eventually, it would be interesting to know whether RANKL inhibition will affect osteocyte-mediated bone adaptation to mechanical strain. Osteocytes may be important target cells for bisphosphonates. In cell lines and experimental animals, osteocyte apoptosis is induced by many stimuli, including glucocorticoid exposure, mechanical loading, microdamage, and weightlessness [78,79]. In contrast to the ability of bisphosphonates to induce osteoclast apoptosis, very low concentrations of several bisphosphonates seem to protect osteocytes (and osteoblasts) from glucocorticoid-induced apoptosis in vitro [80]. The prevention of osteocyte apoptosis by bisphosphonates is mediated by connexin 43 hemichannel opening and subsequent activation of ERKs [81,82]. Bisphosphonates, such as amino-olpadronate, that have little antiresorptive effect are able to inhibit osteocyte apoptosis, supporting the idea that the effects of bisphosphonates on osteocytes are independent of their effects on osteoclasts [81,83,84]. In rats, low doses of risedronate and alendronate have been shown to suppress osteocyte apoptosis following cyclic mechanical loading [85]. The canalicular compartment is bathed in an extracellular fluid that is likely to be able to deliver drug to osteocytes. Studies using a fluorescein-tagged risedronate analog have provided direct evidence of distribution into this compartment [39,86]. The relevance of these observations with regard to the effects of bisphosphonates on targeted bone remodeling initiated by osteocytes remains to be elucidated. Interestingly, alendronate together with treadmill exercise exerted synergistic effects on the mechanical properties of the midshaft femur in rats, suggesting some common biological pathway [87], whereas zoledronate did not exert synergistic/additive effects with exercise or bone axial compression [88,89]. Whether these observations pertain to differential effects of various bisphosphonates on osteocyte function remains to be investigated. OPG) inhibits dendritic-cell-dependent T-cell activation [91,92]. However, when RANKL is produced by activated T cells during inflammatory processes such as colitis and rheumatoid arthritis (RA) in the absence of other immune molecule deficits, antagonizing RANK/RANKL signaling has no effect on proliferation or activation of inflammatory cells [93–97]. Stolina et al. [98] have recently published the first direct comparison of the effects of RANKL inhibition and of TNF and IL-1 inhibition on inflammation and cytokine levels. Rats with established adjuvant-induced arthritis or collagen-induced arthritis were treated with vehicle, TNF-α inhibitor (pegsunercept), IL-1 inhibitor (anakinra), or RANKL inhibitor (OPG-Fc). Anti-TNF-α or anti-IL-1 therapy inhibited parameters of local inflammation (paw swelling) and of systemic inflammation (serum levels of proinflammatory cytokines), and partially reduced local but not systemic bone loss. In contrast, RANKL inhibition by OPG-Fc prevented local and systemic bone loss in both arthritis models without inhibiting any measured local or systemic parameter of inflammation. Furthermore, a recent study showed that administration of RANK-Fc in mice did not affect T-cell cytokine production or proliferation, nor did it modify T-cell infiltration of inflammatory tissues in a model of Staphylococcus aureus-induced arthritis [99]. For a more detailed review of RANKL inhibitor effects on inflammation and immunity, see Ferrari-Lacraz and Ferrari [100]. Bisphosphonates and the acute-phase reaction The acute-phase reaction is a well-known adverse effect of nitrogen-containing bisphosphonates and encompasses influenzalike symptoms such as fatigue, fever, chills, myalgia, and arthralgia. These symptoms can occur with both oral and intravenous bisphosphonates, but are more common with the latter [101–103]. The symptoms are transient and self-limiting, and usually do not recur after subsequent drug administration [104]. Inhibition of FPP synthase by nitrogen-containing bisphosphonates causes accumulation of isopentenyl pyrophosphate (IPP), the metabolite immediately upstream of FPP synthase in the mevalonate pathway by cells (most likely monocytes) in peripheral blood. Since IPP is a ligand for a receptor on the most common subset of γ,δ-T cells in humans, Vγ9Vδ2 T cells, the accumulation of IPP leads to activation of these γ,δ-T cells, which in turn causes the release of TNF-α, thus initiating the proinflammatory acute phase response [105,106]. Statins, which inhibit an enzyme upstream of FPP synthase in the mevalonate pathway, prevent the accumulation of IPP [105,107]. Co-administration of a statin has therefore been suggested as a way to prevent the acutephase reaction in patients [105]. However, this strategy was not found to be successful when it was tried in a small, randomized study in 12 children who were receiving intravenous bisphosphonates; in this study, concurrent administration of atorvastatin did not significantly reduce pain or analgesic/antipyretic usage [108]. Effects on the immune and vascular systems Besides their effects on bone cells as reviewed above, both RANKL inhibitors and bisphosphonates exert some non-skeletal effects, particularly on immune and vascular cells, which are potentially of clinical relevance. Immunomodulatory effects of RANKL inhibition Considering the role of RANK/RANKL in the ontogenesis of the immune system [21,90] and the widespread expression of these molecules during immune and inflammatory reactions, where they appear to play a role as co-stimulatory factors for the activation of T cells and dendritic cells [9], one would expect a RANKL antagonist to influence immune and inflammatory processes. Thus, in severe immune deficiency models, such as CD40 knockout mice, and autoimmune disease models, such as IL-2 knockout mice that develop spontaneous colitis, blockade of RANKL signaling (by RANK-Fc or Fc- Effects on the vascular system Epidemiological studies have found an association between osteoporosis and vascular calcification in postmenopausal women [109] and men [110]. The RANKL signaling pathway may have a role in this association, since OPG knockout mice develop both osteoporosis and calcification of the aorta and renal arteries [111]. Inactivation of the OPG gene in ApoE knockout mice, which are prone to atherosclerosis and vascular calcification, increased each of these vascular phenotypes [112]. Therefore, the effects of RANKL inhibition using denosumab on vascular calcium deposition following glucocorticoid exposure were studied in huRANKL mice [113]. Denosumab treatment reduced aortic calcium deposition in prednisolone-treated mice by up to 50%, based on calcium measurement. Aortic calcium deposition was correlated negatively with bone mineral density (BMD) at the lumbar spine and positively with urinary excretion of deoxypyridinoline, a marker of bone resorption. R. Baron et al. / Bone 48 (2011) 677–692 Bisphosphonates may also exert favorable effects on vascular calcification. The earliest studies with bisphosphonates showed that they can inhibit aortic and kidney calcification in rats, an effect which was attributed to their direct inhibitory effects on crystal growth [43,114]. More recently, in rat models, bisphosphonates inhibited warfarin- and vitamin-D-induced artery calcification and calciphylaxis at doses comparable to those that inhibit bone resorption [115,116]. Case reports have been published where bisphosphonates have been used successfully to treat calciphylaxis in the clinical setting [117,118]. Some in vitro studies, particularly those studying the potential antitumor effects of nitrogen-containing bisphosphonates, have found that these agents, especially zoledronate and pamidronate, have antiangiogenic properties [119,120]. Whether the concentrations necessary to exert these effects can be achieved in vivo remains unknown. Pharmacokinetics and skeletal distribution One way in which denosumab differs from bisphosphonates is in its likely distribution in bone. As denosumab is an antibody, one would expect this agent to be capable of distributing throughout the extravascular space. In particular, there is no evidence of sustained binding of denosumab to bone surfaces. The skeletal disposition of denosumab has been visually assessed in huRANKL mice using immunohistochemistry. In the proximal tibia of a denosumab-treated huRANKL mouse, strong staining was observed within a major blood vessel in the marrow diaphysis, and within a vessel penetrating the cortex (Fig. 5) [62]. There was no apparent staining of bone matrix or bone surfaces, suggesting that denosumab is primarily a circulating soluble protein. A single-dose, dose–response study of denosumab in healthy postmenopausal women demonstrated a pharmacokinetic response that was non-linear and dose-dependent [121] with the drug being detectable in the circulation for up to several weeks; overall, the halflife of denosumab is approximately 26 days [121]. As seen with other antibodies, clearance of denosumab occurs through the reticuloendothelial system, and is thus independent of renal clearance. This study also assessed denosumab pharmacodynamics. At the clinically relevant dose of 1 mg/kg, a single subcutaneous dose of denosumab resulted in a rapid and profound decrease in urinary N-telopeptide/ creatinine that was 80% lower at 1 week and 59% lower at 6 months after administration compared with baseline [121]. Furthermore, the mean serum intact PTH level doubled from its baseline value within 4 days after administration of this dose of denosumab; it returned to baseline levels during follow-up. This transient increase in PTH may 683 occur to mitigate the decrease in serum calcium. Whether it also contributes to promote modeling-based bone mass gain remains unknown at this time. In contrast, some of the bisphosphonate that is originally adsorbed on to bone mineral surfaces becomes buried within bone by being covered by bone that is synthesized de novo at the original site of bone resorption [122]. Whether this accumulation leads to a continuous and progressive decrease in bone remodeling has been studied in animal models and clinical studies. Using a rat model, Reitsma et al. showed that inhibition of bone resorption by pamidronate was dosedependent, appeared to reach a steady-state level within days, and did not become progressively lower, even with repeated dosing [123]. In long-term clinical studies of alendronate, ibandronate, risedronate and zoledronate, a new steady state of bone turnover was achieved within 1–6 months of starting treatment, and this reduced level of bone