OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES Jeffrey A. Dodge Henry U. Bryant Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 1. INTRODUCTION Bone is a living, dynamic tissue that is continuously remodeled during the adult life of an individual. The remodeling process occurs in quantum units called bone-remodeling units [1,2] through the action of osteoclasts and osteoblasts. Osteoclasts are the bone-resorbing cells, which tightly adhere to the bone surface and then secrete acid that dissolves the hydroxyapatite mineral and proteolytic enzymes that degrade the organic matrix of bone. Osteoblasts are the bone-forming cells that synthesize a highly cross-linked, lamellar organic matrix (osteoid) that becomes mineralized by extracellular processes. Osteoblasts usually replenish the bone excavated by osteoclasts. Osteoporosis is a disease of the bone that leads to increased risk of fracture as a consequence of an imbalance between osteoclastic and osteoblastic activities, coupled with an increased rate of bone turnover observed. That is, a net loss of bone mass or inadequate architecture results due to either the excessive bone-resorbing activity of osteoclasts or the impaired bone-forming activity of osteoblasts, such that osteoblasts do not optimally replenish the lost bone. For women, this phenomenon is related to the decline of endogenous levels of the steroid hormone estrogen after menopause. Because the rate of remodeling is approximately 10 times higher in cancellous bone than cortical bone, bone loss following menopause is observed primarily in regions enriched for trabecular bone such as the vertebra and proximal femur. Gradually, perforations in or thinning of the trabecular bone spicules develop with the result that a weakened and inadequate architecture ensues. Osteoporosis is currently defined by the World Health Organization as a condition observed for patients with spinal bone mineral density (BMD) of less than 2.5 standard deviations below the mean of young, normal adults of the same gender [3,4]. Osteoporosis is an ailment of increasing concern among elderly women and men in which bone has been lost to the extent that too little remains to support the mechanical usage requirements of the individual's activities. As a result, these individuals are at risk for spontaneous, atraumatic (or mild trauma) fractures. The inverse relationship between densitometric measures of bone mass and fracture risk was clearly shown for peri- and postmenopausal women in the process of losing bone due to declining levels of circulating estrogens [5-7]. Postmenopausal or type I osteoporosis is observed with escalating frequency in women elder than 50 years of age such that elderly women have a lifetime risk of fractures of approximately 75% [8,9]. At any given age, the risk of osteoporotic fracture is approximately two times greater in women than in men and in white people of Northern European ancestry than in Africans or Asians [10]. Women are at greater risk because of the lower peak bone density achieved in adulthood and greater susceptibility to rapid bone loss associated with menopause. Women also have a greater tendency than men to survive well into the age ofvulnerability [11—13]. Therefore, for these reasons much of the past research activity in the field has been focused on postmenopausal osteoporosis. The most serious consequences to the patient appear to result from hip fractures. Hip fractures account for the major proportion of the measured economic impact of osteoporosis because of the necessity of hospitalization [12,13]. Additionally, mortality within 4 months of hip fracture is currently 20%, with the majority of the survivors facing lifelong impairment. Risk assessment analyses have clearly shown that the risk of hip fractures increases exponentially with age and is currently 40% for white women aged 50 years or more in the United States [8]. As life expectancy continues to increase in most regions worldwide, the total of 323 million individuals aged 65 years or older in 1990 is expected to exceed 1.5 billion by the year 2050. Worldwide, the number of hip fractures may increase from 1.7 million in 1990 to 6.3 million 1 Burger's Medicinal Chemistry, Drug Discovery, and Development, Seventh Edition, edited by Donald J. Abraham and David P. Rotella Copyright © 2010 John Wiley & Sons, Inc. 2 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES by 2025 [14,15]. Assuming a 5% annual inflation rate, costs for hip fractures in the United States alone are projected to increase from an excess of $10 billion in 1990 to $240 billion by 2040 [16,17]. These may be conservative estimates because while most vertebral fractures do not lead to hospitalization, human costs were recently shown to be significant in terms of lost days due to back pain (2 days of bed rest, 10 days of limited activity). As a consequence, a number of therapeutic strategies have been successfully pursued in an effort to satisfy this unmet medical need. Supportive clinical data with molecules with varying modes of actions such as the bispho-sphonates, selective estrogen receptor modulators, and parathyroid hormone analogs suggest that very different pharmacological approaches can be utilized to prevent further bone loss in postmenopausal women. This review will focus on those therapies that act by inhibiting bone resorption. Subsequent chapters address therapies that result in bone formation. 1.1. Calcitonin and Integrin Antagonists Salmon calcitonin is among the most potent inhibitors of the bone-resorbing activity of osteoclasts in vitro [18-20] and is available as intramuscular injection and as nasal spray formulations to treat postmenopausal osteoporosis. While calcitonin has been shown to inhibit osteoclastic activity at low concentrations in vitro, calcitonin signaling is desensitized with continued exposure through the downregulation of calcitonin receptors [21-23]. This may help explain the somewhat limited clinical efficacy observed of 1—1.5% vertebral BMD increase over 3 years for treated patients. Nevertheless, despite this limited BMD efficacy observed for calcitonins and the poor bioavailability observed for nasal calcitonin [24], both formulations were shown to decrease significantly the incidence of vertebral fractures in osteoporotic women [25-28]. Calcitonin also has analgesic effects that appear to help alleviate bone pain in osteoporotic women, which may help explain calcitonin's popularity in some regions of Europe and Japan. An alternative therapeutic strategy to inhibit osteoclastic bone resorption has been to target the integrin mediated attachment of osteoclasts to the bone surface [29]. The Arg-Gly-Asp (RGD)-containing snake venom protein, echistatin, was shown to be a potent inhibitor of the av(33 integrin mediated resorb-ing activity of osteoclasts in vitro [30,31] and in vivo [32,33]. While echistatin itself is not likely to be therapeutically useful [34], RGD peptides and integrin antagonists have been shown to prevent bone loss in ovariectomized animals [35,36]. More recently, av(3a antagonist with improved drug-like properties have been described that 1 and 2 in Fig. 1. Both demonstrated potent antagonist activity in vitro. Compound 2 has good oral bioavailability in rats, dogs, and monkeys and has demonstrated bone-related efficacy in rats and monkeys after oral administration [37]. 