Available online at www.sciencedirect.com

ScienceDirect New therapeutics for osteoporosis Kong Wah Ng and T John Martin Two new approaches for the treatment of osteoporosis are summarized, each having arisen out of important new discoveries in bone biology. Odanacatib (ODN) inhibits the enzyme, cathepsin K, that is essential for the resorbing activity of osteoclasts. It is effective in preventing ovariectomy-induced bone loss in preclinical studies, and a phase II clinical study has shown inhibition of resorption sustained over five years. Outcome of a phase III study is awaited. The finding from mouse and human genetics that Wnt signaling is a powerful inducer of bone formation led to developments aimed at enhancing this pathway. Of the several approaches towards this, the most advanced is with a neutralizing antibody against sclerostin, the osteocyte-derived inhibitor of Wnt signaling. Preclinical studies show a powerful bone anabolic effect, and a clinical phase II study shows dose-dependent increases in bone formation and decreases in bone resorption markers. Addresses University of Melbourne, Department of Medicine, St. Vincent’s Institute of Medical Research, 9 Princes Street, Fitzroy 3065, Victoria, Australia Corresponding author: Martin, T John ([email protected])

Current Opinion in Pharmacology 2013, 16:58–63 This review comes from a themed issue on Musculoskeletal Edited by Alison Gartland and Lynne J Hocking

1471-4892/$ – see front matter, Published by Elsevier Ltd. http://dx.doi.org/10.1016/j.coph.2014.03.004

Introduction Understanding the intercellular control processes that regulate bone modelling and remodelling is essential in planning therapeutic approaches for prevention and treatment of bone fragility. New resorption inhibitors are being developed, based on mechanisms of action that are different from existing drugs. Some of these might offer resorption inhibition without reducing bone formation. Means are now available to promote the formation of new bone, and recent discoveries in bone biology show promise of new molecular targets for anabolic therapies. In this article we will discuss a novel resorption inhibitor, Odanacatib (ODN), and the application of neutralizing antibodies to sclerostin as an anabolic therapy. Remodelling of bone consists of a strict coupling of bone resorption and formation that continues throughout life and is necessary for the removal of damaged and old bone Current Opinion in Pharmacology 2014, 16:58–63

and to maintain normal bone structure [1–4,5]. The process begins with resorption of a volume of bone by osteoclasts, followed by new bone formation by osteoblasts. This process takes place in ‘bone multicellular units’ (BMUs) asynchronously throughout the skeleton. If the volume of bone resorbed exceeds the volume formed, bone loss results, leading to bone fragility and susceptibility to fracture. Intercellular communication processes are crucial in ensuring the balance that is achieved within BMUs throughout the skeleton. These communication pathways favour resorption by promoting osteoclast formation from haemopoietic precursors, or formation by promoting osteoblast differentiation from mesenchymal precursors. The latter activity within the BMU is referred to as the coupling of bone formation to resorption, and the development of the cathepsin K inhibitor, ODN, arose out of this concept of coupling.

ODN — a selective cathepsin K inhibitor Of the eleven different types of cathepsins [6], cathepsin K is the most important in the bone. It is predominantly expressed in osteoclasts and in actively resorbing osteoclasts, is localized at the ruffled border and discharged into the extracellular space when the lysosomal vesicles fuse with the cell membrane. It degrades the two main types of collagen I and II within the acidic microenvironment of resorption lacunae [7–9]. Cathepsin K cleaves the N-telopeptide of collagen to generate NTx and also degrades serum C-terminal telopeptide of type I collagen (1CTP), to generate CTx [10]. The inhibition of bone resorption observed in human and animal models deficient for cathepsin K [11,12,13,14,15,16] identified this enzyme as a suitable target for intervention by small molecules that might be used as therapeutic agents in osteoporosis [17,18,19]. With cathepsin K as target, the aim was direct inhibition of matrix degradation by osteoclasts without affecting either cell formation or survival. Indeed, in cathepsin K null mice the number of osteoclasts tends to be increased [17]. ODN (MK-0822, Merck) is a non-basic and nonlysosomotropic nitrile-based molecule displaying high potency for cathepsin K (IC50 = 0.2 nM). In studies of cathepsin K ( / ) mice or human osteoclast progenitors treated with ODN, loss of cathepsin K causes them to make small shallow resorption lacunae in contrast to untreated osteoclasts, which generate typical deep traillike resorption lacunae. While their resorption efficiency is reduced, ODN-treated osteoclasts can nonetheless continue to release osteogenic factors derived from the viable osteoclasts [17,20,21]. Of particular interest was the finding that in resorption-deficient cathepsin K / mice, www.sciencedirect.com