resorption remained constant for as long as treatment continued [101,124–128]. The findings of these studies suggest that bisphosphonate buried in bone does not affect resorption for at least as long as it remains buried there. The pharmacokinetic half-lives for the elimination of bisphosphonates that are retained in bone are thus not equivalent to the half-lives of bisphosphonates' biochemical effects. The elimination of bisphosphonate from bone depends greatly on remodeling and resorption [122,129]. After release from the skeleton mainly as a result of resorptive action, bisphosphonates are excreted via the kidneys. Indeed, urinary excretion of small amounts of bisphosphonate that has been released from the skeleton can be measured over many weeks, months, or even years after stopping bisphosphonate treatment [130,131]. This means that bisphosphonate must be present in the circulation and available for re-uptake into bone for prolonged periods. This recycling of bisphosphonates back onto bone mineral surfaces has been proposed as the reason for the long duration of action of zoledronate in particular, and also of alendronate [39]. This helps to explain why the effects of bisphosphonates are less rapidly reversible after stopping treatment than those of denosumab (see section ‘Clinical trials’). Animal models of bone loss Osteoprotegerin and bisphosphonates in rodents Single-dose and short-term effects In rodents, blocking RANKL was first achieved by using OPG, because denosumab – which is highly specific for human RANKL – does not recognize mouse or rat RANKL and requires genetically modified mice Fig. 5. Immunohistochemical assessment of denosumab disposition in the proximal tibia of a huRANKL mouse. Left panel: Negligible specific staining was seen in bones from huRANKL mice treated with phosphate-buffered saline (PBS). Right panel: In a denosumab (DMab)-treated huRANKL mouse, strong staining was observed within a major blood vessel in the marrow diaphysis, and within a vessel penetrating the cortex (circled). Reproduced with permission from [62]. 684 R. Baron et al. / Bone 48 (2011) 677–692 expressing huRANKL. In intact female rats, a single injection of recombinant human OPG caused a transient decrease in the serum osteoclastic marker TRACP between 2 and 20 days after injection. The decrease in TRACP returned to a level similar to the control by day 30. Similarly, osteoclast-associated bone surfaces decreased by approximately 80% by day 2, and by up to 90% between days 5 and 10, after which a recovery was noted with more than 50% of the original osteoclastic surfaces being present again 30 days after the injection. Although no detectable change in serum calcium levels was noted throughout the duration of the experiment, there was a transient increase in serum PTH levels in the first 24 h after injection. The amount of bone mineral at the proximal tibia metaphysis increased markedly over 30 days after a single injection of recombinant OPG, which was paralleled by a near doubling of the trabecular bone volume at this site [132]. Histomorphometrical bone turnover indices were not reported in this experiment. In another experiment in ovariectomized rats, 6 weeks' administration of recombinant OPG markedly decreased osteoclast and osteoblast surfaces at the distal femoral metaphyses. This was associated with significantly greater bone mass and trabecular microarchitecture, as well as with increased bone strength at other skeletal sites (vertebral and femoral neck) [133]. A similar experimental design has been used to assess the effects of a single dose of a bisphosphonate. Female rats received a single intravenous injection of zoledronic acid 4 days before bilateral ovariectomy [71]. Bone mass and density and serum bone markers were then measured every 4 weeks for 32 weeks. Zoledronate given at 100 μg/kg (equivalent to the human 5-mg dose) completely prevented the decreases in total BMD and cortical thickness that occurred after ovariectomy in untreated (vehicle) animals, and this persisted for the 32 weeks of the study. This dose also increased cancellous bone mass at 32 weeks vs vehicle. Zoledronate given at a 100 μg/kg dose significantly suppressed osteocalcin (a serum marker of bone formation) by about 30% for the entire study duration, and reduced TRACP5b activity by up to 90% from week 4 to week 8, when TRACP5b levels began to rise gradually; however, TRACP5b activity was still significantly suppressed for the entire duration of the study (weeks 4 to 32). This prolonged inhibition of bone turnover markers by a single injection of zoledronate was reflected by a significant reduction in mineralizing surface, mineral apposition rate, and formation rate of cancellous bone at 32 weeks. Of note, this study provided no information about the effects of zoledronate on osteoclast or osteoblast numbers on bone surfaces. Comparing the results of these two single-dose experiments indicates that robust inhibition of bone remodeling by a single dose of either a RANKL inhibitor or a bisphosphonate increases bone mass and preserves bone structure in rodents. Although rodent models do not spontaneously develop cortical porosity nor cortical thinning, transgenic mice which express a constitutively active PTH/PTH-related protein (PTH/PTHrP) receptor in osteoblasts exhibit not only increased osteoblast and osteoclast numbers on bone surfaces, with a resulting higher cancellous bone volume, but also prominent cortical porosity as well as fibroblastoid transformation of the bone marrow, which are consistent with the phenotype of hyperparathyroidism [134]. OPG, zoledronate, or alendronate all similarly increased trabecular bone volume in these mice compared with vehicle-treated transgenic mice, suggesting that trabecular bone resorption was comparably suppressed by these agents [135]. In contrast, only OPG greatly reduced osteoclast numbers and abrogated the increased cortical porosity and bone marrow fibrosis in this particular model, indicating a key role for osteoclastic cells (rather than bone resorption per se) in these two pathological processes. Whether the observation in this particular model has clinical relevance remains uncertain. Long-term suppression of bone remodeling To study the effects of continuous long-term inhibition of RANKL on bone physical properties, transgenic rats were engineered to overexpress OPG [76]. In these rats, 1 year of continuous overexpression of OPG was found to markedly suppress bone resorption, and also resulted in significant increases in bone mass, cortical area and thickness, and trabecular bone volume, density, and breaking strength in lumbar vertebrae, compared with wild-type rats. There was an overall reduction in femur bending strength, which – as in other rodent osteopetrosis models [136] – appeared to be caused by abnormal long bone geometry and not by any changes in bone material properties [76]. However, considering the lack of haversian bone remodeling in rodents under normal conditions, there is a question of whether this model is fully appropriate to evaluate the consequences of suppressed bone remodeling on cortical bone strength. Long-term administration of alendronate in rodents has proven safe and effective at increasing bone mass and strength. Dogs, rather than rodents, have been used to assess the relationship between bone microdamage and the suppression of remodeling by bisphosphonates. Studies using a beagle dog model have shown that long-term suppression of bone turnover by bisphosphonates is associated with microdamage accumulation in the ribs and vertebrae [137–140]. However, data from the dog model suggest that the level of microdamage accumulation in animals treated with anti-remodeling agents (bisphosphonates or raloxifene) does not correlate with bone toughness nor does it interfere with breaking strength [141]. Intriguingly, a 3-year course of high-dose alendronate therapy in these young gonad-intact dogs did not increase vertebral BMD or vertebral strength compared to vehicle controls [137], which differs from the typical response of mouse, rat, or primate vertebrae to longterm antiresorptive therapy. The applicability of these animal studies to humans remains unclear, as they used dogs that were not estrogen deficient and did not have low BMD [142]. In addition, reduced material properties with bisphosphonates have not been corroborated in any of the numerous long-term studies conducted in adult ovariectomized primates. Whether microdamage accumulation occurs in postmenopausal women treated long term with bisphosphonates remains controversial [143,144]. In summary, long-term inhibition of bone turnover by antiresorptives in preclinical experiments (rodents and dogs) seems to increase cancellous bone mass and strength, whereas its effects on cortical bone remain uncertain. Denosumab compared with bisphosphonates in preclinical models of bone loss Human RANKL knock-in mouse model As denosumab does not recognize murine RANKL, a mouse model expressing a chimeric mouse/human RANKL gene (huRANKL mouse) was developed. These mice have a slightly lower osteoclast number per bone surface, and an approximately 50% greater trabecular bone volume fraction (BV/TV) compared with normal mice. This may be because the affinity of human RANKL is likely to be less for mouse RANK. In intact adult huRANKL mice, short-term administration of denosumab reduced bone resorption and markedly increased cortical and cancellous bone mass and trabecular BV/TV (N2× increase) [62]. In ovariectomized huRANKL mice, denosumab administered at the time of ovariectomy was more effective than alendronate in increasing BMD and preserving trabecular architecture and cortical thickness in vertebrae and distal femur after 4 and 8 weeks [32]. In this experiment, BMD and trabecular bone volume fraction (BV/TV) was higher in estrogen-deficient mice after 8 weeks of denosumab treatment than in intact mice without treatment, indicating that RANKL inhibition had not only prevented the ovariectomy-induced increase in bone turnover, as observed with alendronate, but lowered bone turnover below baseline levels, with a further beneficial effect on bone mass and microstructure. Histomorphometrical analysis showed an increase in osteoclast number per bone surface in alendronate- R. Baron et al. / Bone 48 (2011) 677–692 treated mice, but a marked decrease with denosumab. There was a significant decrease of bone remodeling indices with alendronate and the absence of detectable mineralizing surfaces with denosumab. Bone turnover, as evaluated both systemically (circulating levels of TRACP5b) and