1.2. Cathepsin K Inhibitors Cathepsin K is a lysosomal cysteine protease that is highly expressed in osteoclasts [38-40]. Cathepsin K has been mapped to chromosome lq21, and functional mutations to this gene OMe Figure 1. Integrin antagonists. INTRODUCTION 3 occur naturally, resulting in pycnodysostosis, a rare skeletal dysplasia that is characterized by dwarfism, low rate of bone turnover, and osteosclerosis [41]. Chemical tools represented by peptide aldehyde inhibitors of this enzyme have been shown to inhibit resorbing activity of osteoclasts in vitro with IC50 of 20-100 nM and in rats [42]. Emerging evidence that cathepsin K is the primary enzyme involved in osteoclastic bone resorption has made it an important target for the treatment of osteoporosis [43]. Several studies have shown that cathepsin K deficiency leads to an increase in BMD [44]. Pharmacological studies of cathepsin K inhibitors in rats [45] and monkeys [46] have shown reductions in biochemical markers of bone resorption and increased BMD. Recently, clinical data have been disclosed for the cathepsin K inhibitor balicatib demonstrating a reduction of biochemical markers of bone resorption and in- creases in BMD over 1 year of treatment [47] In addition, a 3-week study of MK-0822 showed a 70-80% reduction in serum CTx and an 80% reduction in urinary NT. Cathepsin inhibitors can be classified by structural class based on the electrophilic nature of subunit, or warhead, that interacts at the active site of the enzyme. Covalent inhibitors can be categorized into cyano or ketone-based molecules. There are also noncovalent inhibitors which are based on an aminoaniline structural subunit. Representative ketone inhibitors include those shown in Fig. 2 and include cyclohexanones 3 [48], azapanones 4 [49], dihydrofuranones 5 [50], and sulfona-midoketones 6 [51], to name a few. This class of inhibitors is generally characterized by electron withdrawing substituents such as alpha-heteroatom or carbonyl functionalities. Nitrile based inhibitors include dipeptide 7 [52] and aromatic nitriles [53]. Noncovalent competi- Figure 2. Cathepskin K inhibitors. 4 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES tive inhibitors include aminoethylaniline derivatives such as 8 [54] that achieve efficacy through lipophilic PI' interactions. 1.3. OPG/RANKL/RANK Inhibitors Osteoprotegerin (OPG) and receptor activator of nuclear factor-/1.67 defines a woman at high risk [184]. Tamoxifen was the first SERM to show reduced risk of breast cancer through a number of large, placebo-controlled, trials. In the Breast Cancer Prevention Trial, tamoxifen was evaluated in 14 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES a cohort of 13,388 women at increased risk of breast cancer and produced a 49% reduction in the relative risk of invasive breast cancer, and a 69% reduced risk of ER-positive mammary tumors [185]. However, despite this substantial reduction in risk, and inclusion of breast cancer risk reduction as an approved use for tamoxifen, the clinical use of tamoxifen for this indication has been rather lackluster—primarily due to a side effect profile that tilts the risk/benefit ratio in a negative direction in the mind of most physicians and women. The increase in endometrial cancer in postmenopausal women likely stems from the uterine stimulatory properties of tamoxifen and represents one area for improvement in other SERMs. To this regard, raloxifene hydrochloride has recently received approval for reducing the risk of invasive breast cancer in postmenopausal women with osteoporosis and in postmenopausal women at high risk for invasive breast cancer. After 8 years of following 4011 postmenopausal women with osteoporosis, a 66% reduction in the incidence of invasive breast cancer was observed with raloxifene use [186]. In the Study of Tamoxifen and Raloxifene (STAR) Trial, a head-to-head comparison of the two SERMs was conducted in 19,000 postmenopausal women at high risk of breast cancer, where tamoxifen and raloxifene hydrochloride were found to produce similar reductions in the incidence of invasive breast cancer [187], with the primary benefit being due to a reduced risk of ER-positive invasive breast cancers [188]. The most significant differences between raloxifene hydrochloride and tamoxifen in the STAR trial were significantly fewer uterine-associated adverse events with raloxifene hydrochloride (most notably the lack of endometrial cancer) while tamoxifen appeared to have a greater effect on noninvasive breast cancer incidence than raloxifene [187]. These differences between tamoxifen and raloxifene hydrochloride, although subtle indicate a difference from preclinical and even early clinical indicators, and as such, demonstrate the need for thorough clinical evaluation before accurate therapeutic risk/benefit assessment and approval of indications can be made for human use. To this regard, several SERMs in development, such as acolbifene and bazedox-ifene [189,190], have preclinical and early clinical profiles that are promising for potential use in reduction of risk for breast cancer, however, until sufficient clinical evaluation has been completed, it is too early to predict the ultimate utility of these molecules to this regard. 2. SUMMARY SERMs are a diverse class of molecules that affect a broad spectrum of biological systems with potential therapeutic benefit for a variety of diseases. Current concern over long-term use of estrogen-containing regimens has created an opportunity for application of SERMs to chronic indications such as osteoporosis treatment or prevention. The unique SERM profile also allows their use in other chronic indications of interest to postmenopausal women, most notably, breast cancer risk reduction and treatment. However, safety considerations are a very important consideration for SERM use in these chronic indications. The pleiotropic nature the ER and its role in numerous physiologic systems raises the importance of considering potential SERM benefits and/or adverse events in the cardiovascular system and other tissues. REFERENCES 1. Frost HM. Remodeling dynamics. In: Frost HM, editor. Bone Remodeling Biodynamics. Boston: Little Brown; 1963. p 65-78. 