New therapeutics for osteoporosis Ng and Martin 59

bone formation was high in both trabecular and cortical bone [16]. In preclinical studies, ODN presented good pharmacokinetic parameters such as minimal in vitro metabolism, a long half-life, and oral bioavailability [22,23]. In ovariectomized rabbits, ODN was an effective inhibitor of resorption without inhibiting bone formation [B Pennybacker et al., abstract 1030, Annual Meeting of American Society for Bone and Mineral Research, San Diego, September 2011]. Osteoclast numbers on bone increased, but their resorption capacity was disabled, with shallow resorption pits evident. Monkey treatment studies show that ODN is an effective resorption inhibitor, which is dose-dependent [24,25]. Unlike conventional antiresorptives however, ODN treatment in the monkey resulted in a compartmentspecific increase in bone formation, with increased periosteal bone formation and cortical thickness whereas trabecular bone formation was reduced, alongside reduced bone resorption. Thus the drug did not reproduce the preservation of bone formation as seen in the cathepsin K / mice or in the ODN-treated rabbits, and currently the increased periosteal bone formation in the monkey study has no ready explanation.

Clinical studies of ODN Phase I studies indicated safety of ODN, and a once-daily regimen showed no obvious advantage over a once-weekly regimen [26,27]. In phase II studies, when analysed for up to five years [28–30], subjects showed almost linear increases in bone mineral density (BMD) from baseline at the lumbar spine (11.9%), femoral neck (9.8%) and total hip (8.5%). The bone resorption effect was maintained through five years of treatment, while bone formation was only slightly reduced relative to baseline. Reversibility of ODN effect after stopping treatment was striking. After ODN discontinuation at month 24, bone turnover markers increased transiently before returning to baseline by month 36 [30]. Such rapid reversibility observed upon stopping treatment was similar to that seen with estrogens, selective estrogen receptor modulators (SERMs) and denosumab, but not oral or intravenous bisphosphonates [31–35]. There is as yet, no published fracture data for ODN. An international multicentre placebo-controlled phase III fracture outcome trial that enrolled more than 16,000 postmenopausal women taking 50 mg ODN once weekly in a double-blind treatment (ClinicalTrials.gov registration number: NCT00529373) is completed and the results are anticipated in 2014. The large phase III placebo-controlled study with ODN will also provide important insight regarding safety and tolerability. There are two other ongoing phase III studies with ODN, one in www.sciencedirect.com

postmenopausal women (ClinicalTrials.gov registration number: NCT00729183) and the other in men (NCT0112600). These ongoing fracture outcome studies are necessary to determine whether the increased bone mass demonstrated in the phase II studies is translated into better fracture risk reduction [17,36]. If cathepsin K inhibitors are safe and at least as effective in fracture reduction as other inhibitors, they could offer theoretical advantages over bisphosphonates. For instance, ODN and other selective cathepsin K inhibitors might be more effectively combined with anabolic therapy such as parathyroid hormone (PTH), than resorption inhibitors that lead to inhibition of bone formation [B Pennybacker et al., abstract 1030, Annual Meeting of American Society for Bone and Mineral Research, San Diego, September 2011]. While cathepsin K is highly expressed in osteoclasts, very low levels exist in other adult tissues, including the heart, liver, skeletal muscle, placenta, ovary, testis, small intestine, colon as well as embryonic lung and neonatal dermal fibroblasts. It is also expressed in synovial fibroblasts and macrophages of rheumatoid arthritic joints, breast and prostate tumors. Therefore inhibition of cathepsin K could affect the turnover of type I collagen in organs other than bone [18]. While there is good potential for ODN to be a useful treatment for osteoporosis, rheumatoid arthritis and metastatic bone disease, the clinician will need to be vigilant for potential off-target effects.