at the tissue level (bone formation rate [BFR]), was inversely correlated with BMD, trabecular BV/TV and vertebral cortical thickness (R values ≥ 0.7). Of note, administration of intermittent daily PTH concomitantly with alendronate or denosumab resulted in similar mineral apposition rates in both groups, indicating the intact response of osteoblastic bone formation. However, the PTHstimulated BFR remained significantly lower in the denosumab group than in the alendronate group, probably reflecting the smaller remodeling space with denosumab. In addition, there are a number of experiments in rodents combining alendronate (and/or zoledronate) with PTH, which confirm that in general those drugs exert additive effects on the skeleton [69,145,146]. This is in contrast to clinical studies with alendronate and PTH, which show limited additive effects with this combination [147]. The reasons why the response to PTH may be smaller in patients who have received or are receiving alendronate is unknown. The interaction between denosumab and PTH in clinical studies has not been studied to date. Male huRANKL mice were used to study the effects of denosumab on glucocorticoid-induced bone loss. In adult male huRANKL mice treated with prednisolone, bone loss was associated with suppressed vertebral bone formation and increased bone resorption [148]. Denosumab prevented this prednisolone-induced bone loss through a pronounced antiresorptive effect. Biomechanical compression tests of lumbar vertebrae revealed a detrimental effect of prednisolone on bone strength that was prevented by denosumab. Male huRANKL mice were also used to compare the effects of denosumab and alendronate on fracture healing [149]. Unilateral transverse femur fractures were generated and the mice were then treated with twice weekly doses of alendronate (0.1 mg/kg), denosumab (10 mg/kg), or phosphate buffered saline (control group). Within each treatment group, the mice were further divided into subgroups receiving treatment for either 21 or 42 days. Denosumab treatment almost completely suppressed serum TRACP5b levels, whereas alendronate reduced the levels of this osteoclastic marker by only about 25%. Qualitative histological analysis showed that the 21- and 42-day alendronate and denosumab groups had greater amounts of unresorbed cartilage or mineralized cartilage matrix compared with the controls, and unresorbed cartilage could still be seen in the denosumab group at 42 days. Although alendronate and denosumab delayed the removal of cartilage and the remodeling of the fracture callus, this did not diminish the mechanical integrity of the healing fractures. In contrast, strength and stiffness were enhanced in alendronate- and denosumab-treated animals at day 42 compared with the control group. This study shows that neither the suppression of osteoclasts by denosumab nor inhibition of osteoclastic activity by alendronate impairs the fracture consolidation by the callus process. Whether anti-resorptives delay osteonal healing by interfering with remodeling processes at the fractured bone ends, however, remains controversial. Primate model In a long-term (16-month) toxicology safety study of denosumab in ovariectomized adult cynomolgus monkeys (available in abstract format only), once-monthly subcutaneous injection of denosumab significantly increased bone mineral content in vertebrae and the proximal femur, as well as in the more cortical femur diaphysis [150]. Preliminary evidence indicates that peak load was significantly increased in both vertebrae and the femur neck upon compression testing, whereas the intrinsic properties of the bone materials remained unaltered, suggesting that denosumab improved bone strength at these sites primarily by increasing bone mass. Denosumab 685 markedly inhibited histomorphometrical indices of bone remodeling after 6 months and until study completion, as observed on both rib biopsies and tibia metaphyses. One of the mechanisms by which denosumab could have increased cortical volumetric BMD in these animals is by inhibiting haversian remodeling with a significant reduction of intracortical porosity compared with ovariectomized controls, which was inversely correlated with femur bone strength [151]. In a subsequent study, ovariectomized cynomolgus monkeys were switched to once-monthly denosumab after 6 months of continuous alendronate treatment [152]. Switching to denosumab further decreased serum CTx (C-terminal telopeptide of type I collagen) to virtually undetectable levels, and also further decreased bone-specific alkaline phosphatase (BSAP) and osteocalcin. This decrease in biochemical markers of bone turnover with denosumab reflected a complete disappearance of osteoclasts from cancellous bone surfaces, i.e. in contrast to the continuous alendronate group in which osteoclast surfaces remained virtually unchanged. In this experiment, both denosumab and alendronate markedly inhibited bone remodeling indices in cancellous bone, although switching to denosumab led to a significant improvement of the compressive strength of vertebral trabecular cores. Both drugs also reduced cortical porosity at the rib and tibial diaphysis by about 50%, although cortical porosity was non-significantly lower in monkeys switched from alendronate to denosumab. Cortical bone turnover was also more greatly suppressed in monkeys switched from alendronate to denosumab than in monkeys treated with alendronate alone. Bone mass and strength remained well correlated with 12 months of denosumab or 12 months of alendronate therapy in ovariectomized cynomolgus monkeys. This observation suggests that bone quality was not adversely affected by either agent, and that increments in bone mass with either agent should translate to proportional improvements in bone strength. Whether denosumab exerts structural effects on cortical bone that are different from bisphosphonates, however, remains unclear. Clinical trials In randomized controlled trials in postmenopausal women with osteoporosis, bisphosphonates have significantly reduced the risk of vertebral fractures by 39–70% compared with placebo [101,153–159]. Analyses of randomized controlled trials with alendronate, risedronate, and zoledronic acid have also shown significant antifracture efficacy at nonvertebral sites and the hip, with up to 25% and 40% relative risk reduction, respectively [101,154,157–159]. Similar results were found in the FREEDOM trial, after 3 years of treatment, where denosumab (60 mg subcutaneously every 6 months) significantly reduced the risk of vertebral fractures by 68%, nonvertebral fractures by 20%, and hip fractures by 40%, compared with placebo [160]. Comparative fracture trials of denosumab vs bisphosphonates have not been performed. In two double-blind, randomized clinical studies, the efficacy and safety of denosumab vs alendronate were evaluated in treatmentnaïve patients or those previously treated with alendronate. Both studies showed significantly greater BMD increases at all measured sites (including the one-third distal radius, which is considered a predominantly cortical site) in denosumab-treated patients compared to those treated with alendronate (approximately + 1% BMD with denosumab vs alendronate) [161,162]. Decreases in concentrations of bone turnover markers (serum CTX and P1NP) were also significantly greater with denosumab than with alendronate treatment. Whether these differences will translate into clinically relevant benefits is unclear. Furthermore, the effects of discontinuing denosumab or alendronate treatment in postmenopausal women with low BMD were studied in a phase 2 trial [163]. Decreases in BMD at the lumbar spine and total hip were faster in patients who discontinued denosumab 686 R. Baron et al. / Bone 48 (2011) 677–692 than in patients who discontinued alendronate, suggesting that denosumab has a faster offset of action than oral weekly alendronate at these sites. Whether the rapid reversal of bone turnover that occurred with denosumab withdrawal could negatively affect bone structure and strength remains to be investigated. The effects of denosumab and alendronate in preventing microarchitectural deterioration of cortical and trabecular bone were compared in a placebo-controlled, randomized, double-blind, phase 2 study in postmenopausal women with moderately low bone mass [164]. Morphologic changes were assessed using high-resolution peripheral quantitative computed tomography (HR-pQCT). In the placebo group, both cortical and trabecular volumetric BMD decreased over 1 year at the distal radius (by 1.5% and 2%, respectively). These changes were prevented by alendronate and denosumab. In cortical bone, there was actually a small net gain of volumetric BMD with denosumab that was significantly greater than with alendronate. More surprisingly, cortical thickness at the distal radius, which did not significantly change from baseline in the placebo group, increased by 2% with alendronate and 3% with denosumab. Cortical thickness at the distal tibia increased by approximately 1% in the placebo group, and by 4% with alendronate and 5% with denosumab. Consequently, the calculated polar moment of inertia, an index of bone strength at the distal radius, significantly improved with both agents, more significantly so with denosumab. In a recent study by Genant and colleagues of 332 postmenopausal osteopenic women, using QCT, the decrease in cortical thickness at the distal radius observed in the placebo group was prevented in the group treated with denosumab [165] whereas volumetric BMD and the polar moment of inertia tended to be greater after 24 months of denosumab than at baseline, particularly at the ultradistal site. These results are consistent with dual-energy X-ray absorptiometry results previously reported from the same study, in which areal BMD increased by 1.4% at the one-third radius in women receiving denosumab and decreased by 2.1% in those receiving placebo. A possible explanation for the increase in bone mass and structure seen with denosumab at the radius could be due to a reduction in cortical porosity as shown by the monkey studies (above). Although QCT has limited spatial resolution and precision in the evaluation of cortical and trabecular bone structure [166], these results could indicate potential differences between denosumab and bisphosphonates, which have not been shown to produce such large changes at the radius. Whether this will lead to detectable differences in nonvertebral fracture risk reduction is currently uncertain. Effects on cortical porosity have been observed in iliac crest bone biopsies