2. Parfitt AM. Quantum concept of bone remodeling and turnover: implications for the pathogenesis of osteoporosis. Calcif Tissue Int 1979;28:1-5. 3. Kanis JA, Melton LJ, Christiansen C, Jonston CC, KhalaevN. The diagnosis of osteoporosis. J Bone Miner Res 1994;9:1137-1141. 4. Kanis JA, Devogelaer JP, Gennari C. Practical guide for the use of bone mineral measurements in the assessment of treatment of osteoporosis: a position paper of the European Foundation for Osteoporosis and Bone Disease. Osteoporosis Int 1996;6:256-261. REFERENCES 15 5. Hui SL, Slemenda CW, Johnston CC. Age and bone mass as predictors of fracture in a prospective study. J Clin Invest 1988;81:1804-1809. 6. Slemenda CW, Hui SL, Longcope C, Wellman H, Johnston CC. Predictors of bone mass in perimenopausal women, a prospective study of clinical data using photon absorptiometry. Ann Intern Med 1990;112:96-101. 7. Marshall D, Johnell 0, Wedel H. Meta-analysis of how well measures of bone mineral density predict occurrence of osteoporotic fractures. Br Med J 1999;312:1254-1259. 8. Melton LJ, Chrischilles EA, Cooper C, Lane AW, Riggs BL. How many women have osteoporosis? J Bone Miner Res 1992;7:1005-1010. 9. Eddy D, Cummings SR, Dawson-Hughes B. Guidelines for the prevention, diagnosis and treatment of osteoporosis: cost-effectiveness analysis and review of the evidence. Osteoporosis Int 1998;8:1-88. 10. Johnell 0, Gullberg B, Allander E, Kanis JA. The apparent incidence of hip fractures in Europe: a study of national register sources. Osteoporosis Int 1992;2:298-302. 11. Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud KE, Cauley J, Black D, Vogt TM. Risk fractors for hip fracture in white women. N Engl J Med 1995;332:767-773. 12. Barrett-Connor E. The economic and human costs of osteoporotic fracture. Am J Med 1995;98(Suppl 2A): 3S-8S. 13. Nevitt MC, Ettinger B, Black DM, Stone K, Jamal SA, Ensrud K, Segal M, Genant HK, Cummings SR. The association of radiographi-cally detected vertebral fractures with back pain and function: a prospective study. Ann Intern Med 1998;128:793-800. 14. Cooper C, Campion G, Melton LJ. Hip fractures in the elderly: a worldwide projection. Osteoporosis Int 1992;2:285-289. 15. Kanis JA, McCloskey EV. Evaluation of the risk of hip fracture. Bone 1996;18(Suppl 3): 127s-132s. 16. Schneider EL, Guralnik JM. The aging of America. J Am Med Assoc 1990;263: 2335-2340. 17. Cummings SR, Rubin SM, Black D. The future of hip fractures in the United States: numbers, costs, and potential effects of postmenopausal estrogen. Clin Orthop 1990; 252:163-166. 18. Nicholson GC, Moseley JM, Sexton PM, Mendelsohn GAO, Martin TJ. Abundant calcitonin receptors in isolated rat osteoclasts. J Clin Invest 1986;64:355-360. 19. Arnett TR, Demptser DW. A comparative study of disaggregated chick and rat osteoclasts in vitro: effects of calcitonin and prostaglandins. Endocrinology 1987;120:602-608. 20. Murrills RJ, Shane E, Lindsay R, Dempster DW. Bone resorption by isolated human osteoclasts in vitro: effects of calcitonin. J Bone Miner Res 1989;4:259-268. 21. Raisz LG, Wener JA, Trummel CL, Feinblatt JF, Au WYW. Induction, inhibition and escape as phenomena in bone resorption. Excerpta Med Int Congr Ser 1972;243:446-449. 22. Tashjian AH, Wright DR, Ivey JL, Pont A. Calcitonin binding sites in bone: relationships to biological response and "escape". Recent Prog Horm Res 1978;34:285-299. 23. Nicholson GC, Moseley JM, Yates AJP, Martin TJ. Control of cAMP production in osteoclasts: calcitonin-induced persistent activation and homologous desensitization of adenylate cyclase. Endocrinology 1987;120:1902-1908. 24. Kohno T, Murasugi N, Sakurai H, Watabe K, Nakamuta Koida M, Sugie Y, Nomura M, Yanagawa A. A sandwich transfer enzyme immuno assay for salmon calcitonin: determination of the bioavailability of intranasal salmon calcitonin in human. J Clin Lab Anal 1997;11:380-387. 25. Overgaard K, Hansen MA, Jensen SB, Christiansen C. Effect of salcatonin given in-tranasally on bone mass and fracture rates in established osteoporosis. Br Med J 1992;305: 556-561. 26. Cardona JM, Pastor E. Calcitonin versus etidronate for the treatment of postmenopausal osteoporosis: a meta-analysis of published clinical trials. Osteoporosis Int 1997;7:165-174. 27. Thamsborg G, Jensen JE, Kollerup G, Hauge EM, Meisen F, Sorensen OH. Effect of nasal salmon calcitonin on bone remodeling and bone mass in postmenopausal osteoporosis. Bone 1996;18:207-212. 28. Stock JL, Avioli LV, Baylink DJ, Chesnut C, Genant HK, Maricic MJ, Silverman SL, Schaffer AV, Feinblatt J. Calcitonin-salmon nasal spray reduces the incidence of new vertebral fractures in postmenopausal women: 3 year interim results of the proof study. J Bone Miner Res 1997;12(Suppl 1): S149. 29. Horton MA, Rodan GA. Integrins as therapeutic targets in bone. In: Horton MA, editor. Adhesion Receptors as Therapeutic Targets. New York: CRC Press; 1996. p 223-245. 30. Sato MMK, Sardana WA, Grasser VM, Garsky JM, Murray AH, Gould RJ. Echistatin is a 16 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES potent inhibitor of bone resorption in culture. J Cell Biol 1990;111:1713-1723. 31. Sato MV, Garsky RJ, Majeska TA, Einhorn J, Murray Tashjian AH, Gould RJ. Structure-activity studies of the s-echistatin inhibition of bone resorption: implications for therapeutic utility. J Bone Miner Res 1994;9:1441-1449. 32. Fisher JE, Caulfield MP, Sato M, Quartuccio HA, Gould RJ, Garsky VM, Rodan GA, Rosenblatt M. Inhibition of osteoclastic bone resorption in vivo by echistatin, an arginyl-glycyl-asartyl (RGD)-containing protein. Endocrinology 1993;132:1411-1413. 33. Yamamoto M, Fisher JE, Gentile M, Seedor JG, Leu CT, Rodan SB, Rodan GA. The integ-rin ligand echistatin prevents bone loss in ovariectomized mice and rats. Endocrinology 1998;139:1411-1419. 34. Fisher JE, Caulfield MO, Sato M, Quartuccio HA, Gould RJ, Garsky VM, Rodan GA, Rosenblatt M. Response to letter. Endocrinology 1993;133:2408. 35. Engleman VW, Nickols AG, Ross FP, Horton MA, Griggs DW, Settle SL, Ruminski PG, Teitelbaum SL. A peptidomimetic antagonist of the avB3 integrin inhibits bone resorption in vitro and prevents osteoporosis in vivo. J Clin Invest 1997;99:22284-22292. 36. Lark MW, Stroup G, Cousins RD. Potent and selective inhibition of human cathepsin K leads to inhibition of bone resorption in vivo in a nonhuman primate. J Bone Miner Res 1998; 23:S219. 37. Hutchinson JJ, Halczenko W, Brashear KM, Breslin MJ, Coleman PJ, Duong LT, Gentile MA, Fisher JE, Hartman GD, Huff JR, Kimmel DB, Liu C-T, Meissner RS, Merkle K, Nagy R, Pennypacker B, Perkins JJ, Preuksaritanont T, Rodan G, Zartman AE, Rodan S, Duggan ME. Nonpeptide alphavbeta3 antagonists. 8. In vitro and in vivo evaluation of a potent alphavbeta3 antagonist for the prevention and treatment of osteoporosis. J Med Chem 2003;46:4790. 