Anabolic therapy The first opportunity to restore bone after it had been lost came with the development of PTH as a highly effective anabolic therapy for the skeleton, despite its betterknown action as a resorptive hormone. The approved therapies in several countries are PTH(1–34) and PTH(1–84). The anabolic effectiveness of PTH requires that it be administered intermittently, with daily injections that rapidly achieve a peak level in blood, which is not maintained [37,38]. Studies of PTH pre-treatment and post-treatment bone biopsies in women indicated that the predominant PTH effect was to increase remodelling, with some lesser effect on modelling [39,40]. Current views on the anabolic action of PTH are that it increases the activation of the BMUs in remodelling, that it acts on committed osteoblast precursors to promote their differentiation, inhibits osteoblast and osteocyte apoptosis [41], and inhibits the production of the bone formation inhibitor, sclerostin [42]. Special attention is being paid to activation of the Wnt signaling pathway to achieve an anabolic effect in bone. The first link between Wnt signaling and human bone disease came from observations that inactivating mutations in the low density lipoprotein receptor-related protein 5 (LRP5) cause the osteoporosis-pseudoglioma syndrome (OPPG, OMIM 259770) characterized by Current Opinion in Pharmacology 2014, 16:58–63

60 Musculoskeletal

severely decreased bone mass [43]. Conversely, a syndrome of high bone mass was found to be caused by a gain-of-function mutation of LRP5 (OMIM 601884) [44]. These genetic syndromes were reproduced with the appropriate genetic manipulations in mice [45,46]. The Wnt/b-catenin signaling pathway offers several targets that may be suitable for pharmacological intervention at a number of specific points. These include extracellular agonists and the points of interaction of antagonists. The primary aim of these interventions is to increase Wnt/b-catenin canonical signaling (see http:// www.stanford.edu/group/nusselab/cgi-bin/wnt/) in order to increase bone mass. This has been achieved in animal models with the inhibition of dickkopf-1 (DKK-1), glycogen synthase kinase-3b (GSK-3b), or of sclerostin, as described below. Inhibition of DKK-1

LRP5 can form a ternary complex with DKK-1 and Kremen (a receptor for DKK), which triggers rapid internalization and depletion of LRP5, leading to inhibition of the canonical Wnt signaling pathway. Inhibition of the interactions between DKK-1 and LRP5 would release LRP5 to activate the Wnt pathway. Genetic studies with mice lacking a single allele of DKK-1 showed a markedly increased trabecular bone volume and elevated trabecular bone formation rate [47]. The production of DKK-1 by multiple myeloma cells has been invoked as a contributing factor to the reduced bone formation in the lytic bone lesions of myeloma [48]. A study using antibodies raised against DKK-1 in the treatment of a mouse model of multiple myeloma showed increased numbers of osteoblasts, reduced number of osteoclasts, and reduced myeloma burden in the antibody-treated mice [49]. Initial drug development attempts in osteoporosis also use monoclonal anti-DKK-1 reagents, but the possibility of directing small molecules to prevent the DKK-1–LRP5 interaction is being explored. Inhibition of GSK-3

Inhibition of GSK-3 would prevent the phosphorylation of b-catenin, leading to stabilization of b-catenin independently of Wnt interactions with the receptor complex. Mice treated with lithium chloride as a GSK-3 inhibitor showed increased bone formation and bone mass [50]. Treatment of ovariectomized rats with an orally active dual GSK a/b inhibitor, LY603281-31-8, for two months resulted in an increase in the number of trabeculae and connectivity as well as trabecular area and thickness. BMD at cancellous and cortical sites was increased, and this was associated with increased bone formation on histomorphometry and increased strength [51]. Inhibition of sclerostin