from women treated with risedronate [167]. In summary, whereas there is accumulating evidence that denosumab could improve bone mass and estimated strength at cortical bone sites (particularly the distal radius) potentially more than alendronate, the exact structural mechanisms by which denosumab exerts these effects, for instance, through a greater suppression of cortical porosity and/or endocortical resorption remain unclear. Effects of denosumab and bisphosphonates on bone histomorphometry Bone histology is used in the evaluation of drugs used in osteoporosis to assess both efficacy and safety. It is well known from studies of different bisphosphonates that there is a reduction in activation frequency and bone turnover as assessed by the BFR and mineralizing surfaces, using quantitative histomorphometric techniques. However, the mineral apposition rate has been shown to decrease, not change or even increase with bisphosphonates, depending on the method used to evaluate (or discard) biopsies where double tetracycline labels were undetectable. Results from iliac crest biopsies from patients treated with denosumab have recently been published by Reid and colleagues, and as expected showed that denosumab produces a marked and sustained inhibition of osteoclasts, activation frequency and bone turnover [38]. The bone microarchitecture remains normal and there was no evidence of adverse effects on mineralization or the formation of lamellar bone. The inhibition of bone turnover with denosumab was greater than with alendronate, which is consistent with the reduction in biochemical markers of bone resorption. Hence, in a comparative study with alendronate and denosumab, histomorphometry analysis revealed that all biopsies from alendronate-treated subjects showed double label in trabecular and cortical bone whereas this was reduced in denosumab-treated subjects (trabecular bone: 90% alendronate vs 20% denosumab and cortical bone: 100% vs 53%, respectively) [38]. Table 1 provides a summary of the results obtained with different bisphosphonates compared with denosumab. Safety and specific adverse events: denosumab vs bisphosphonates The safety profile of denosumab has been well-documented in the phase 3 FREEDOM trial with no increase in the risk of cancer, cardiovascular disease, fracture healing, hypocalcemia or infection observed in women treated with denosumab vs those receiving placebo. However, compared with placebo, a higher number of serious adverse events of cellulitis were reported in denosumabtreated women (0.3% [n = 12] vs b0.1% [n = 1]) [160]. In phase 3 clinical trials and clinical practice, bisphosphonates have been shown to be generally well tolerated [168]. Some of the most commonly reported side effects include those on the gastrointestinal tract and kidney function as well as transient acute phase reactions (extensively reviewed by Pazianas et al. [168]). There is currently concern regarding the decrease in bone resorption associated with bone loss therapy and conditions such as osteonecrosis of the jaw (ONJ) and atypical subtrochanteric fractures. Bisphosphonate-related ONJ was originally reported in patients receiving treatment to prevent hypercalcemia or other skeletal complications of cancer [169,170]. For patients with metastatic bone disease receiving denosumab, the incidence of ONJ observed, to date, is similar to the incidence observed with bisphosphonates [171,172]. In postmenopausal osteoporosis, rare cases of ONJ have been reported in both women receiving bisphosphonates as well as denosumab [64,170,173], but no causal association has been established. In patients receiving long-term bisphosphonate (particularly alendronate) therapy for 5–10 years, there have been reports of atypical subtrochanteric femoral fractures, which have been linked with impaired bone remodeling [174]. However, these fractures are rare, even after 10 years of treatment [175], and a causal association with bisphosphonate treatment remains unproven. To date, there have been no reports of subtrochanteric fractures with denosumab [160]. Both these topics have been extensively reviewed elsewhere [168]. Another area of interest and possible distinction between denosumab and bisphosphonates is in their effects on the kidney. As an antibody, denosumab is not known to have effects on the kidneys, nor have adverse events been identified. In contrast, bisphosphonates are known to be excreted unaltered through the kidneys via filtration and possibly by proximal tubular secretion. Intravenous administration produces exposure to high initial concentrations of bisphosphonates in the kidney and may be associated with acute renal injury [176] and nephrotoxicity [177]. Although this has not been shown for oral bisphosphonates when used as labeled for the treatment of osteoporosis, the administration of bisphosphonates is contraindicated in patients with severe renal impairment (creatinine clearance b30 mL/min) due to “lack of sufficient clinical experience”, meaning that patients with at least stage 4 renal insufficiency were excluded from the registration studies with these drugs [178–182]. Recent publications indicate that oral bisphosphonates, including alendronate [183] and risedronate [184,185], may be safe and effective in reducing fractures in patients with glomerular filtration rates less R. Baron et al. / Bone 48 (2011) 677–692 687 Table 1 Effects of antiresorptives on the main histomorphometrical dynamic and structural bone parameters.a Trabecular MAR (μm/day) Bone forming rate (μm3/μm2/day) MS/BS (%) Activation frequency (/year) BV/TV (%) 2D 3D Trabecular number (/mm) 2D 3D Cortical thickness (mm) 2D 3D Alendronateb PBO Zoledronic acid PBO Risedronate PBO Ibandronatec PBO Denosumabd PBO 0.63–0.70 0.003–0.019 0.25–2.30 0.035–0.223 0.59 0.039 6.37 0.451 0.60 0.05 0.45 0.1 0.53 0.15 4.79 0.27 0.55 BFR/BV 0.085 1.1 0.15 0.58 (/year) 0.18 5.5 0.34 0.48 0.01 2.0 0.1 0.40 0.02 3.1 0.2 0.30 0.004 0.12 0.002 0.75 0.14 3.08 0.2 17.1 19.4 13.4 16.2 16.9 16.2 14.2 12.9 19.3 20.4–22.7e 19.0 23.0–20.0e 15.3 NA? 12.6 NA? 13.5 25.2 12.5 16.2 1.19 1.46 1.07 1.31 NA 1.36 NA 1.22 1.4 1.32–1.31e 1.3 1.50–1.31e 1.2 NA 1.1 NA NA 1.46 NA 1.31 NA NA NA NA 0.624 0.72 (0.79)f 0.511 0.63 1.1 0.91–0.83e 1.0 0.86–0.74e NA NA NA NA 0.658 0.565 0.765 0.710 Table adapted from Ferrari [188] (with kind permission from Springer Science + Business Media B.V.), with data from [189]. BFR: bone forming rate; BV: bone volume; BV/TV: trabecular bone volume fraction; MAR: mineral apposition rate; MS/BS: mineralizing surface per bone surface; NA: not available; PBO: placebo. a As evaluated on iliac crest bone biopsies after 3 years of treatment in postmenopausal women. b 5 or 10 mg/day for 2 and 3 years. c Results are shown only for the 2.5 mg oral daily dose. Greater inhibition of bone turnover indices may be achieved with 3 mg IV every 3 months [190]. d Dynamic indices could be evaluated on 7/37 biopsies pooled from 2 and 3 years; 3D structural indices at 3 years. e The two numbers for each treatment indicate values that were evaluated separately in a subgroup of women with low or high turnover, respectively (MS/BS b or N median). f The result in parentheses is from a subgroup with previous non-bisphosphonate antiresorptive treatment. than 30 mL/min. Similarly, denosumab also reduces fractures in patients with impaired renal function [186]. However, in clinical practice, because of the possible presence of adynamic bone disease in patients with severe renal impairment, in whom any further reduction of bone turnover might be detrimental, antiresorptive drugs should only be used with caution. Clinical perspectives and conclusions Treatment with denosumab appears to have at least three major points of difference compared with bisphosphonate treatment. Firstly, in treatment-naïve women and those transitioning to denosumab from alendronate, who thus already have low levels of bone turnover markers, biochemical bone turnover markers are reduced. This could reflect either a more profound inhibition of bone remodeling (i.e., a more profound inhibition of the activation frequency of new BMUs) with denosumab at any skeletal site or a difference in their inhibitory effects depending on their ability to gain access to different sites within the skeleton. The strong affinity of bisphosphonates for HAP and bone mineral may limit their even distribution throughout the skeleton, particularly to sites deep within the bone. However, bisphosphonates with lower affinity for bone mineral appear to penetrate deeper into the network [187]. Secondly, compared with alendronate, denosumab treatment results in greater gains in BMD after 1 year at all sites, including cortical bone sites such as the one-third radius (Table 2). As denosumab is a circulating antibody, it is expected to reach all sites within bone, including intracortical sites such as the distal radius, which is not typically seen with bisphosphonates, as summarized in Table 2. Whether denosumab has significantly greater effects on cortical porosity and/or thickness than bisphosphonates, however, remains unclear. Lastly, the effects of denosumab on bone turnover are quickly reversible with discontinuation, actually leading to a transient rebound phenomenon, and can be restored with subsequent retreatment. Results of the phase 2 trial in postmenopausal women with low BMD also suggest that denosumab has a faster offset of action at the lumbar spine and total hip than weekly oral alendronate. However, the speed of offset of action of bisphosphonates varies depending on their chemical structure, dose, and frequency of administration. Other novel approaches to the treatment of osteoporosis include cathepsin-K inhibitors, such as odanacatib, and new selective estrogen receptor modulators, such as bazedoxifene and lasofoxifene. The place of denosumab and other novel agents in the management of osteoporosis Table 2 Comparison of RANKL inhibitors and bisphosphonates on cortical bone. Study design Results OPG in rodent models Transgenic mice expressing constitutively active PTH/PTH-related protein receptors on cells of the osteoblast lineage [135] Intracortical porosity: OPG ↓↓↓ Alendronate ↓ Zoledronate ↓ Denosumab in rodent models Knock-in mice expressing a chimeric human/mouse RANKL gene (huRANKL mice) [191] Ovariectomized huRANKL mice [32] Cortical volumetric bone density and thickness: Denosumab ↑ Alendronate ↑ Cortical thickness in vertebrae and distal femur: Denosumab ↑↑a Alendronate ↑ Denosumab in cynomolgus monkey models Cortical porosity at the rib and tibial Ovariectomized cynomolgus monkeys switched to