38. Tezuka K, Tezuka Y, Maejima A, Sato T, Nemoto K, Kamioka H, Hakeda Y, Kumegawa M. Molecular cloning of a possible cysteine proteinase predominantly expressed in osteoclasts. J Biol Chem 1994;269:1106-1109. 39. Bossard MJ, Tomaszek TA, Thompson SK, Amegadzie BY, Hannings CR, Jones C, Kurdla JT, McNulty DE, Drake FH, Gowen M, Levy MA. Proteolytic activity of human osteoclast cathepsin K. J Biol Chem 1996;271: 12517-12524. 40. Drake FH, Dodds R, Connor JI, Debouck J, Richardson S, Lee E, Rieman D, Barthlow R, Hastings G, Gowen M. Cathepsin K, but not cathepsins B, L, or S, is abundantly expressed in human osteoclasts. J Biol Chem 1996;271: 12511-12516. 41. Gelb BD, Shi GP, Chapman HA, Desnick RJ. Pycnodysostosis, a lysosomal disease caused by cathepsin K deficiency. Science 1996;273: 1236-1238. 42. Votta BJ, Levy MA, Badger A, Bradbeer J, Dodds RA, James IE, Thompson D, Bossard MJ, Carr T, Connor JR, Tomaszek TA, Szewc-zuk L, Drake FH, VeberDF, Gowen M. Peptide aldehyde inhibitors of cathepsin Kinhibit bone resorption both in vitro and in vivo. J Bone Miner Res 1997;12:1396-1406. 43. Grabowska UB, Chambers TJ, Shiroo M. Recent developments in cathepsin K inhibitor design. Curr Opin Drug Discov Dev 2005;8: 619-630. 44. Saftig P, Hunziker E, Wehmeyer 0, 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 Sei USA 1998;95:13453-13458. 45. Barrett DG, Boncek VM, Catalano JG, Deaton DN, Hassell AM, Jurgensen CH, Long ST, McFadyen RB, Miller AB, Miller LR, Payne JA, Ray JA, Samano V, Shewchuk LM, Ta-vares FX, Wells-Knecht KJ, Willard DH, Wright LL, Zhou HQ. P2-P3 conformationally constrained ketoamide-based inhibitors of cathepsin K. Bioorg Med Chem Lett 2005;15:3540-3546. 46. Kumar S, Dare L, Vasko-Moser JA, James IE, Blake SM, Rickard DJ, Hwang SM, Tomaszek T, Yamashita DS, Marquis RW, Oh H, Jeong JU, Veber DF, Gowen M, Lark MW, Stroup G. A highly potent inhibitor of cathepsin K (rela-catib) reduces biomarkers of bone resorption both in vivo and an acute model of elevated bone turnover in vivo in monkeys. Bone 2007;40:122-131. 47. Adami A, Supronik J, Hala T, Brown JP, Gar-nero P, Haemmerle S, Ortmann CE, Bouisset F, Trechsel U. Effect of one year treatment with the cathepsin-K inhibitor, balicatib, on bone mineral density (BMD) in postmenopausal women with osteopenia/osteoporosis. J Bone Min Res 2006;21:S24. 48. Kim MK, Kim HD, Park JH, Lim JI, Yang JS, Kwak WY, Sung SY, Kim HJ, Kim SH, Lee CH, Shim JY, Bae MH, Shin YA, Huh Y, Han TD, REFERENCES 17 Chong W, Choi H, Ahn BN, Yang SO, Son MH. An orally active cathepsin k inhibitor, furan-2-carboxylic acid, l-{l-[4-fluoro-2-(2-oxo-pyrroli-din-l-yl)-phenyl]-3-oxo-piperidin-4-ylcarba-moyl}-cyclohexyl)-amide (OST-4077), inhibits osteoclast Activity in vitro and bone loss in ovariectomized rats. J Pharmacol Exp Ther 2006;318:555-562. 49. Yamashita DS, Marquis RW, Xie R, Nidamarthy SD, Oh HJ, Jeong JU, Erhard KF, Ward KW, Roethke TJ, Smith BR, Cheng HY, Geng X, Lin F, Offen PH, Wang B, Nevins N, Head MS, Haltiwanger RC, Narducci Sargeant AA, Liable-Sands LM, Zhao B, Smith WW, Janson CA, Gao E, Tomaszek T, McQueney M, James IE, Gress CJ, Zembryki DL, Lark MW, Veber DF. Structure-activity relationships of 5-, 6-, and 7-methyl-substituted azepan-3-one cathepsin K inhibitors. J Med Chem 2006;49: 1587-1593. 50. Quibell M, Benn A, Flinn N, Monk T, Ramjee M, Ray P, Wang Y, Watts J. Synthesis and evaluation of cis-hexahydropyrrolo[3,2-b]pyr-rol-3-one peptidomimetic inhibitors of CAC1 cysteinyl proteinases. Bioorg Med Chem 2005;12:609-625. 51. Barrett DG, Catalano JG, Deaton DN, Long ST, McFadyenm RB, Miller AB, Miller LR, Ray JA, Samano V, Tavares FX, Wells-Knecht KJ, Wright LL, Zhou HQ. Acyclic, orally bioavail-able ketone-based cathepsin K inhibitors, bioorganic and medicinal chemistry letters. Bioorg Med Chem Lett 2007;12:22-30. 52. Missbach, M, Gamse, R, Trechsel, U, WO Patent 2005049028. 2005. 53. Altmann E, Cowan-Jacob SW, Missbach M. Novel purine nitrile derived inhibitors of the cysteine protease cathepsin K. J Med Chem 2004;47:5833-5863. 54. Shinozuka T, Shimada S, Matsui S, Yamane T, Ama M, Fukuda T, Taki M, Naito S. Potent and selective cathepsin K inhibitors. Bioorg Med Chem 2006;14:6789-6806.Rodan GA, Martin TJ. Role of osteoblasts in hormonal control of bone resorption: a hypothesis. Calcif Tissue Int 1981;33:349-351. 55. Lacey DL, Timms E, Tan HL, Kelley MJ, Dun-stan C, Burgess T, Elliot R, Colombero A, Elliot G, Scully S. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998;93:165-176. 56. Simonet WS, Lacey DL, Dunstan C, Kelly M, Chang MS, Luthy R, Nhuyen HQ, Wooden S, Bennett L, Boone T, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997;89:309-319. 57. Tsuda E, Goto M, Mochizuki SI, Yano Y, Kobayashi F, Morinaga T, Higashio K. Isolation of a novel cytokine from human fibroblasts that specifically inhibits osteoclastogenesis. Bio-chemBiophys Res Commun 1997;234:137-142. 58. Hsu H, Lacey DL, Dunstan C, Solovyev I, Colombero A, Timms E, Tan HL, Elliot G, Kelley MJ, Sarosi I, et al Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc Natl Acad Sei USA 1999;96:3540-3545. 59. Burgess T, Qian YX, Kaufman S, Ring BD, Van G, Capparelli C, Kelley M, Hsu H, Boyle WJ, Dunstan CR, et al. The ligand for osteoprotegerin (OPGL) directly activates mature osteoclasts. J Cell Biol 1999;145:527-538. 60. Mizuno A, Kanno T, Hoshi M, Shibata 0, Yano K, Fujise N, Kinosaki M, Yamaguchi K, Tsuda E, Murakam A, et al. Transgenic mice over-expressing soluble osteoclast differentiation factor (sODF) exhibit severe osteoporosis. J Bone Miner Metab 2002;20:337-344. 61. Bucay N, Sarosi I, Dunstan C, Morony S, Tarpley J, Capparelli C, Scully S, Tan HL, Xu W, Lacey DL, et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998; 12: 1260-1268. 62. Mizuno A, Amizuka N, Irie K, Murakami A, Fujise N, Kanno T, Sato Y, Nakagawa N, Ya-suda H, Mochizuki SI, et al. Severe osteoporosis in mice lacking osteoclastogenesis inhibitory factor/osteoprotegerin. Biochem Biophys Res Commun 1998;247:610-615. 63. Nakamura M, Udagawa N, Matsuura S, Mogi M, Nakamura H, Horiuchi H, Saito N, Hiraoka BY, Kobayahsi Y, Takaoka K, et al. Osteoprotegerin regulates bone formation through a coupling mechanism with bone resorption. Endocrinology 2003;144:5441-5449. 64. Vanderkerken De Leenheer KE, Shipman C, Asosingh K, Willems A, van Camp, B, Croucher, P. Recombinant osteoprotegerin decreases tumor burden and increases survival in a murine model of multiple myeloma. Cancer Res 2003;63:287-289. 65. Redlich K, Hayer S, Maier A, Dunstan CR, Tohidast-Akrad M, Lang SL, Turk B, Pietsch-mann P, Woloszczuk W, Haralambous S, et al. Tumor necrosis factor a-mediated joint destruction is inhibited by targeting osteoclasts with osteoprotegerin. Arthritis Rheum 2002; 46:785-792. 