Sclerostin, the protein product of SOST, is produced in bone exclusively by osteocytes and is a circulating inhibitor Current Opinion in Pharmacology 2014, 16:58–63

of the Wnt-signaling pathway that achieves this by binding to LRP5 and LRP6 [52]. High bone density in sclerosteosis is caused by an inactivating mutation in SOST gene (OMIM 607363) [53]. Inhibition of production or action of sclerostin resulting in enhanced Wnt canonical signaling was predicted to lead to increased bone mass, and did so with impressive effect in preclinical studies, where monoclonal antibody against sclerostin has been shown to promote bone formation rapidly in monkeys and ovariectomized rats. Considerable increases in bone formation rates and in the amounts of trabecular bone took place rapidly without increases in resorption parameters [54]. At the time of writing, a phase I study of anti-sclerostin (AMG 785, Amgen) had been published [55], in which healthy men and women were treated for up to 85 days with escalating doses of AMG 785, resulting in doserelated increases in bone formation markers and a decrease in the resorption marker, serum CTx. The latter observation might relate to changes in osteoblast differentiation, with less cells of the osteoblast lineage available for presentation of the critical osteoclast differentiation factor, Receptor Activator of Nuclear Factor kB Ligand (RANKL), to osteoclast precursors. In this short study, BMD increased significantly at the spine (5.3%) and hip (2.8%), with five subjects at the highest dose developing detectable antibodies, two of which were neutralizing. The treatment with AMG 785 was generally well tolerated, and further clinical studies will be expected with what appears to be a most promising way of increasing the amount of bone. Of particular interest is the fact that PTH rapidly reduces sclerostin mRNA and protein production by osteoblasts in vitro and in bone in vivo [42,56] raising the possibility that transient reduction of sclerostin output by osteocytes in response to intermittent PTH could mediate enhanced osteoblast differentiation and bone formation [57] and reduced osteoblast apoptosis [58]. Such a mechanism would offer real possibilities as a drug target, and the mechanism of this inhibition is all the more interesting with the recent finding [59] that the cyclic AMPmediated effect of PTH to diminish sclerostin production operates through a long range enhancer, MEF2, the discovery of which came from the pursuit of the nature of the van Buchem’s disease mutation. There may be small molecule approaches amenable to sclerostin regulation, in addition to antibody neutralization of its activity. Safety and specificity

Any manipulation of the Wnt canonical signaling pathway will need to be shown to be safe, and that its action can be targeted specifically to bone. Wnt proteins are critical signaling proteins involved in developmental biology, with roles in early axis specification, brain patterning, intestinal development, and limb development. In adults, Wnt proteins play a vital role in tissue maintenance, with www.sciencedirect.com

New therapeutics for osteoporosis Ng and Martin 61

aberrations in Wnt signaling leading to diseases such as adenomatous polyposis [60]. Inhibition of GSK-3 results in increased cyclin D1, cyclin E, and c-Myc, and overexpression of these cell cycle regulators has been linked to tumor formation [61]. All relevant possibilities of side effects of enhanced Wnt signaling need to be kept in mind throughout preclinical studies. Exciting possibilities of new anabolic therapies are becoming more evident. The early evidence that it might be possible to develop resorption inhibitors that can uncouple bone resorption from bone formation is exciting, potentially offering an advantage over currently available antiresorptive agents. Nonetheless, all these predictions are based largely on preclinical data, and it is hoped that properly conducted clinical trials in the coming years will see the emergence of new therapies that are effective, durable, and safe, at an affordable cost.