denosumab after 6 months of diaphysis, and cortical bone turnover: continuous alendronate treatment [152] Switched to denosumab ↓↓b Alendronate alone ↓ Clinical studies of denosumab vs alendronate DECIDE — 12-month, randomized, double- BMD at all sites including predominantly cortical (hip and blind, phase 3 trial in postmenopausal distal radius): women with low BMD [161] Denosumab ↑↑c Alendronate ↑ STAND — randomized, double-blind, phase Femoral neck and one-third radius BMD: 3 trial in postmenopausal women with Switched to denosumab ↑↑c low BMD switching from alendronate [162] Continued alendronate ↑ HR-pQCT phase 2 study in postmenopausal Cortical volumetric BMD and women with low BMD thickness at distal radius and tibia: Denosumab ↑↑d Alendronate ↑ Key: ↑ = increased; ↓ = decreased; a greater number of arrows indicates a greater effect. BMD: bone mineral density; HR-pQCT: high-resolution peripheral quantitative computed tomography; OPG: osteoprotegerin; PTH: parathyroid hormone. a Significantly better than alendronate at vertebra. b Not significantly better than alendronate. c Significantly better than alendronate. d Significantly better than alendronate on cortical volumetric BMD. 688 R. Baron et al. / Bone 48 (2011) 677–692 Table 3 Summary of differences between bisphosphonates and denosumab. Characteristics Bisphosphonate Denosumab Chemistry Targets Chemical agent Selective uptake by hydroxyapatite. Inhibition of FPP synthase (for nitrogen bisphosphonates) [129] Bone mineral surface [129,193] Mature osteoclasts. Possible effects on osteoclast precursors and osteocytes [41] Inhibits osteoclast resorptive function and survival by disrupting intracellular signaling pathways. Dysfunctional osteoclasts may persist [39,195] Oral or IV (depending on the bisphosphonate). Daily, weekly, or monthly (oral), quarterly or yearly (IV) Fast onset of action if given parenterally, slower orally Monoclonal antibody Selectively binds RANKL [192] Distribution Major bone target cells Mechanism of action Mode of administration Onset of action (serum CTX) Inhibition of bone resorption (serum CTX) Circulating in blood and extracellular fluid [194] Osteoclast precursors and mature osteoclasts [194] Prevents the formation, function, and survival of osteoclasts. Depletes osteoclasts [194] Subcutaneous (60 mg 6 monthly) Faster onset of action than oral alendronate [192] Appears to reduce bone resorption to a greater extent than alendronate, e.g. DECIDE study [161] Effect on BMD Denosumab produces significantly greater gains in BMD at all measured skeletal sites when compared with alendronate [161] Duration of action and reversibility of effect Depends on type of BP and length of treatment. Slow offset of action Fully reversible and relatively rapid offset of action [192] after stopping treatment Contraindications Pregnancy Pregnancy Severe renal impairment Hypocalcemia Abnormalities of the esophagus which delay esophageal emptying Hypersensitivity to the active substance or to any of the excipients (tablet formulations) Inability to stand or sit upright for at least 30–60 min (tablet formulations) Patients at increased risk of aspiration (alendronate oral solution) Hypocalcemia Hypersensitivity to any component of the product BP: bisphosphonate; CTX: C-terminal telopeptide; FPP: farnesyl pyrophosphate; IV: intravenous. will be determined by these agents' long-term effectiveness and safety compared with existing treatments. In summary, by targeting the RANKL pathway, denosumab provides a new approach for the treatment of postmenopausal osteoporosis (Table 3). Denosumab has a fundamentally different mechanism of action from that of bisphosphonates. This may explain differences in the degree and rapidity of reduction of bone resorption, their potential differential effects on trabecular and cortical bone, and the reversibility of their actions. In phase 3 studies, denosumab has shown efficacy against vertebral, nonvertebral, and hip fractures compared with placebo. Targeting the RANKL pathway represents a significant example of the rapid translation of advances in basic science into a novel clinical therapy. Whether this novel approach will result in further benefit for women with osteoporosis will be seen as clinicians gather experience with this new drug. Acknowledgments The authors drafted and wrote this review article. Writing/editing assistance was also provided by Reza Sayeed (Bioscript Stirling Ltd.) funded by Amgen (Europe) GmbH and GlaxoSmithKline. Graphics support was provided by Jennifer Keysor (Surfmedia). Thanks also to Antonia Panayi, Robert Stad, and Nathalie Franchimont of Amgen (Europe) GmbH and Paul Kostenuik and Cesar Libanati of Amgen Inc for providing many helpful discussions and comments during the preparation of this review. References [1] Schaffler MB. Role of bone turnover in microdamage. Osteoporos Int 2003;14 (Suppl 5):S73–7 (discussion S77–80). [2] Martin T, Gooi JH, Sims NA. Molecular mechanisms in coupling of bone formation to resorption. Crit Rev Eukaryot Gene Expr 2009;19:73–88. [3] Bruzzaniti A, Baron R. Molecular regulation of osteoclast activity. Rev Endocr Metab Disord 2006;7:123–39. [4] Russell G, Mueller G, Shipman C, Croucher P. Clinical disorders of bone resorption. Novartis Found Symp 2001;232:251–67 (discussion 267–71). [5] Khosla S, Riggs BL, Robb RA, Camp JJ, Achenbach SJ, Oberg AL, et al. Relationship of volumetric bone density and structural parameters at different skeletal sites to sex steroid levels in women. J Clin Endocrinol Metab 2005;90:5096–103. [6] Seeman E. Invited review: pathogenesis of osteoporosis. J Appl Physiol 2003;95: 2142–51. [7] Zebaze RM, Ghasem-Zadeh A, Bohte A, Iuliano-Burns S, Mirams M, Price RI, et al. Intracortical remodelling and porosity in the distal radius and post-mortem femurs of women: a cross-sectional study. Lancet 2010;375:1729–36. [8] Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, Riggs BL. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest 2003;111:1221–30. [9] Theill LE, Boyle WJ, Penninger JM. RANK-L and RANK: T cells, bone loss, and mammalian evolution. Annu Rev Immunol 2002;20:795–823. [10] Guerrini MM, Sobacchi C, Cassani B, Abinun M, Kilic SS, Pangrazio A, et al. Human osteoclast-poor osteopetrosis with hypogammaglobulinemia due to TNFRSF11A (RANK) mutations. Am J Hum Genet 2008;83:64–76. [11] Sobacchi C, Frattini A, Guerrini MM, Abinun M, Pangrazio A, Susani L, et al. Osteoclast-poor human osteopetrosis due to mutations in the gene encoding RANKL. Nat Genet 2007;39:960–2. [12] Cundy T, Davidson J, Rutland MD, Stewart C, DePaoli AM. Recombinant osteoprotegerin for juvenile Paget's disease. N Engl J Med 2005;353:918–23. [13] Bekker PJ, Holloway D, Nakanishi A, Arrighi M, Leese PT, Dunstan CR. The effect of a single dose of osteoprotegerin in postmenopausal women. J Bone Miner Res 2001;16: 348–60. [14] Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa S, Okamura H, et al. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature 1990;345:442–4. [15] Elford PR, Felix R, Cecchini M, Trechsel U, Fleisch H. Murine osteoblastlike cells and the osteogenic cell MC3T3-E1 release a macrophage colony-stimulating activity in culture. Calcif Tissue Int 1987;41:151–6. [16] Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998;93:165–76. [17] Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature 2003;423:337–42. [18] Asagiri M, Takayanagi H. The molecular understanding of osteoclast differentiation. Bone 2007;40:251–64. [19] Mori Y, Tsuji S, Inui M, Sakamoto Y, Endo S, Ito Y, et al. Inhibitory immunoglobulin-like receptors LILRB and PIR-B negatively regulate osteoclast development. J Immunol 2008;181:4742–51. [20] Takayanagi H. Osteoimmunology and the effects of the immune system on bone. Nat Rev Rheumatol 2009;5:667–76. [21] Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 1999;397:315–23. [22] Theoleyre S, Wittrant Y, Tat SK, Fortun Y, Redini F, Heymann D. The molecular triad OPG/RANK/RANKL: involvement in the orchestration of pathophysiological bone remodeling. Cytokine Growth Factor Rev 2004;15:457–75. [23] Wright HL, McCarthy HS, Middleton J, Marshall MJ. RANK, RANKL and osteoprotegerin in bone biology and disease. Curr Rev Musculoskelet Med 2009;2:56–64. [24] Kearns AE, Khosla S, Kostenuik PJ. Receptor activator of nuclear factor κB ligand and osteoprotegerin regulation of bone remodeling in health and disease. Endocr Rev 2008;29:155–92. R. Baron et al. / Bone 48 (2011) 677–692 [25] Fuller K, Owens JM, Jagger CJ, Wilson A, Moss R, Chambers TJ. Macrophage colony-stimulating factor stimulates survival and chemotactic behavior in isolated osteoclasts. J Exp Med 1993;178:1733–44. [26] Burgess TL, Qian Y, Kaufman S, Ring BD, Van G, Capparelli C, et al. The ligand for osteoprotegerin (OPGL) directly activates mature osteoclasts. J Cell Biol 1999;145:527–38. [27] Lacey DL, Tan HL, Lu J, Kaufman S, Van G, Qiu W, et al. Osteoprotegerin ligand modulates murine osteoclast survival in vitro and in vivo. Am J Pathol 2000;157: 435–48. [28] Niida S, Kaku M, Amano H, Yoshida H, Kataoka H, Nishikawa S, et al. Vascular endothelial growth factor can substitute for macrophage colony-stimulating factor in the support of osteoclastic bone resorption. J Exp Med 1999;190:293–8. [29] Niida S, Kondo T, Hiratsuka S, Hayashi S, Amizuka N, Noda T, et al. VEGF receptor 1 signaling is essential for osteoclast development and bone marrow formation in colony-stimulating factor 1-deficient mice. Proc Natl Acad Sci USA 2005;102: 14016–21. [30] Stewart AF, Vignery A, Silverglate A, Ravin ND, LiVolsi V, Broadus AE, et al. Quantitative bone histomorphometry in humoral hypercalcemia of malignancy: uncoupling of bone cell activity. J Clin Endocrinol Metab 1982;55:219–27. [31] Martin TJ, Sims NA. Osteoclast-derived activity in the coupling of bone formation to resorption. Trends Mol Med 2005;11:76–81. [32] Pierroz DD, Bonnet N, Baldock PA, Ominsky MS, Stolina M, Kostenuik PJ, et al. Are osteoclasts needed for the bone anabolic response to PTH? A study of intermittent PTH with denosumab or alendronate in knock-in mice expressing humanized RANKL. J Biol Chem 2010;285:28164–73. [33] Pederson L, Ruan M, Westendorf JJ, Khosla S, Oursler MJ. Regulation of bone formation by osteoclasts involves Wnt/BMP signaling and the chemokine sphingosine-1-phosphate. Proc Natl Acad Sci USA 2008;105:20764–9. [34] Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, et al. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab 2006;4:111–21. [35] Sims N. EPHs and ephrins: many pathways to regulate osteoblasts and osteoclasts. IBMS BoneKEy 2010;7:304–13. [36] Henriksen K, Neutzsky-Wulff AV, Bonewald LF, Karsdal MA. Local communication on and within bone controls bone remodeling. Bone 2009;44:1026–33. [37] Recker RR, Marin F, Ish-Shalom S, Moricke R, Hawkins F, Kapetanos G, et al. Comparative effects of teriparatide and strontium ranelate on bone biopsies and biochemical markers of bone turnover in postmenopausal women with osteoporosis. J Bone Miner Res 2009;24:1358–68. [38] Reid IR, Miller PD, Brown JP, Kendler DL, Fahrleitner-Pammer A, Valter I, et al. Effects of denosumab on bone histomorphometry: the FREEDOM and STAND studies. J Bone Miner Res 2010;25:2256–65. [39] Russell RG, Watts NB, Ebetino FH, Rogers MJ. Mechanisms of action of bisphosphonates: similarities and differences and their potential influence on clinical efficacy. Osteoporos Int 2008;19:733–59. [40] Hughes DE, MacDonald BR, Russell RGG, Gowen M. Inhibition of osteoclast-like cell formation by bisphosphonates in long-term cultures of human bone marrow. J Clin Invest 1989;83:1930–5. [41] Boonekamp PM, van der Wee-Pals LJ, van Wijk-van Lennep MM, Thesing CW, Bijvoet OL. Two modes of action of bisphosphonates on osteoclastic resorption of mineralized matrix. Bone Miner 1986;1:27–39. [42] Mabilleau G, Kwaasi AA, Sabokbar A, Tsoumpra M, Ebetino FH, Russell RGG. Bisphosphonates affect human osteoclast development in vitro. Bone 2010;46: S63–4 (Abstract 149). [43] Francis MD, Russell RG, Fleisch H. Diphosphonates inhibit formation of calcium phosphate crystals in vitro and pathological calcification in vivo. Science 1969;165:1264–6. [44] Benedict JJ. The physical chemistry of the disphosphonates — its relationship to their medical activity. In: Donath A, Courvoisier B, editors. Symposium CEMO (Centre d'Etude des Maladies Ostéo-articulaires de Genève) IV: diphosphonates and bone, Nyon (Switzerland), November 1–4, 1981. Geneva: Editions Médecine et Hygiène; 1982. p. 1–19. [45] Ebrahimpour A, Francis MD. Bisphosphonate therapy in acute and chronic bone loss: physical chemical considerations in bisphosphonate-related therapies. In: Bijvoet OLM, Fleisch HA, Canfield RE, Russell RGG, editors. Bisphosphonate on bones. Amsterdam: Elsevier; 1995. p. 125–36. [46] Fleisch H. Bisphosphonates in bone disease: from the laboratory to the patient. 4th ed. New York: Academic Press; 2000. [47] Henneman ZJ, Nancollas GH, Ebetino FH, Russell RG, Phipps RJ. Bisphosphonate binding affinity as assessed by inhibition of carbonated apatite dissolution in vitro. J Biomed Mater Res A 2008;85:993–1000. [48] Nancollas GH, Tang R, Phipps RJ, Henneman Z, Gulde S, Wu W, et al. Novel insights into actions of bisphosphonates on bone: differences in interactions with hydroxyapatite. Bone 2006;38:617–27. [49] Ebetino FH, Francis MD, Rogers MJ, Russell RGG. Mechanisms of action of etidronate and other bisphosphonates. Rev Contemp Pharmacother 1998;9:233–43. [50] Sato M, Grasser W, Endo N, Akins R, Simmons H, Thompson DD, et al. Bisphosphonate action. Alendronate localization in rat bone and effects on osteoclast ultrastructure. J Clin Invest 1991;88:2095–105. [51] Thompson K, Rogers MJ, Coxon FP, Crockett JC. Cytosolic entry of bisphosphonate drugs requires acidification of vesicles after fluid-phase endocytosis. Mol Pharmacol 2006;69:1624–32. [52] Coxon FP, Taylor A. Vesicular trafficking in osteoclasts. Semin Cell Dev Biol 2008;19:424–33. [53] van Beek E, Löwik C, Que I, Papapoulos S. Dissociation of binding and antiresorptive properties of hydroxybisphosphonates by substitution of the hydroxyl with an amino group. J Bone Miner Res 1996;11:1492–7. 689 [54] Frith JC, Mönkkönen J, Auriola S, Mönkkönen H, Rogers MJ. The molecular mechanism of action of the antiresorptive and antiinflammatory drug clodronate: evidence for the formation in vivo of a metabolite that inhibits bone resorption and causes osteoclast and macrophage apoptosis. Arthritis Rheum 2001;44:2201–10. [55] Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997;89:309–19. [56] Ominsky MS, Kostenuik PJ, Cranmer P, Smith SY, Atkinson JE. The RANKL inhibitor OPG-Fc increases cortical and trabecular bone mass in young gonadintact cynomolgus monkeys. Osteoporos Int 2007;18:1073–82. [57] Body JJ, Greipp P, Coleman RE, Facon T, Geurs F, Fermand JP, et al. A phase I study of AMGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases. Cancer 2003;97: 887–92. [58] Schwarz EM, Ritchlin CT. Clinical development of anti-RANKL therapy. Arthritis Res Ther 2007;9(Suppl 1):S7. [59] Sordillo EM, Pearse RN. RANK-Fc: a therapeutic antagonist for RANK-L in myeloma. Cancer 2003;97:802–12. [60] McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med 2006;354:821–31. [61] Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov Today 2006;11:81–8. [62] Kostenuik PJ, Nguyen HQ, McCabe J, Warmington KS, Kurahara C, Sun N, et al. Denosumab, a fully human monoclonal antibody to RANKL, inhibits bone resorption and increases BMD in knock-in mice that express chimeric (murine/ human) RANKL. J Bone Miner Res 2009;24:182–95. [63] Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S, Silverman C, et al. Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. J Biol Chem 1998;273:14363–7. [64] Amgen Europe BV. Prolia™ (denosumab) European Union summary of product characteristics. http://www.ema.europa.eu/docs/en_GB/document_library/ EPAR_-_Product_Information/human/001120/WC500093526.pdf. 26 May 2010 Accessed Sep 7, 2010. [65] Weinstein RS, Roberson PK, Manolagas SC. Giant osteoclast formation and longterm oral bisphosphonate therapy. N Engl J Med 2009;360:53–62. [66] Stolina M, Dwyer D, Middleton S, Adamu S, Ominsky MS, Zack D, et al. OPG prevents osteopenia in arthritic rats via different mechanisms than bisphosphonate therapy. [abstract]J Bone Miner Res 2007;22(Suppl 1):S475. [67] Tat SK, Padrines M, Theoleyre S, Couillaud-Battaglia S, Heymann D, Redini F, et al. OPG/membranous–RANKL complex is internalized via the clathrin pathway before a lysosomal and a proteasomal degradation. Bone 2006;39:706–15. [68] Lambert C, Oury C, Dejardin E, Chariot A, Piette J, Malaise M, et al. Further insights in the mechanisms of interleukin-1β stimulation of osteoprotegerin in osteoblastlike cells. J Bone Miner Res 2007;22:1350–61. [69] Samadfam R, Xia Q, Goltzman D. Co-treatment of PTH with osteoprotegerin or alendronate increases its anabolic effect on the skeleton of oophorectomized mice. J Bone Miner Res 2007;22:55–63. [70] Rogers MJ. New insights into the molecular mechanisms of action of bisphosphonates. Curr Pharm Des 2003;9:2643–58. [71] Gasser JA, Ingold P, Venturiere A, Shen V, Green JR. Long-term protective effects of zoledronic acid on cancellous and cortical bone in the ovariectomized rat. J Bone Miner Res 2008;23:544–51. [72] Teti A, Zallone A. Do osteocytes contribute to bone mineral homeostasis? Osteocytic osteolysis revisited. Bone 2009;44:11–6. [73] Bonewald LF, Johnson ML. Osteocytes, mechanosensing and Wnt signaling. Bone 2008;42:606–15. [74] Kamiya N, Ye L, Kobayashi T, Mochida Y, Yamauchi M, Kronenberg HM, et al. BMP signaling negatively regulates bone mass through sclerostin by inhibiting the canonical Wnt pathway. Development 2008;135:3801–11. [75] Kramer I, Halleux C, Keller H, Pegurri M, Gooi JH, Weber PB, et al. Osteocyte Wnt/ beta-catenin signaling is required for normal bone homeostasis. Mol Cell Biol 2010;30:3071–85. [76] Ominsky MS, Stolina M, Li X, Corbin TJ, Asuncion FJ, Barrero M, et al. One year of transgenic overexpression of osteoprotegerin in rats suppressed bone resorption and increased vertebral bone volume, density, and strength. J Bone Miner Res 2009;24:1234–46. [77] Li X, Ominsky MS, Stolina M, Warmington KS, Geng Z, Niu QT, et al. Increased RANK ligand in bone marrow of orchiectomized rats and prevention of their bone loss by the RANK ligand inhibitor osteoprotegerin. Bone 2009;45: 669–76. [78] Aguirre JI, Plotkin LI, Stewart SA, Weinstein RS, Parfitt AM, Manolagas SC, et al. Osteocyte apoptosis is induced by weightlessness in mice and precedes osteoclast recruitment and bone loss. J Bone Miner Res 2006;21:605–15. [79] Bonewald LF. Osteocytes as dynamic multifunctional cells. Ann NY Acad Sci 2007;1116:281–90. [80] Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC, Bellido T. Prevention of osteocyte and osteoblast apoptosis by bisphosphonates and calcitonin. J Clin Invest 1999;104:1363–74. [81] Plotkin LI, Manolagas SC, Bellido T. Dissociation of the pro-apoptotic effects of bisphosphonates on osteoclasts from their anti-apoptotic effects on osteoblasts/ osteocytes with novel analogs. Bone 2006;39:443–52. [82] Plotkin LI, Lezcano V, Thostenson J, Weinstein RS, Manolagas SC, Bellido T. Connexin 43 is required for the anti-apoptotic effect of bisphosphonates on osteocytes and osteoblasts in vivo. J Bone Miner Res 2008;23:1712–21. 690 R. Baron et al. / Bone 48 (2011) 677–692 [83] Plotkin LI, Goellner J, Vyas K, Shelton RS, Wynne RA, Weinstein RS, et al. A bisphosphonate analog that lacks anti-remodeling activity prevents osteocyte and osteoblast apoptosis in vivo. [abstract]J Bone Miner Res 2007;22(Suppl 1):S4. [84] Kogianni G, Mann V, Ebetino F, Nuttall M, Nijweide P, Simpson H, et al. Fas/CD95 is associated with glucocorticoid-induced osteocyte apoptosis. Life Sci 2004;75: 2879–95. [85] Follet H, Li J, Phipps RJ, Hui S, Condon K, Burr DB. Risedronate and alendronate suppress osteocyte apoptosis following cyclic fatigue loading. Bone 2007;40: 1172–7. [86] Roelofs AJ, Thompson K, Ebetino FH, Rogers MJ, Coxon FP. Bisphosphonates: molecular mechanisms of action and effects on bone cells, monocytes and macrophages. Curr Pharm Des 2010;16:2950–60. [87] Fuchs RK, Shea M, Durski SL, Winters-Stone KM, Widrick J, Snow CM. Individual and combined effects of exercise and alendronate on bone mass and strength in ovariectomized rats. Bone 2007;41:290–6. [88] Lespessailles E, Jaffre C, Beaupied H, Nanyan P, Dolleans E, Benhamou CL, et al. Does exercise modify the effects of zoledronic acid on bone mass, microarchitecture, biomechanics, and turnover in ovariectomized rats? Calcif Tissue Int 2009;85:146–57. [89] Stadelmann VA, Bonnet N, Pioletti DP. Combined effects of zoledronate and mechanical stimulation on bone adaptation in axially loaded mouse tibia. Clin Biomech 2011;26:101–5. [90] Dougall WC, Glaccum M, Charrier K, Rohrbach K, Brasel K, De Smedt T, et al. RANK is essential for osteoclast and lymph node development. Genes Dev 1999;13: 2412–24. [91] Bachmann MF, Wong BR, Josien R, Steinman RM, Oxenius A, Choi Y. TRANCE, a tumor necrosis factor family member critical for CD40 ligand-independent T helper cell activation. J Exp Med 1999;189:1025–31. [92] Ashcroft AJ, Cruickshank SM, Croucher PI, Perry MJ, Rollinson S, Lippitt JM, et al. Colonic dendritic cells, intestinal inflammation, and T cell-mediated bone destruction are modulated by recombinant osteoprotegerin. Immunity 2003;19:849–61. [93] Byrne FR, Morony S, Warmington K, Geng Z, Brown HL, Flores SA, et al. CD4+CD45RBHi T cell transfer induced colitis in mice is accompanied by osteopenia which is treatable with recombinant human osteoprotegerin. Gut 2005;54:78–86. [94] Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, et al. Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature 1999;402:304–9. [95] Romas E, Sims NA, Hards DK, Lindsay M, Quinn JW, Ryan PF, et al. Osteoprotegerin reduces osteoclast numbers and prevents bone erosion in collagen-induced arthritis. Am J Pathol 2002;161:1419–27. [96] Schett G, Middleton S, Bolon B, Stolina M, Brown H, Zhu L, et al. Additive boneprotective effects of anabolic treatment when used in conjunction with RANKL and tumor necrosis factor inhibition in two rat arthritis models. Arthritis Rheum 2005;52:1604–11. [97] Redlich K, Hayer S, Maier A, Dunstan CR, Tohidast-Akrad M, Lang S, et al. Tumor necrosis factor α-mediated joint destruction is inhibited by targeting osteoclasts with osteoprotegerin. Arthritis Rheum 2002;46:785–92. [98] Stolina M, Schett G, Dwyer D, Vonderfecht S, Middleton S, Duryea D, et al. RANKL inhibition by osteoprotegerin prevents bone loss without affecting local or systemic inflammation parameters in two rat arthritis models: comparison with anti-TNF-alpha or anti-IL-1 therapies. Arthritis Res Ther 2009;11:R187. [99] Verdrengh M, Bokarewa M, Ohlsson C, Stolina M, Tarkowski A. RANKL-targeted therapy inhibits bone resorption in experimental Staphylococcus aureus-induced arthritis. Bone 2010;46:752–8. [100] Ferrari-Lacraz S, Ferrari S. Do RANKL inhibitors (denosumab) affect inflammation and immunity? Osteoporos Int 2010;22:435–46. [101] Black DM, Delmas PD, Eastell R, Reid IR, Boonen S, Cauley JA, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med 2007;356:1809–22. [102] Bock O, Boerst H, Thomasius FE, Degner C, Stephan-Oelkers M, Valentine SM, et al. Common musculoskeletal adverse effects of oral treatment with once weekly alendronate and risedronate in patients with osteoporosis and ways for their prevention. J Musculoskelet Neuronal Interact 2007;7:144–8. [103] Delmas PD, Adami S, Strugala C, Stakkestad JA, Reginster JY, Felsenberg D, et al. Intravenous ibandronate injections in postmenopausal women with osteoporosis: one-year results from the dosing intravenous administration study. Arthritis Rheum 2006;54:1838–46. [104] Strampel W, Emkey R, Civitelli R. Safety considerations with bisphosphonates for the treatment of osteoporosis. Drug Saf 2007;30:755–63. [105] Thompson K, Rogers MJ. Statins prevent bisphosphonate-induced γ, δ-T-cell proliferation and activation in vitro. J Bone Miner Res 2004;19:278–88. [106] Raikkonen J, Crockett JC, Rogers MJ, Monkkonen H, Auriola S, Monkkonen J. Zoledronic acid induces formation of a pro-apoptotic ATP analogue and isopentenyl pyrophosphate in osteoclasts in vivo and in MCF-7 cells in vitro. Br J Pharmacol 2009;157:427–35. [107] Benzaid I, Clezardin P. Nitrogen-containing bisphosphonates and human γδ T cells. IBMS BoneKEy 2010;7:208–17. [108] Srivastava T, Haney CJ, Alon US. Atorvastatin may have no effect on acute phase reaction in children after intravenous bisphosphonate infusion. J Bone Miner Res 2009;24:334–7. [109] Hak AE, Pols HA, van Hemert AM, Hofman A, Witteman JC. Progression of aortic calcification is associated with metacarpal bone loss during menopause: a population-based longitudinal study. Arterioscler Thromb Vasc Biol 2000;20: 1926–31. [110] Szulc P, Kiel DP, Delmas PD. Calcifications in the abdominal aorta predict fractures in men: MINOS study. J Bone Miner Res 2008;23:95–102. [111] Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998;12:1260–8. [112] Bennett BJ, Scatena M, Kirk EA, Rattazzi M, Varon RM, Averill M, et al. Osteoprotegerin inactivation accelerates advanced atherosclerotic lesion progression and calcification in older ApoE−/− mice. Arterioscler Thromb Vasc Biol 2006;26:2117–24. [113] Helas S, Goettsch C, Schoppet M, Zeitz U, Hempel U, Morawietz H, et al. Inhibition of receptor activator of NF-κB ligand by denosumab attenuates vascular calcium deposition in mice. Am J Pathol 2009;175:473–8. [114] Fleisch HA, Russell RG, Bisaz S, Muhlbauer RC, Williams DA. The inhibitory effect of phosphonates on the formation of calcium phosphate crystals in vitro and on aortic and kidney calcification in vivo. Eur J Clin Invest 1970;1:12–8. [115] Price PA, Faus SA, Williamson MK. Bisphosphonates alendronate and ibandronate inhibit artery calcification at doses comparable to those that inhibit bone resorption. Arterioscler Thromb Vasc Biol 2001;21:817–24. [116] Price PA, Omid N, Than TN, Williamson MK. The amino bisphosphonate ibandronate prevents calciphylaxis in the rat at doses that inhibit bone resorption. Calcif Tissue Int 2002;71:356–63. [117] Monney P, Nguyen QV, Perroud H, Descombes E. Rapid improvement of calciphylaxis after intravenous pamidronate therapy in a patient with chronic renal failure. Nephrol Dial Transplant 2004;19:2130–2. [118] Schliep S, Schuler G, Kiesewetter F. Successful treatment of calciphylaxis with pamidronate. Eur J Dermatol 2008;18:554–6. [119] Santini D, Schiavon G, Angeletti S, Vincenzi B, Gasparro S, Grilli C, et al. Last generation of amino-bisphosphonates (N-BPs) and cancer angio-genesis: a new role for these drugs? Recent Pat Anticancer Drug Discov 2006;1:383–96. [120] Stresing V, Daubine F, Benzaid I, Monkkonen H, Clezardin P. Bisphosphonates in cancer therapy. Cancer Lett 2007;257:16–35. [121] Bekker PJ, Holloway DL, Rasmussen AS, Murphy R, Martin SW, Leese PT, et al. A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 2004;19:1059–66. [122] Rodan G, Reszka A, Golub E, Rizzoli R. Bone safety of long-term bisphosphonate treatment. Curr Med Res Opin 2004;20:1291–300. [123] Reitsma PH, Bijvoet OL, Verlinden-Ooms H, van der Wee-Pals LJ. Kinetic studies of bone and mineral metabolism during treatment with (3-amino-1-hydroxypropylidene)-1,1-bisphosphonate (APD) in rats. Calcif Tissue Int 1980;32: 145–57. [124] Bone HG, Hosking D, Devogelaer JP, Tucci JR, Emkey RD, Tonino RP, et al. Ten years' experience with alendronate for osteoporosis in postmenopausal women. N Engl J Med 2004;350:1189–99. [125] Tonino RP, Meunier PJ, Emkey R, Rodriguez-Portales JA, Menkes CJ, Wasnich RD, et al. Skeletal benefits of alendronate: 7-year treatment of postmenopausal osteoporotic women. Phase III Osteoporosis Treatment Study Group. J Clin Endocrinol Metab 2000;85:3109–15. [126] Delmas PD, Recker RR, Chesnut III CH, Skag A, Stakkestad JA, Emkey R, et al. Daily and intermittent oral ibandronate normalize bone turnover and provide significant reduction in vertebral fracture risk: results from the BONE study. Osteoporos Int 2004;15:792–8. [127] Sorensen OH, Crawford GM, Mulder H, Hosking DJ, Gennari C, Mellstrom D, et al. Long-term efficacy of risedronate: a 5-year placebo-controlled clinical experience. Bone 2003;32:120–6. [128] Ste-Marie LG, Sod E, Johnson T, Chines A. Five years of treatment with risedronate and its effects on bone safety in women with postmenopausal osteoporosis. Calcif Tissue Int 2004;75:469–76. [129] Lin JH. Bisphosphonates: a review of their pharmacokinetic properties. Bone 1996;18:75–85. [130] Khan SA, Kanis JA, Vasikaran S, Kline WF, Matuszewski BK, McCloskey EV, et al. Elimination and biochemical responses to intravenous alendronate in postmenopausal osteoporosis. J Bone Miner Res 1997;12:1700–7. [131] Cremers S, Sparidans R, den HJ, Hamdy N, Vermeij P, Papapoulos S. A pharmacokinetic and pharmacodynamic model for intravenous bisphosphonate (pamidronate) in osteoporosis. Eur J Clin Pharmacol 2002;57:883–90. [132] Capparelli C, Morony S, Warmington K, Adamu S, Lacey D, Dunstan CR, et al. Sustained antiresorptive effects after a single treatment with human recombinant osteoprotegerin (OPG): a pharmacodynamic and pharmacokinetic analysis in rats. J Bone Miner Res 2003;18:852–8. [133] Ominsky MS, Li X, Asuncion FJ, Barrero M, Warmington KS, Dwyer D, et al. RANKL inhibition with osteoprotegerin increases bone strength by improving cortical and trabecular bone architecture in ovariectomized rats. J Bone Miner Res 2008;23:672–82. [134] Calvi LM, Sims NA, Hunzelman JL, Knight MC, Giovannetti A, Saxton JM, et al. Activated parathyroid hormone/parathyroid hormone-related protein receptor in osteoblastic cells differentially affects cortical and trabecular bone. J Clin Invest 2001;107:277–86. [135] Ohishi M, Chiusaroli R, Ominsky M, Asuncion F, Thomas C, Khatri R, et al. Osteoprotegerin abrogated cortical porosity and bone marrow fibrosis in a mouse model of constitutive activation of the PTH/PTHrP receptor. Am J Pathol 2009;174:2160–71. [136] Jämsä T, Rho JY, Fan Z, MacKay CA, Marks Jr SC, Tuukkanen J. Mechanical properties in long bones of rat osteopetrotic mutations. J Biomech 2002;35:161–5. [137] Allen MR, Burr DB. Three years of alendronate treatment results in similar levels of vertebral microdamage as after one year of treatment. J Bone Miner Res 2007;22:1759–65. R. Baron et al. / Bone 48 (2011) 677–692 [138] Komatsubara S, Mori S, Mashiba T, Ito M, Li J, Kaji Y, et al. Long-term treatment of incadronate disodium accumulates microdamage but improves the trabecular bone microarchitecture in dog vertebra. J Bone Miner Res 2003;18:512–20. [139] Komatsubara S, Mori S, Mashiba T, Li J, Nonaka K, Kaji Y, et al. Suppressed bone turnover by long-term bisphosphonate treatment accumulates microdamage but maintains intrinsic material properties in cortical bone of dog rib. J Bone Miner Res 2004;19:999–1005. [140] Mashiba T, Hirano T, Turner CH, Forwood MR, Johnston CC, Burr DB. Suppressed bone turnover by bisphosphonates increases microdamage accumulation and reduces some biomechanical properties in dog rib. J Bone Miner Res 2000;15: 613–20. [141] Allen MR, Burr DB. Skeletal microdamage: less about biomechanics and more about remodeling. Clin Rev Bone Miner Metab 2008;6:24–30. [142] Burr DB, Allen MR. Low bone turnover and microdamage? How and where to assess it? J Bone Miner Res 2008;23:1150–1. [143] Chapurlat RD, Delmas PD. Bone microdamage: a clinical perspective. Osteoporos Int 2009;20:1299–308. [144] Dobnig H, Stepan JJ, Burr DB, Li J, Michalska D, Sipos A, et al. Teriparatide reduces bone microdamage accumulation in postmenopausal women previously treated with alendronate. J Bone Miner Res 2009;24:1998–2006. [145] Johnston S, Andrews S, Shen V, Cosman