66. Kostenuik PJ, Bolon B, Morony S, Daris M, Geng Z, Carter C, Sheng J. Gene therapy with 18 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES human recombinant osteoprotegerin reverses established osteopenia in ovariectomized mice. Bone 2004;34:656-664. 67. Byrne FR, Morony S, Warmington K, Geng Z, Brown HL, Flores SA, Fiorino M, Yin SL, Hill D, Porkess V, et al. CD4 + CD45RBHl T cell transfer induced colitis in mice is accompanied by osteopenia which is treatable with recombinant human osteoprotegerin. Gut 2005;54:78-86. 68. Allen MR, Bloomfield SA. Hindlimb unloading has a greater effect on cortical compared to cancellous bone in mature female rats. J Appl Physiol 94:2003; 642-650. 69. Kodama Y, NakayamaK, Fuse H, Fukumoto S, Kawahara H, Takahashi H, Kurokawa H, Takahashi H, Kurokawa T, Sekiguchi C, et al. Inhibition of bone resorption by Pamidronate cannot restore normal gain in cortical bone mass and strength in tail-suspended rapidly growing rats. J Bone Miner Res 1997;12: 1058-1067. 70. Ichinose Y, Tanaka H, Inoue M, Mochizuki S, Tsuda S, Seino Y. Osteoclastogenesis inhibitor factor/osteoprotegerin reduced bone loss induced by mechanical unloading. Calcif Tissue Int 2004;75:338-343. 71. Mochizuki S, Fujise N, Higashio K, Tsuda E. Osteoclastogenesis inhibotory factor/osteoprotegerin ameliorates the decrease in both bone mineral density and bone strength in immobilized rats. J Bone Miner Metab 2002;20:14-20. 72. Ross AB, Bateman TA, Kostenuik PJ, Ferguson VL, Lacey DL, Dunstan CR, Simske SJ. The effects of osteoprotegerin on the mechanical properties of rat bone. J Mater Sei Mater Med 2001;12:583-588. 73. Bateman TA, Dunstan CR, Ferguson VL, Lacey DL, Ayers RA, Simske SJ. Osteoprotegerin mitigates tail suspension-induced osteopenia. Bone 2000;26:443-449. 74. Kostenuik PJ, Bateman TA, Morony S, Warmington K, Geng Z, Simske SJ, Ferguson VL, Dunstan CR, Lacey DL. OPG prevents relative osteopenia and deficits in skeletal strength in mice during a 12-day spaceflight. J Bone Miner Res 2002;17(Suppl 1): S209. 75. Rodan G, Fleisch HA. Bisphosphonates: mechanisms of action. J Clin Invest 1996;97: 2692-2696. 76. Fleisch H. Bisphosphonates in bone disease: from the laboratory to the patient. 3rd ed. Parthenon Publishing; 1997. 77. Reginster JYL, Halkin V, Gösset C, Deroisy R. The role of bisphosphonates in the treatment of osteoporosis. Drugs Today 1997;33:563-570. 78. Jeal W, Barradell LB, McTavish D. Alendronate: a review of its pharmacological properties and therapeutic efficacy in postmenopausal osteoporosis. Drugs 1997;53:415-434. 79. Yates J, Rodan GA. Alendronate and osteoporosis. Drug Discov Today 1998;3:69-78. 80. Watts NB, Harris ST, Genant HK, Wasnich RD, Miller PD, Jackson RD, Licata AA, Ross P, Woodson GC, Yanover MJ, Mysiw JW, Kohse L, Rao MB, Steiger P, Richmond B, Chesnut CH. Intermittent cyclical etidronate treatment of postmenopausal osteoporosis. N Engl J Med 1990;323:73-79. 81. Harris ST, Watts NB, JacksonRD, Genant HK, Wasnich RD, Ross P, Miller PD, Licata AA, Chesnut CH. Four-year study of intermittent cyclic etidronate treatment of postmenopausal osteoporosis: three years of blinded therapy followed by one year of open therapy. Am J Med 95:1993; 557-567. 82. Howsey J, Riggs BL, Kelly PJ, Hoffman DL, Bordier P. The treatment of osteoporosis with disodium ethane-l-hydroxy-l,l-diphosphonate. J Lab Clin Med 1971;78:574-581. 83. Heaney RP, Saville PD. Etidronate disodium in postmenopausal women. Clin Pharmacol Ther 1976;20:593-604. 84. Boyce BF, Smith L, Fogelman I, Johnston E, Ralston S, Boyle IT. Focal osteomalacia due to low-dose diphosphonate therapy in Paget's disease. Lancet 1984; 821-824. 85. Gibbs, CJ, Aaron, JE, Peacock, M. Osteomalacia in Paget's disease treated with short-term, high dose sodium etidronate. Br Med J 1986;292:1227-1229. 86. Devogelaer JP, Broil H, Correa-Rotter R, Cum-ming DC, Deuxchaisnes CN, Geusens P, Hosk-ing D, Jaeger P, Kaufman JM, Leite M, Leon J, Liberman U, Menkes CJ, Meunier PJ, Reid I, Rodriguez J, Romanowicz A, Seeman E, Ver-meulen A, Hirsch LJ, Lombardi A, Plezia K, Santora AC, Yates AJ, Yuan W. Oral alendronate induces progressive increases in bone mass of the spine, hip, and total body over 3 years in postmenopausal women with osteoporosis. Bone 1996;18:141-150. 87. Black DM, Cummings SR, Karpf DB, Cauley JA, Thompson DE, Nevitt MC, Bauer DC, Genant HK, Haskell WL, Marcus R, Ott SM, Torner JC, Quandt SA, Reiss TF, Ensrud KE. Randomized trial of effect of alendronate on risk of fracture in women with existing vertebral fractures. Fracture Intervention Trial Research Group. Lancet 1996;348: 1535-1541. REFERENCES 19 88. Liberman UA, Weiss SR, Broil J, Minne HW, Dequeker J, Favus M, Seeman E, Recker R, Capizzi T, Santora AC, Lombardi A, Shah R, Hirsch LJ, Karpf DB. Effect of oral alendronate on bone mineral density and the incidence of fractures in postmenopausal osteoporosis. N Engl J Med 1995;333:1437-1443. 89. Hosking D, Chilvers C, Christiansen C, Ravn P, Wasnich R, Ross P, McClung M, Balske A, Thompson D, Daley M, Yates AJ. Prevention of bone loss with alendronate in postmenopausal women under 60 years of age. N Engl J Med 1998;338:485-492. 90. Sato M, Grasser W, Endo N, Akins R, Simmons H, Thompson DD, Golub E, Rodan GA. Bisphosphonate action: alendronate localization in rat bone and effects on osteoclast ultrastructure. J Clin Invest 1991;69:2095-2105. 91. Breuil, V, Cosman F, Stein, L, Horbert, W, Nieves, J, Shen, V, Lindsay, R, Dempster, DW. Human osteoclast formation and activity in vitro: effects of alendronate. J Bone Miner Res 1998;13:1721-1729. 92. Lin JH, Chen IW, deLuna FA. Nonlinear kinetics of alendronate, plasma protein binding and bone uptake. Drug Metab Dispos 1994;22:400-405. 93. Lin JH. Bisphosphonates: a review of their pharmacokinetic properties. Bone 1996;18: 75-85. 94. Lin JH, Chen IW, deLuna FA. On the absorption of alendronate in rats. J Pharm Sei 1994;83:1741-1746. 95. Azuma Y, Sato H, Oue Y, Okabe K, Ohta T, Tsuchimoto M, Kiyoki M. Alendronate distributed on bone surfaces inhibits osteoclastic bone resorption in vitro and in experimental hypercalcemia models. Bone 1995;16:235-245. 96. Gertz BJ, Holland SD, Kline WF, Matuszewski BK, Porras AG. Clinical pharmacology of alendronate sodium. Osteoporosis Int 1993; (Suppl 3): S13-S16. 97. Sato M, Grasser W. Effects of bisphosphonates on isolated rat osteoclasts as examined by reflected light microscopy. J Bone Miner Res 1990;5:31-40. 98. 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;67:1930-1935. 99. Schmidt A, Rutledge SU, Endo N, Opas EE, Tanaka H, Wesolowski G, Leu CT, Huang Z, Ramachandaran C, Rodan SB, Rodan GA. Protein—tyrosine phosphatase activity regulates osteoclast formation and function: inhibition by alendronate. Proc Natl Acad Sci USA 1996;93:3068-3073. 