References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest

11. Maroteaux P, Lamy M: 2 cases of a condensing osseous disease: pycnodysostosis. Arch Fr Pediatr 1962, 19:267-274. 12. Gelb BD, Shi GP, Chapman HA, Desnick RJ: Pycnodysostosis, a  lysosomal disease caused by cathepsin K deficiency. Science 1996, 273:1236-1238. Description of the bone abnormalities of cathepsin K deficiency in man. 13. Saftig P, Hunziker E, Wehmeyer O, Jones S, Boyde A, Rommerskirch W, Moritz JD, Schu P, von Figura K: Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin-K-deficient mice. Proc Natl Acad Sci USA 1998, 95:13453-13458. 14. Gowen M, Lazner F, Dodds R, Kapadia R, Feild J, Tavaria M, Bertoncello I, Drake F, Zavarselk S, Tellis I et al.: Cathepsin K  knockout mice develop osteopetrosis due to a deficit in matrix degradation but not demineralization. J Bone Miner Res 1999, 14:1654-1663. Useful early paper on the impaired bone resorption of cathepsin K deficiency. 15. Chen W, Yang S, Abe Y, Li M, Wang Y, Shao J, Li E, Li YP: Novel pycnodysostosis mouse model uncovers cathepsin K function as a potential regulator of osteoclast apoptosis and senescence. Hum Mol Genet 2007, 16:410-423. 16. Pennypacker B, Shea M, Liu Q, Masarachia P, Saftig P, Rodan S,  Rodan G, Kimmel D: Bone density, strength, and formation in adult cathepsin K (S/S) mice. Bone 2009, 44:199-207. Mice null for cathepsin K gene have decreased bone resorption without decrease in bone formation. 17. Duong Le T: Therapeutic inhibition of cathepsin K — reducing bone resorption while maintaining bone formation. Bonekey Rep 2012, 1:67.

1.

Frost HM: Dynamics of bone remodeling. Bone Biodynamics Little. Boston: Brown; 1964, 315-333.

18. Ng KW: Potential role of odanacatib in the treatment of osteoporosis. Clin Interv Aging 2012, 7:235-247.

2.

Parfitt AM: The coupling of bone formation to bone resorption: a critical analysis of the concept and of its relevance to the pathogenesis of osteoporosis. Metab Bone Dis Relat Res 1982, 4:1-6.

19. Zerbini CA, McClung MR: Odanacatib in postmenopausal  women with low bone mineral density: a review of current clinical evidence. Ther Adv Musculoskelet Dis 2013, 5:199-209. Evaluation of current status clinical studies of cathepsin K inhibition.

3.

Eriksen EF: Normal and pathological remodeling of human trabecular bone: three dimensional reconstruction of the remodeling sequence in normals and in metabolic bone disease. Endocr Rev 1986, 7:379-408.

20. Baron R, Ferrari S, Russell RG: Denosumab and bisphosphonates: different mechanisms of action and effects. Bone 2011, 48:677-692.

4.

Hauge EM, Qvesel D, Eriksen EF, Mosekilde L, Melsen F: Cancellous bone remodeling occurs in specialized compartments lined by cells expressing osteoblastic markers. J Bone Miner Res 2001, 16:1575-1582.

5. 

Martin T, Gooi JH, Sims NA: Molecular mechanisms in coupling of bone formation to resorption. Crit Rev Eukaryot Gene Expr 2009, 19:73-88. This review discusses mechanisms of cell communication in bone remodeling and particularly the ways in which osteoclasts might contribute to bone formation in the coupling process.

6.

Reiser J, Adair B, Reinheckel T: Specialized roles for cysteine cathepsins in health and disease. J Clin Invest 2010, 120:3421-3431.

7.

Yamaza T, Goto T, Kamiya T, Kobayashi Y, Sakai H, Tanaka T: Study of immunoelectron microscopic localization of cathepsin K in osteoclasts and other bone cells in the mouse femur. Bone 1998, 23:499-509.

8.

Va¨a¨ra¨niemi J, Halleen JM, Kaarlonen K, Ylipahkala H, Alatalo SL, Andersson G, Kaija H, Vihko P, Va¨a¨na¨nen HK: Intracellular machinery for matrix degradation in bone-resorbing osteoclasts. J Bone Miner Res 2004, 19:1432-1440.