F, Lindsay R, Dempster DW, et al. The effects of combination of alendronate and human parathyroid hormone(1–34) on bone strength are synergistic in the lumbar vertebra and additive in the femur of C57BL/6J mice. Endocrinology 2007;148:4466–74. [146] Pettway GJ, Meganck JA, Koh AJ, Keller ET, Goldstein SA, McCauley LK. Parathyroid hormone mediates bone growth through the regulation of osteoblast proliferation and differentiation. Bone 2008;42:806–18. [147] Black DM, Greenspan SL, Ensrud KE, Palermo L, McGowan JA, Lang TF, et al. The effects of parathyroid hormone and alendronate alone or in combination in postmenopausal osteoporosis. N Engl J Med 2003;349:1207–15. [148] Hofbauer LC, Zeitz U, Schoppet M, Skalicky M, Schuler C, Stolina M, et al. Prevention of glucocorticoid-induced bone loss in mice by inhibition of RANKL. Arthritis Rheum 2009;60:1427–37. [149] Gerstenfeld LC, Sacks DJ, Pelis M, Mason ZD, Graves DT, Barrero M, et al. Comparison of effects of the bisphosphonate alendronate versus the RANKL inhibitor denosumab on murine fracture healing. J Bone Miner Res 2009;24: 196–208. [150] Ominsky MS, Schroeder J, Smith SY, Farrell DJ, Atkinson JE, Kostenuik PJ. Denosumab (AMG 162), a fully human RANKL antibody, improves cortical and cancellous bone mass and bone strength in ovariectomized cynomolgus monkeys. [abstract] J Bone Miner Res 2007;22(Suppl 1) [Abstract 1082]. [151] Ominsky M, Smith S, Jolette J, Vlasseros F, Samadfam R, Kostenuik P. Decreased fluorochrome labeling in bone biopsies from denosumab-treated cynomolgus monkeys is related to reductions in cortical porosity and trabecular eroded surfaces. [abstract] J Bone Miner Res 2009;24(Suppl 1) [Abstract A09001682]. [152] Ominsky MS, Jolette J, Smith SY, Vlasseros F, Samadfam R, Kostenuik PJ. Transition from alendronate to denosumab resulted in further reductions in local and systemic bone turnover parameters and reduced cortical porosity in ovariectomized cynomolgus monkeys. [abstract] J Bone Miner Res 2008;23(Suppl 1):S61. [153] Chesnut IC, Skag A, Christiansen C, Recker R, Stakkestad JA, Hoiseth A, et al. Effects of oral ibandronate administered daily or intermittently on fracture risk in postmenopausal osteoporosis. J Bone Miner Res 2004;19:1241–9. [154] Reginster J, Minne HW, Sorensen OH, Hooper M, Roux C, Brandi ML, et al. Randomized trial of the effects of risedronate on vertebral fractures in women with established postmenopausal osteoporosis. Vertebral Efficacy with Risedronate Therapy (VERT) Study Group. Osteoporos Int 2000;11:83–91. [155] Fogelman I, Ribot C, Smith R, Ethgen D, Sod E, Reginster JY. Risedronate reverses bone loss in postmenopausal women with low bone mass: results from a multinational, double-blind, placebo-controlled trial. BMD-MN Study Group. J Clin Endocrinol Metab 2000;85:1895–900. [156] Watts NB, Harris ST, Genant HK, Wasnich RD, Miller PD, Jackson RD, et al. Intermittent cyclical etidronate treatment of postmenopausal osteoporosis. N Engl J Med 1990;323:73–9. [157] Black DM, Cummings SR, Karpf DB, Cauley JA, Thompson DE, Nevitt MC, et al. Randomised trial of effect of alendronate on risk of fracture in women with existing vertebral fractures. Fracture Intervention Trial Research Group. Lancet 1996;348:1535–41. [158] Cummings SR, Black DM, Thompson DE, Applegate WB, Barrett-Connor E, Musliner TA, et al. Effect of alendronate on risk of fracture in women with low bone density but without vertebral fractures: results from the Fracture Intervention Trial. JAMA 1998;280:2077–82. [159] Liberman UA, Weiss SR, Broll J, Minne HW, Quan H, Bell NH, et al. Effect of oral alendronate on bone mineral density and the incidence of fractures in postmenopausal osteoporosis. The Alendronate Phase III Osteoporosis Treatment Study Group. N Engl J Med 1995;333:1437–43. [160] Cummings SR, Martin JS, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 2009;361:756–65. [161] Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, de Gregorio LH, et al. Comparison of the effect of denosumab and alendronate on BMD and biochemical markers of bone turnover in postmenopausal women with low bone mass: a randomized, blinded, phase 3 trial. J Bone Miner Res 2009;24:153–61. [162] Kendler DL, Roux C, Benhamou CL, Brown JP, Lillestol M, Siddhanti S, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res 2010;25: 72–81. 691 [163] Miller PD, Bolognese MA, Lewiecki EM, McClung MR, Ding B, Austin M, et al, AMG 162 Bone Loss Study Group. Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after long-term continued, discontinued, and restarting of therapy: a randomized blinded phase 2 clinical trial. Bone 2008;43:222–9. [164] Seeman E, Delmas PD, Hanley DA, Sellmeyer D, Cheung AM, Shane E, et al. Microarchitectural deterioration of cortical and trabecular bone: differing effects of denosumab and alendronate. J Bone Miner Res 2010;25:1886–94. [165] Genant HK, Engelke K, Hanley DA, Brown JP, Omizo M, Bone HG, et al. Denosumab improves density and strength parameters as measured by QCT of the radius in postmenopausal women with low bone mineral density. Bone 2010;47:131–9. [166] Ferrari S. Bisphosphonates and denosumab: do they thicken bone cortices, and can these changes be assessed by high-resolution pQCT? IBMS BoneKEy 2010;7:182–6. [167] Borah B, Dufresne T, Nurre J, Phipps R, Chmielewski P, Wagner L, et al. Risedronate reduces intracortical porosity in women with osteoporosis. J Bone Miner Res 2010;25:41–7. [168] Pazianas M, Cooper C, Ebetino FH, Russell RG. Long-term treatment with bisphosphonates and their safety in postmenopausal osteoporosis. Ther Clin Risk Manag 2010;6:325–43. [169] Marx RE. Pamidronate (Aredia) and zoledronate (Zometa) induced avascular necrosis of the jaws: a growing epidemic. J Oral Maxillofac Surg 2003;61:1115–7. [170] Almazrooa SA, Woo SB. Bisphosphonate and nonbisphosphonate-associated osteonecrosis of the jaw: a review. J Am Dent Assoc 2009;140:864–75. [171] Taylor KH, Middlefell LS, Mizen KD. Osteonecrosis of the jaws induced by antiRANK ligand therapy. Br J Oral Maxillofac Surg 2010;48:221–3. [172] Aghaloo TL, Felsenfeld AL, Tetradis S. Osteonecrosis of the jaw in a patient on denosumab. J Oral Maxillofac Surg 2010;68:959–63. [173] Kanis JA, Burlet N, Cooper C, Delmas PD, Reginster JY, Borgstrom F, et al. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int 2008;19:399–428. [174] Odvina CV, Zerwekh JE, Rao DS, Maalouf N, Gottschalk FA, Pak CY. Severely suppressed bone turnover: a potential complication of alendronate therapy. J Clin Endocrinol Metab 2005;90:1294–301. [175] Abrahamsen B, Eiken P, Eastell R. Subtrochanteric and diaphyseal femur fractures in patients treated with alendronate: a register-based national cohort study. J Bone Miner Res 2009;24:1095–102. [176] Green JR, Seltenmeyer Y, Jaeggi KA, Widler L. Renal tolerability profile of novel, potent bisphosphonates in two short-term rat models. Pharmacol Toxicol 1997;80: 225–30. [177] Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int 2008;74:1385–93. [178] Genentech Inc.. Boniva® (ibandronate sodium) tablets US prescribing information. http://www.gene.com/gene/products/information/boniva/pdf/pi.pdf. Jan 2010 Accessed Sep 7, 2010. [179] Genentech Inc. Boniva® (ibandronate sodium) injection US prescribing information. http://www.gene.com/gene/products/information/boniva/pdf/ Injection_pi.pdf. Mar 2010 Accessed Sep 7, 2010. [180] Merck and Co. Inc. Fosamax® (alendronate sodium) US prescribing information. http://www.merck.com/product/usa/pi_circulars/f/fosamax/fosamax_pi.pdf. Mar 2010 Accessed Sep 7, 2010. [181] Novartis Pharmaceuticals Corporation. Reclast® (zoledronic acid) US prescribing information. http://www.pharma.us.novartis.com/product/pi/pdf/reclast.pdf. Jan 2010 Accessed Sep 7, 2010. [182] Warner Chilcott Pharmaceuticals Inc.. Actonel® (risedronate sodium) US prescribing information. http://www.actonel.com/global/prescribing_information.pdf. Mar 2010 Accessed Sep 7, 2010. [183] Jamal SA, Bauer DC, Ensrud KE, Cauley JA, Hochberg M, Ishani A, et al. Alendronate treatment in women with normal to severely impaired renal function: an analysis of the fracture intervention trial. J Bone Miner Res 2007;22:503–8. [184] Miller PD, Roux C, Boonen S, Barton IP, Dunlap LE, Burgio DE. Safety and efficacy of risedronate in patients with age-related reduced renal function as estimated by the Cockcroft and Gault method: a pooled analysis of nine clinical trials. J Bone Miner Res 2005;20:2105–15. [185] Miller PD, Miday R, Klemes A, Ramsey D. Renal safety across a wide range of dosing regimens of risedronate. J Bone Miner Res 2008;23(Suppl 1):SA402. [186] Jamal SA, Ljunggren O, Stehman-Breen C, Cummings S, McClung M, Goemaere S, et al. The effects of denosumab on bone mineral density (BMD) and fracture by level of renal function. [abstract]Bone 2010;47(Suppl 1):S207–8. [187] Roelofs AJ, Coxon FP, Ebetino FH, Lundy MW, Henneman ZJ, Nancollas GH, et al. Fluorescent risedronate analogues reveal bisphosphonate uptake by bone marrow monocytes and localization around osteocytes in vivo. J Bone Miner Res 2010;25:606–16. [188] Ferrari S. Comparing and contrasting the effects of strontium ranelate and other osteoporosis drugs on microarchitecture. Osteoporos Int 2010;21(Suppl 2): S437–42. [189] Chavassieux PM, Arlot ME, Reda C, Wei L, Yates AJ, Meunier PJ. Histomorphometric assessment of the long-term effects of alendronate on bone quality and remodeling in patients with osteoporosis. J Clin Invest 1997;100: 1475–80. [190] Recker RR, Ste-Marie LG, Langdahl B, Czerwinski E, Bonvoisin B, Masanauskaite D, et al. Effects of intermittent intravenous ibandronate injections on bone quality and micro-architecture in women with postmenopausal osteoporosis: the DIVA study. Bone 2010;46:660–5. [191] Ominsky MS, Li X, Tan H, Asuncion FJ, Barrero M, Tian XY, et al. The effects of alendronate or denosumab on cortical and trabecular bone mass, bone strength, 692 R. Baron et al. / Bone 48 (2011) 677–692 and bone mass–strength relationships in mice. [abstract]J Bone Miner Res 2008;23(Suppl 1):S40. [192] Miller PD. Denosumab: anti-RANKL antibody. Curr Osteoporos Rep 2009;7:18–22. [193] Masarachia P, Weinreb M, Balena R, Rodan GA. Comparison of the distribution of 3H-alendronate and 3H-etidronate in rat and mouse bones. Bone 1996;19:281–90. [194] McClung MR. Inhibition of RANKL as a treatment for osteoporosis: preclinical and early clinical studies. Curr Osteoporos Rep 2006;4:28–33. [195] Hiraga T, Tanaka S, Yamamoto M, Nakajima T, Ozawa H. Inhibitory effects of bisphosphonate (YM175) on bone resorption induced by a metastatic bone tumor. Bone 1996;18:1–7.
© Copyright 2025 Paperzz