100. Hughes DE, Wright KR, Uly HL, Sasaki A, Yoneda T, Roodman GD, Mundy GR, Boyce BF. Bisphosphonates promote apoptosis in murine osteoclasts in vitro and in vivo. J Bone Miner Res 1995;10:1478-1487. 101. Sahni M, Guenther HL, Fleisch H, Collin P, Martin TJ. Bisphosphonates act on rat bone resorption through the mediation of osteoblasts. J Clin Invest 1993;71:2004-2011. 102. Owens JM, Fuller K, Chambers TJ. Osteoclast activation: potent inhibition by the bi-sphosphonate alendronate through a nonre-sorptive mechanism. J Cell Physiol 1997; 172:79-86. 103. Czabo C, Martin M, Oldfield E. An investigation of bone resorption and Dictyostelium dis-coideum growth inhibition by bisphosphonate drugs. J Med Chem 2002;45:2894.Dunford J, Thompson K, Coxon F. Structure—activity relationships for inhibition of farnesyl diphosphate synthase in vitro and inhibition of bone resorption in vivo by nitrogen-containing bisphosphonates. J Pharm Exp Ther 2001; 296:235. 104. Chavassieux PM, Ariot 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;100:1475-1480. 105. Sato M, Bryant H, Iversen P, Helterbrand J, Smietana F, Bemis K, Higgs R, Owan I, Takano T, Burr D. Advantages of raloxifene over alendronate or estrogen on nonreproduc-tive and reproductive tissues in the long-term dosing of ovariectomized rats. J Pharmacol Exp Ther 1996;279:298-305. 106. Balena R, Toolan BC, Shea M, Markatos A, Myers ER, Lee SC, Opas EE, Seedor JG, Klein H, Frankenfield D, Quartuccio H, Fioravanti C, Clair J, Brown E, Hayes WC, Rodan GA. The effects of 2-year treatment with the aminobisphosphonate alendronate on bone metabolism, bone histomorphome-try, and bone strength in ovariectomized nonhuman primates. J Clin Invest 1993;92: 2577-2586. 107. Balena R, Markatos A, Seedor JG, Gentile M, Stark C, Peter CP, Rodan GA. Long-term safety of the aminobisphosphonate alendronate in adult dogs. II. Histomorphometric ana- 20 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES lysis of the L5 vertebra. J Pharmacol Exp Ther 1996;276:277-283. 108. Burr DB, Forwood MR, Fyhrie DP, Martin RB, Schaffler MP, Turner CH. Bone microdamage and skeletal fragility in osteoporotic and stress fractures. J Bone Miner Res 1997;12:6-15. 109. Gertz BJ, Holland SD, Kline WF, Bogdan K, Matuszewski BK, Freeman A, Porras AG. Studies of the oral bioavailability of alendronate. Clin Pharmacol Ther 1995;58:288-298. 110. Pizzani E, Valenzuela G. Esophagitis associated with alendronate sodium. Va Med Q 1997;124(3): 181-182. 111. Levine L, Nelson D. Esophageal stricture associated with alendronate therapy. Am J Med 1997;102(5): 489-491. 112. Ravn P, Clemmensen B, Riis B, Christiansen C. The effect on bone mass and bone markers of different doses of Ibandronate: a new bispho-sphonate for prevention and treatment of postmenopausal osteoporosis. Bone 1996;19:527; Morri H, Nishizawa Y, Taketani Y. J Bone Miner Res 2002;17:M324. Intravenous zolen-dronate dosed every 3, 6, or 12 months has beneficial effects on BMD in the spine and hip. 113. Reid IR, Brown JP, Burkhardt P, Horowitz Z, Richardson P, Treschel U, Widmer A, Devoge-laer JP, Kaufman JM, Jaeger P, Body JJ, Meunier PJ. Intravenous zoledronic acid in postmenopausal women with low bone mineral density. N Eng J Med 2002;346: 653. 114. Black LJ, Rowley ER, Bekele A, Sato M, Magee DE, Williams DC, Cullinan GJ, Bendele R, Kauffman RF, Bensch W, Frolik CA, Termine JD, Bryant HU. Raloxifene (LY139482 HC1) prevents bone loss and reduces serum cholesterol without causing uterine hypertrophy in ovariectomized rats. J Clin Invest 1994;93: 63-69. 115. Draper MW, Flowers DE, Huster WJ, Nield JA, Harper KD, Arnaud C. A controlled trial of raloxifene (LY139481) HC1: impact on bone turnover and serum lipid profile in healthy postmenopausal women. J Bone Miner Res 1997;11:835-842. 116. Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Petters-son K, Warner M, Gustafsson JA. Mechanisms of estrogen action. Physiol Rev 2001;81: 1535-1565. 117. Konyalioglu S, Durmaz G, Yalcin A. The potential antioxidant effect of raloxifene treatment: a study on heart, liver andbrain cortex of ovariectomized female rats. Cell Biochem Funct 2008;25:259-266. 118. Simoncini T, DeCaterina R, Genazzani AR. Selective estrogen receptor modulators: different actions on vascular cell adhesion molecule (VCAM-1) expression in human endothelial cells. J Clin Endocrinol Metab 1999;84: 815-818. 119. Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, BonnT, EngstromO, OhmanL, Green GL, Gustafsson JA. Molecularbasis of agonism and antagonism in the estrogen receptor. Nature 1997;389:753-768. 120. McDonnel DP, Clemm DL, Hermann T, Goldman ME, Pike JW. Analysis of estrogen receptor function in vitro reveals three distinct classes of anti-estrogens. Mol Endocrinol 1995;9:659-669. 121. Jordan VC, Collins MM, Rowsby L, Prestwich G. A monohydroxylated metabolite of tamoxifen with potent anti-estrogenic activity. J Endocrinol 1997;75:305-316. 122. Shiau K, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard A, Greene GL. The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 1998;95:927-937. 123. Kuiper GGJM, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA 1996;93: 5925-5930. 124. Monroe DG, Secreto FJ, Subramaniam M, Getz BJ, Khosla S, Spelsberg TC. Estrogen receptor alpha and beta heterodimers exert unique effects on estrogen- and tamoxifen-de-pendent gene expression in human U20S osteosarcoma cells. Mol Endocrinol 2005; 19: 1555-1568. 125. Saunders PT, Maguire SM, Gaughan J, Millar MR. Expression of oestrogen receptor beta (ER beta) in multiple rat tissues visualised by im-munohistochemistry. J Endocrinol 1997; 154: R13-R16. 126. Chaidarun SS, Alexander JM. A tumor-specific truncated estrogen receptor splice variant enhances estrogen-stimulated gene expression. Molec Endocrinol 1998;12:1355-1366. 127. Rey JM, Pujol P, Dechaud H, Edouard E, He-don B, Maudelonde T. Expression of oestrogen receptor-alpha splicing variants and oestrogen receptor-beta in endometrium of infertile patients. Mol Hum Reprod 1998;4:641-647. 128. MclnerneyEM, RoseDW, FlynnSE, WestinSE, Mullen TM, Krones A, Inostroza J, Torchia J, Nolte RT, Assa-Munt N, Milburn MV, Glass CK, Rosenfeld MG. Determinants of coactivator REFERENCES 21 LXXLL motif specificity in nuclear receptor transcriptional activation. Genes Dev 1998;12: 3357-3368. 129. Onate SA, Tsai SY, Tsai M-J, O'Malley BW. Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 1995;270:1354-1357. 130. Chen H, Lin RJ, Lin RJ, Schütz RL, Chankra-varti D, Nash A, Privalsky ML, Nakatani Y, Evans RM. Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300. Cell 1997;90(3): 569-580. 131. Voegel JJ, Heine MJ, Zechel C, Chambon P, Gronemeyer H. TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. EMBO J 1996;15:3667-3675. 132. Lanz RB, McKenna NJ, Onate SA, Albrecht U, Wong J, Tsai SY, Tsai M-J, O'Malley BW. A steroid receptor coactivator, SRA, functions as an RNA and is present in an SRC-1 complex. Cell 1999;97:17-27. 