9.

Xia L, Kilb J, Wex H, Li Z, Lipyansky A, Breuil V, Stein L, Palmer JT, Dempster DW, Bro¨mme D: Localization of rat cathepsin K in osteoclasts and resorption pits: inhibition of bone resorption and cathepsin K-activity by peptidyl vinyl sulfones. Biol Chem 1999, 380:679-687.

10. Garnero P, Borel O, Byrjalsen I, Ferreras M, Drake FH, McQueney MS, Foged NT, Delmas PD, Delaisse´ JM: The collagenolytic activity of cathepsin K is unique among mammalian proteinases. J Biol Chem 1998, 273:32347-32352. www.sciencedirect.com

21. Leung P, Pickarski M, Zhuo Y, Masarachia PJ, Duong LT: The effects of the cathepsin K inhibitor odanacatib on osteoclastic bone resorption and vesicular trafficking. Bone 2011, 49:623-635. 22. Gauthier JY, Chauret N, Cromlish W: The discovery of odanacatib (MK-0822), a selective inhibitor of cathepsin K. Bioorg Med Chem Lett 2008, 18:923-928. 23. Podgorski I: Future of anticathepsin K drugs: dual therapy for skeletal disease and atherosclerosis? Future Med Chem 2009, 1:21-34. 24. Cusick T, Chen CM, Pennypacker BL, Pickarski M, Kimmel DB, Scott BB: Duong le T: odanacatib treatment increases hip bone mass and cortical thickness by preserving endocortical bone formation and stimulating periosteal bone formation in the ovariectomized adult rhesus monkey. J Bone Miner Res 2012, 27:524-537. 25. Masarachia PJ, Pennypacker BL, Pickarski M, Scott KR, Wesolowski GA, Smith SY, Samadfam R, Goetzmann JE, Scott BB, Kimmel DB et al.: Odanacatib reduces bone turnover and increases bone mass in the lumbar spine of skeletally mature ovariectomized rhesus monkeys. J Bone Miner Res 2012, 27:509-523. 26. Rodan SB, Duong LT: Cathepsin K — a new molecular target for osteoporosis. IBMS BoneKEy 2008, 5:16-24. 27. Stoch SA, Zajic S, Stone J, Miller DL, Van Dyck K, Gutierrez MJ, De Decker M, Liu L, Liu Q, Scott BB et al.: Effect of the cathepsin K inhibitor odanacatib on bone resorption biomarkers in healthy postmenopausal women: two double-blind, randomized, placebo-controlled phase I studies. Clin Pharmacol Ther 2009, 86:175-182.