133. Westin S, Kurokawa R, Nolte RT, Wisely GB, Mclnerney EM, Rose DW, Milburn MV, Rosenfeld MG, Glass CK. Interactions controlling the assembly of nuclear-receptor heterodimers and coactivators. Nature 1998;395:199-202. 134. Korzus E, Torchia J, Rose DW, Xu L, Kurokawa R, Mclnerney EM, Mullen TM, Glass CK, Rosenfeld MG. Transcription factor-specific requirements for coactivators and their acetyltransferase functions. Science 1998;279: 703-707. 135. Mclnerney EM, Tsai MJ, O'Malley BW, Katzenellenbogen BS. Analysis of estrogen receptor transcriptional enhancement by a nuclear hormone receptor coactivator. Proc Natl Acad Sei USA 1996;93:10069-10073. 136. Smith CL, Nawaz Z, O'Malley BW. Coactivator and corepressor regulation of the agonist/ antagonist activity of the mixed antiestrogen, 4-hydroxytamoxifen. Mol Endocrinol 1997;11: 657-666. 137. Montano MM, Ekena K, Delage-Mourroux R, Chang W, Martini P, Katzenellenbogen BS. An estrogen receptor-selective coregulator that potentiates the effectiveness of antiestrogens and represses the activity of estrogens. Proc Natl Acad Sei USA 1999;96:6947-6952. 138. Xu J, Liao L, Ning G, Yoshida-Komiya H, Deng C, O'Malley BW. The steroid receptor coactivator SRC-3 (p/CIP/RAC3/AIBl/ACTR/ TRAM-1) is required for normal growth, puberty, female reproductive function, and mam- mary gland development. Proc Natl Acad Sci USA 2000;97:6379-6384. 139. Smith CL, DeVera DG, Lamb DJ, Zafar N, Yong-Hiu J, Beaudet AL, O'Malley BW. Genetic ablation of the steroid receptor coactivator ubiquitin ligase, E6-AP, results in tissue selective steroid hormone resistance and defects in reproduction. Mol Cell Biol 2002;22:525-535. 140. Bautista S, Valles H, Walker RL, Anzick S, Zellinger R, Meltzer P, Theillet C. In breast cancer, amplification of the steroid receptor coactivator gene AIB1 is correlated with estrogen and progesterone receptor positivity. Clin Cancer Res 1998;4:2925-2929. 141. Shang Y, Brown M. Molecular determinants for the tissue specificity of SERMs. Science 2002;295:2465-2468. 142. Grese TA, Sluka JP, Bryant HU, Cullinan GC, Glasebrook AL, Jones CD, Matsumoto K, Palk-owitz AD, Sato M, Termine JD, Winter MA, Yang NN, Dodge JA. Molecular determinants of tissue selectivity in estrogen receptor modulators. Proc Natl Acad Sci USA 1997;94: 14105-14110. 143. Sato M, Turner CH, Wang T, Adrian MD, Rowley E, Bryant HU. LY353381.HC1: a novel raloxifene analog with improved SERM potency and efficacy in vivo. J Pharmacol Exp Ther 1998;287:1-7. 144. Sato M, Rippy MK, Bryant HU. Raloxifene, tamoxifen, nafoxidine and estrogen effects on reproductive and nonreproductive tissues in ovariectomized rats. FASEB J 1996; 10: 905-912. 145. Ke HZ, Chen HK, Qi H, Pirie CM, Simmons HA, Ma YF, Jee WSS, Thompson DD. Effects of droloxifene on prevention of cancellous bone loss and bone turnover in the axial skeleton of aged, ovariectomized rats. Bone 1995; 17: 491-496. 146. Nuttall ME, Bradbeer JN, Stroup GB, Nadeau DP, Hoffman SJ, Zhao H, Rehm S, Gowen M. Idoxifene: a novel selective estrogen receptor modulator prevents bone loss and lowers cholesterol levels in ovariectomized rats and decreases uterine weight in intact rats. Endocrinology 1998;139:5224-5234. 147. Jimenez MA, Magee DE, Bryan HU, Turner RT. Clomiphene prevents cancellous bone loss from tibia of ovariectomized rats. Endocrinology 1998;138:1794-1800. 148. Komm BS, Kharode YP, Bodine PV, Harris HA, Miller CP, Lyttle CR. Bazedoxifene acetate: a selective estrogen receptor modulator 22 OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES with improved selectivity. Endocrinology 2005;146:3999-4008. 149. Ke HZ, Paralkar VM, Grasser WA, Crawford DT, Qi H, Simmons HA, Pirie CM, Chidsey-Frink KL, Owen TA, Smock SL, Chen HK, Jee WS, Cameron KO, Rosati RL, Brown TA, Dasilva-Jardine P, Tompson DD. Effects of CP336,156, a new, non-steroidal estrogen agonist/antagonist on bone, serum cholesterol, uterus and body composition in rat models. Endocrinology 1998;139:2068-2076. 150. Galbiati E, Caruso PL, Amari G, Armani E, Ghirardi S, Delcanale M, Civelli M. Effects of 3-phenyl-4-[[4-[2-(l-piperidinyl)ethoxy]phenyl]-methyl]-2H-l-benzopyran-7-ol (CHF 4056), a novel nonsteroidal estrogen agonist/antagonist, on reproductive and nonreproductive tissue. J Pharmacol Exp Ther 300:2002; 802-809. 151. Qu Q, Zheng H, Dahlund J, Laine A, Cock-croft N, Peng Z, Koskinen M, Hemminki K, Kangas L, Vaananen K, Harkonen P. Selective estrogenic effects of a novel tripheny-lethylene comound, FC-1271a on bone, cholesterol level, and reproductive tissue in intact and ovariectomized rats. Endocrinology 2000;141:809-820. 152. Martel C, Picard S, Belanger RV, Labrie C, Labrie F. Prevention of bone loss by EM-800 and raloxifene in the ovariectomized rat. J Steroid Biochem Molec Biol 2000;74:45-56. 153. Turner CH, Sato M, Bryant HU. Raloxifene preserves bone strength and bone mass in ovariectomized rats. Endocrinology 1994; 135: 2001-2005. 154. Cano A, Dapia S, Noguera I, Pineda B, Her-menegildo C, del Val R, Caeiro JR, Garcia-Perez MA. Comparative effects of 17b-estra-diol, raloxifene and genistein on bone 3D microarchitecture and volumetric bone mineral density in the ovariectomized mice. Osteo-poros Int 2008; (Epub ahead of print). 155. Komm BS, Bodine PV, Minck DR. Effects of bazedoxifene on bone loss: a 12 month study in ovariectomized rats. J Bone Miner Res2007;22 (Suppl 1): S206. 156. Lees CJ, Register TC, Turner CH, Wang T, Stancill M, Jerome CP. Effects of raloxifene on bone density, biomarkers, and histomorpho-metric and biomechanical measures in ovariectomized cynomolgus monkeys. Menopause 2002;9:320-328. 157. Lees CJ, Shen V, Brommage R. Effects of lasofoxifene on bone in surgically postmenopausal cynomolgus monkeys. Menopause 2007;14:97-105. 158. Hotchkiss CE, Stavisky R, Nowak J, Brommage R, Lees CJ, Kaplan J. Levormeloxifene prevents increased bone turnover and vertebral bone loss following ovariectomy in cynomolgus monkeys. Bone 2001;29:7-15. 159. Frolik CA, Bryant H, Black EC, Magee DE, Chandrasekhar S. Time dependent changes in biochemical bone markers and serum cholesterol in ovariectomized rats: effects of raloxifene HCI, tamoxifen, estrogen and alendronate. Bone 1996;18:621-627. 160. Evans G, Bryant HU, Magee D, Satom M, Turner RT. The effects of raloxifene on tibia histomorphometry in ovariectomized rats. Endocrinology 1994;134:2283-2288. 161. Helvering LM, Liu R, Kulkarni NH, Wei T, Chen P, Huang S, Lawrence F, Halladay DL, Miles RR, Ambrose EM, Sato M, Ma YL, Frolik CA, Dow ER, Bryant HU, Onyia JE. Expression profiling of rat femur revealed suppression of bone formation genes by treatment with alendronate and estrogen but not raloxifene. Molec Pharmacol 2005;68:1225-1238. 