Current Opinion in Pharmacology 2014, 16:58–63

62 Musculoskeletal

28. Bone HG, McClung MR, Roux C, Recker RR, Eisman JA, Verbruggen N, Hustad CM, DaSilva C, Santora AC, Ince BA: Odanacatib, a cathepsin-K inhibitor for osteoporosis: a twoyear study in postmenopausal women with low bone density. J Bone Miner Res 2010, 25:937-947. 29. Brixen K, Chapurlat R, Cheung AM, Keaveny TM, Fuerst T, Engelke K, Recker R, Dardzinski B, Verbruggen N, Ather S et al.: Bone density, turnover, and estimated strength in postmenopausal women treated with odanacatib: a randomized trial. J Clin Endocrinol Metab 2013, 98:571-580. 30. Eisman JA, Bone HG, Hosking DJ, McClung MR, Reid IR, Rizzoli R, Resch H, Verbruggen N, Hustad CM, DaSilva C et al.: Odanacatib in the treatment of postmenopausal women with low bone mineral density: three-year continued therapy and resolution of effect. J Bone Miner Res 2011, 26:242-251. 31. Neele SJ, Evertz R, De Valk-De Roo G, Roos JC, Netelenbos JC: Effect of 1 year of discontinuation of raloxifene or estrogen therapy on bone mineral density after 5 years of treatment in healthy postmenopausal women. Bone 2002, 30:599-603. 32. Khosla S, Burr D, Cauley J, Dempster DW, Ebeling PR, Felsenberg D, Gagel RF, Gilsanz V, Guise T, Koka S et al.: American Society for Bone and Mineral Research. Bisphosphonate-associated osteonecrosis of the jaw: report of a task force of the American Society for Bone and Mineral Research. J Bone Miner Res 2007, 22:1479-1491. 33. Miller PD, Bolognese MA, Lewiecki EM, McClung MR, Ding B, Austin M, Liu Y, San Martin J, Amg 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, 43:222-229. 34. Bone HG, Bolognese MA, Yuen CK, Kendler DL, Miller PD, Yang YC, Grazette L, San Martin J, Gallagher JC: Effects of denosumab treatment and discontinuation on bone mineral density and bone turnover markers in postmenopausal women with low bone mass. J Clin Endocrinol Metab 2011, 96:972-980. 35. Grey A, Bolland MJ, Wattie D, Horne A, Gamble G, Reid IR: The antiresorptive effects of a single dose of zoledronate persist for two years: a randomized, placebo-controlled trial in osteopenic postmenopausal women. J Clin Endocrinol Metab 2009, 94:538-544. 36. Costa AG, Cusano NE, Silva BC, Cremers S, Bilezikian JP: Cathepsin K: its skeletal actions and role as a therapeutic target in osteoporosis. Nat Rev Rheumatol 2011, 7:447-456. 37. Frolik CA, Black EC, Cain RL, Satterwhite JH, BrownAugsburger PL, Sato M, Hock JM: Anabolic and catabolic bone effects of human parathyroid hormone (1–34) are predicted by duration of hormone exposure. Bone 2003, 33:372-379. 38. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK et al.: Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001, 344:1434-1441. 39. Misof BM, Roschger P, Cosman F, Kurland ES, Tesch W, Messmer P, Dempster DW, Nieves J, Shane E, Fratzl P et al.: Effects of intermittent parathyroid hormone administration on bone mineralization density in iliac crest biopsies from patients with osteoporosis: a paired study before and after treatment. J Clin Endocrinol Metab 2003, 88:1150-1156. 40. Ma YL, Zeng Q, Donley DW, Ste-Marie LG, Gallagher JC, Dalsky GP, Marcus R, Eriksen EF: Teriparatide increases bone formation in modeling and remodeling osteons and enhances IGF-II immunoreactivity in postmenopausal women with osteoporosis. J Bone Miner Res 2006, 21:855-864. 41. Jilka RL: Molecular and cellular mechanisms of the anabolic effect of intermittent PTH. Bone 2007, 40:1434-1446. 42. Keller H, Kneissel M: SOST is a target gene for PTH in bone. Bone 2005, 37:148-158. Current Opinion in Pharmacology 2014, 16:58–63

43. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D et al.:  Osteoporosis-Pseudoglioma Syndrome Collaborative Group. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 2001, 107:513-523. Genetic abnormality identifying the Wnt pathway role in bone formation. 44. Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, Wu D,  Insogna K, Lifton RP: High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 2002, 346:1513-1521. Another very important report identifying genetic abnormality showing importance of Wnt signaling effect in bone. 45. Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA 2nd, Hartmann C, Li L, Hwang TH, Brayton CF et al.: Cbfa1independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol 2002, 157:303-314. 46. Babij P, Zhao W, Small C, Kharode Y, Yaworsky PJ, Bouxsein ML, Reddy PS, Bodine PV, Robinson JA, Bhat B et al.: High bone mass in mice expressing a mutant LRP5 gene. J Bone Miner Res 2003, 18:960-974. 47. Morvan F, Boulukos K, Cle´ment-Lacroix P, Roman Roman S, SucRoyer I, Vayssie`re B, Ammann P, Martin P, Pinho S, Pognonec P, Mollat P et al.: Deletion of a single allele of the Dkk1 gene leads to an increase in bone formation and bone mass. J Bone Miner Res 2006, 21:934-945. 48. Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B, Shaughnessy JD Jr: The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med 2003, 349:2483-2494. 49. Yaccoby S, Ling W, Zhan F, Walker R, Barlogie B, Shaughnessy JD Jr: Antibody-based inhibition of DKK1 suppresses tumorinduced bone resorption and multiple myeloma growth in vivo. Blood 2007, 109:2106-2111. 50. Cle´ment-Lacroix P, Ai M, Morvan F, Roman-Roman S,  Vayssie`re B, Belleville C, Estrera K, Warman ML, Baron R, Rawadi G: Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci USA 2005, 102:17406-17411. Illustrative of a low molecular weight drug approach to stimulating Wnt signaling and enhancing bone formation. 51. Kulkarni NH, Onyia JE, Zeng Q, Tian X, Liu M, Halladay DL, Frolik CA, Engler T, Wei T, Kriauciunas A et al.: Orally bioavailable GSK-3alpha/beta dual inhibitor increases markers of cellular differentiation in vitro and bone mass in vivo. J Bone Miner Res 2006, 21:910-920. 52. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D: Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem 2005, 280:19883-19887. 53. Ott SM: Sclerostin and Wnt signaling — the pathway to bone strength. J Clin Endocrinol Metab 2005, 90:6741-6743. 54. Li X, Warmington KS, Niu QT, Asuncion FJ, Barrero M, Grisanti M,  Dwyer D, Stouch B, Thway TM, Stolina M et al.: Inhibition of sclerostin by monoclonal antibody increases bone formation, bone mass, and bone strength in aged male rats. J Bone Miner Res 2010, 25:2647-2656. Shows very powerful bone anabolic effect of antibody against sclerostin in osteoporotic rats. 55. Padhi D, Jang G, Stouch B, Fang L, Posvar E: Single-dose,  placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J Bone Miner Res 2011, 26:19-26. Phase I clinical study of anti-sclerostin. 56. Silvestrini G, Ballanti P, Leopizzi M, Sebastiani M, Berni S, Di Vito M, Bonucci E: Effects of intermittent parathyroid hormone (PTH) administration on SOST mRNA and protein in rat bone. J Mol Histol 2007, 38:261-269. 57. van Bezooijen RL, ten Dijke P, Papapoulos SE, Lo¨wik CW: SOST/ sclerostin, an osteocyte-derived negative regulator of bone formation. Cytokine Growth Factor Rev 2005, 16:319-327. www.sciencedirect.com

New therapeutics for osteoporosis Ng and Martin 63

58. Sutherland MK, Geoghegan JC, Yu C, Winkler DG, Latham JA: Unique regulation of SOST, the sclerosteosis gene, by BMPs and steroid hormones in human osteoblasts. Bone 2004, 35:448-454. 59. Leupin O, Kramer I, Collette NM, Loots GG, Natt F, Kneissel M, Keller H: Control of the SOST bone enhancer by PTH using MEF2 transcription factors. J Bone Miner Res 2007, 22:1957-1967.

www.sciencedirect.com

60. Krishnan V, Bryant HU, Macdougald OA: Regulation of bone mass by Wnt signaling. J Clin Invest 2006, 116:1202-1209. 61. Dong J, Peng J, Zhang H, Mondesire WH, Jian W, Mills GB, Hung MC, Meric-Bernstam F: Role of glycogen synthase kinase 3beta in rapamycin-mediated cell cycle regulation and chemosensitivity. Cancer Res 2005, 65:1961-1972.

Current Opinion in Pharmacology 2014, 16:58–63

New therapeutics for osteoporosis.

Two new approaches for the treatment of osteoporosis are summarized, each having arisen out of important new discoveries in bone biology. Odanacatib (...
249KB Sizes 0 Downloads 3 Views