162. Ettinger B, Black DM, Mitlak BM, Knickerbocker RK, Nickelsen T, Genant HK, Christiansen C, Delmas PD, Zanchetta JR, Stakkes-tad J, Gluer CC, Krueger K, Cohen FJ, Eckert S, Ensrud KE, Avioli LV, Lips P, Cummings SR. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene. J Am Med Assoc 1999;282:637-645. 163. Heany RP, Draper MW. Raloxifene and estrogen: comparative bone-remodelling kinetics. Clin Endocrinol Metab 1997;82:3425-3429. 164. Ott SM, Oleksik A, Lu Y, Harper KD, Lips P. Bone histomorphometric and biochemical marker results of a two year placebo controlled trial of raloxifene in postmenopausal women. J Bone Miner Res 2002;17:341-348. 165. Weinstein RS, Parfitt AM, Marcus R, Green-wald M, Crans G, Muchmore DB. Effects of raloxifene, hormone replacement therapy, and placebo on bone turnover in postmenopausal women. Osteoporos Intl 2003;14:814-822. 166. Johnell O, Scheele WM, Lu Y, Reginste rJ-Y, Need AG, Seeman E. Additive effects of raloxifene and alendronate on bone density and biochemical markers of bone remodeling in postmenopausal women with osteoporosis. J Clin Endocrinol Metab 2002;87: 985-992. 167. Allen MR, Iwata K, Sato M, Burr DB. Raloxifene enhances vertebral mechanical proper- REFERENCES 23 ties independent of bone density. Bone 2006;39:1130-1135. 168. Li J, Sato M, Jerome C, Turner CH, Fan Z, Burr DB. Microdamage accumulation in the monkey vertebrae does not occur when bone turnover is suppressed by 50% or less with estrogen or raloxifene. J Bone Miner Res 2005;23:48-54. 169. Uusi-Rasi K, Beck TJ, Semanick LM, Daph-tary MM, Crans GG, Desaiah D, Harper D. Structural effects of raloxifene on the proximal femur: results from the multiple outcomes of raloxifene evaluation trial. Osteoporosis Intl 2006;17:575-586. 170. Delmas PD, Genant HK, Crans GG, Stock JL, Wong M, Siris E, Adach JC. Severity of prevalent vertebral fractures and the risk of subsequent vertebral and nonvertebral fractures: results from the MORE trial. Bone 2003;33: 522-532. 171. Alexandersen P, Riss BJ, Stakkestad JA, Delmas PD, Christiansen C. Efficacy of levorme-loxifene in the prevention of postmenopausal bone loss and on the lipid profile compared to low dose hormone replacement therapy. J Clin Endocrinol Metab 2001;86:755-760. 172. Christgau S, Tanko LB, Cloos PA, Mouritzen U, Christiansen C, Delaisse JM, Hoegh-Anderson P. Suppression of elevated cartilage turnover in postmenopausal women and in ovariectomized rats by estrogen and a selective estrogen-receptor modulator (SERM). Menopause 2004;11:508-518. 173. Chestnut C, Weiss S, Mulder H, Wasnich R, Greenwald R, Eastell R, Fitts D, Jensen C, Haines A, MacDonald B. Idoxifene increases bone mineral density in osteopenic postmenopausal women. Bone 1998;23(Suppl): S389. 174. Gardner M, Taylor A, Wei G, Calcagni A, Duncan B, Milton A. Clinical pharmacology of multiple doses of lasofoxifene in postmenopausal women. J Clin Pharmacol 2006;46: 52-58. 175. Patat A, McKeand W, Baird-Bellaire S, Ermer J, LeCo zF. Absolute/relative bioavailability of bazedoxifene acetate in healthy postmenopausal women. J Clin Pharmacol Ther 2003;73:43. 176. Silverman SL, Christiansen K, Genant HK, Zanchetta JR, Valter L, de Villiers TJ, Con-stantine G, Chines AA. Efficacy of bazedoxifene in reducing new vertebral fracture risk in postmenopausal women with osteoporosis from a 3-year randomized, placebo- and active-controlled trial. J Bone Miner Res 2007;22(Suppl 1): S58. 177. Miller PD, Christiansen C, HoeckHC, Kendler DL, Lewiecki EM, Woodson G, Ciesielska M, Chines AA, Constantine G, Delmas PD. Efficacy of bazedoxifene for prevention of postmenopausal osteoporosis: results of a 2-year, phase III, placebo- and active-controlled study. J Bone Miner Res 2007;22(Suppl 1): S59. 178. Tiitinen A, Nikander E, Hietanen P, Metsa-Heikkila M, Ylikorkala O. Changes in bone mineral density during and after 3 years use of tamoxifen or toremifene. Maturitas 2004;48:321-327. 179. Marttunen MB, Hietanen P, Tiitinen A, Ylikorkala O. Comparison of effects of tamoxifen and toremifene on bone biochemistry and bone mineral density in postmenopausal breast cancer patients. J Clin Endocrinol Metab 1998;83:1158-1162. 180. Love RR, Mazess RB, Barden HS, Epstein S, Newcomb PA, Jordan VC, Carbone PP, DeMets DL. Effects of tamoxifen on bone mineral density in postmenopausal women with breast cancer. N Eng J Med 1992;326: 852-856. 181. Powles TJ, Hickish T, Kanis JA, Tidy A, Ashley S. Effect of tamoxifen on bone mineral density measured by dual-energy X-ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 1996; 114:78-84. 182. Nierengarten MB. Toremifene might improve side effects of ADT. Lancet Oncol 2007;8:287. 183. Smith MR. Treatment related osteoporosis in men with prostate cancer. Clin Cancer Res 2006;12:6315S-6319S. 184. Costantino JP, Gail MH, Pee D, Anderson S, Redmond CK, Benichou J, Wieand HS. Validation studies for models projecting the risk of invasive and total breast cancer incidence. J Natl Cancer Inst 1999;91:1541-1548. 185. Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM, Vogel V, Robidoux A, Dimitrov N, Atkins J, Daly M, Wieand S, Tan-Chiu E, Ford L, Wolmark N. Tamoxifen for prevention of breast cancer: Report of the National Surgical Adjuvant Breast and Bowel Project P-l Study. J Natl Cancer Inst 1998;90:1371-1388. 186. Martino S, Cauley JA, Barrett-Connor E, Powles TJ, Mershon J, Disch D, Secrest RJ, Cummings SR. Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 2004;96:1751-1761. OSTEOPOROSIS THERAPY: MECHANISTIC ANTIRESORPTIVES 24 187. Vogel VG, Costantino JP, Wickerham DL, Cro-nin WM, Cecchini RS, Atkins JN, Bevers TB, Fehrenbacher L, Pajon ER, Wade JL, Robi-doux A, Margolese RG, James J, Lippman SM, Runowicz CD, Ganz PA, Reis SE, McCaskill-Stevens W, Ford LG, Jordan VC, Wolmark N. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: The NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 Trial. J Amer Med Assoc 2006;295:2727-2741. 188. Barrett-Connor E, Mosca L, Collins P, Geiger MJ, Grady D, Kornitzer M, McNabb M, Wenger N. Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women. N Engl J Med 2006;335:125-137. 189. Labrie F, Champagne P, Labrie C, Roy J, Laverdiere J, Provencher L, Potvin M, Drolet Y, Panasci L, Esperance B, Dufresne J, Latreille J, Robert J, Samson B, Jolivet J, Yelle L, Cusan L, Diamond P, Candas B. Activity and safety of the antiestrogen EM-800, the orally active precursor of acolbifene, in tamoxifen-resistant breast cancer. J Clin Oncol 2004;22:864-871. 190. Adachi JD, Chesnut CH, Brown JP, Christiansen C, Russo LA, Fernandes CE, Menegoci JC, King A, Chines AA, Bessac L, Chakrabarti D. Safety and tolerability of bazedoxifene in postmenopausal women with osteoporosis: results from a 3-year, randomized, placebo- and active-controlled clinical trial. J Bone Miner Res 2007;22(Suppl 1): S460.