A single dose of zoledronate preserves bone mineral density for up to 2 years after a second course of romosozumab

M R McClung, M A Bolognese, J P Brown, J-Y Reginster, B L Langdahl, J Maddox, Y Shi, M Rojeski, P D Meisner, A Grauer, M R McClung, M A Bolognese, J P Brown, J-Y Reginster, B L Langdahl, J Maddox, Y Shi, M Rojeski, P D Meisner, A Grauer

Abstract

This phase 2 study evaluated the efficacy and safety of transitioning to zoledronate following romosozumab treatment in postmenopausal women with low bone mass. A single dose of 5 mg zoledronate generally maintained the robust BMD gains accrued with romosozumab treatment and was well tolerated.

Introduction: Follow-on therapy with an antiresorptive agent is necessary to maintain the skeletal benefits of romosozumab therapy. We evaluated the use of zoledronate following romosozumab treatment.

Methods: This phase 2, dose-finding study enrolled postmenopausal women with low bone mineral density (BMD). Subjects who received various romosozumab doses or placebo from months 0-24 were rerandomized to denosumab (60 mg SC Q6M) or placebo for 12 months, followed by open-label romosozumab (210 mg QM) for 12 months. At month 48, subjects who had received active treatment for 48 months were assigned to no further active treatment and all other subjects were assigned to zoledronate 5 mg IV. Efficacy (BMD, P1NP, and β-CTX) and safety were evaluated for 24 months, up to month 72.

Results: A total of 141 subjects entered the month 48-72 period, with 51 in the no further active treatment group and 90 in the zoledronate group. In subjects receiving no further active treatment, lumbar spine (LS) BMD decreased by 10.8% from months 48-72 but remained 4.2% above the original baseline. In subjects receiving zoledronate, LS BMD was maintained (percentage changes: - 0.8% from months 48-72; 12.8% from months 0-72). Similar patterns were observed for proximal femur BMD in both groups. With no further active treatment, P1NP and β-CTX decreased but remained above baseline at month 72. Following zoledronate, P1NP and β-CTX levels initially decreased but approached baseline by month 72. No new safety signals were observed.

Conclusion: A zoledronate follow-on regimen can maintain robust BMD gains achieved with romosozumab treatment.

Keywords: Antiresorptive; Bone mineral density; Follow-on regimen; Osteoporosis; Romosozumab.

Conflict of interest statement

MRM has received consulting fees and honoraria from Amgen and consulting fees from Myovant. MAB has received contract fees from and has been a speaker for Amgen. JPB has received research funding from Amgen, Eli Lilly, Mereo Biopharma, Radius Health, and Servier; has received consulting fees from Amgen, Eli Lilly, Orimed, and Servier; and has received lecture fees from Amgen and Eli Lilly. J-YR has received research funding from IBSA-Genevrier, Mylan, CNIEL, and Radius Health; has received lecture fees from IBSA-Genevrier, Mylan, CNIEL, and Dairy Research Council; and has received consulting fees from or participated in paid advisory boards for IBSA-Genevrier, Mylan, Radius Health, and Pierre Fabre. BLL has received research funding from Amgen and Novo Nordisk; has received consulting fees from Amgen, Eli Lilly, and UCB; and has received lecture fees from Amgen, Eli Lilly, and UCB. JM, YS, and MR are employees of and hold stock in Amgen. PDM is an employee of and holds stock in UCB Pharma. AG was an employee of Amgen at the time of the study and holds stock in Amgen.

Figures

Fig. 1
Fig. 1
Study schema. a Subjects were randomized 1:1:1:1:1:1:1:1 to the first 24 months of treatment. Administration of placebo and the various romosozumab doses was blinded; alendronate and teriparatide were administered open-label. At month 24, subjects were rerandomized (1:1) within treatment group to placebo or denosumab (60 mg SC Q6M) for 12 months, followed by a 12-month second course of romosozumab 210 mg QM. b For subjects who reached month 48 of the study, eligibility for the month 48 to 72 zoledronate follow-on period was assessed by the investigator using a 3-step approach with no randomization. Subjects were assigned to no further active treatment if they (1) had been assigned to active treatment throughout the first 48 months (romosozumab any dose and schedule, followed by denosumab 60 mg Q6M, and then followed by romosozumab 210 mg QM); (2) had no clinical vertebral or fragility fracture between months 24 and 48; (3) had a BMD T-score > − 2.5 at the lumbar spine, total hip, or femoral neck at month 48; or (4) had any contraindication to zoledronate. All other subjects were assigned to receive a single IV dose of zoledronate 5 mg. aSubjects transitioned to romosozumab 140 mg QM at month 12, were randomized in the denosumab extension period, completed the study at month 36, and are not included in the present analysis. bSubjects completed the study at month 12 and are not included in the present analysis. cOf the subjects randomized to romosozumab 210 mg QM in the double-blind period, 12 entered the no further active treatment group and 17 entered the zoledronate group during the follow-on phase. IV intravenous, PO orally, QD every day, QM every month, Q3M every 3 months, QW every week, SC subcutaneous
Fig. 2
Fig. 2
Percentage change in BMD at the lumbar spine, total hip, and femoral neck in all subjects who received romosozumab during months 36–48 and who were then assigned to no further active treatment (n = 51; a–c) or zoledronate (n = 90; d–f) from months 48 to 72. Data are mean (95% CI). BMD bone mineral density, CI confidence interval, DMAb denosumab, QM every month, Q6M every 6 months
Fig. 3
Fig. 3
Percentage change in P1NP and β-CTX in all subjects who received romosozumab during months 36–48 and who were then assigned to no further active treatment (n = 51; a, b) or zoledronate (n = 90; c, d) from months 48 to 72. Data are median (Q1, Q3). β-CTX β-isomer of the C-terminal telopeptide of type I collagen, DMAb denosumab, IV intravenous, P1NP procollagen type I N-terminal propeptide, Q1 quartile 1, Q3 quartile 3, QM every month, Q6M every 6 months

References

    1. Ferrari S, Adachi JD, Lippuner K, Zapalowski C, Miller PD, Reginster JY, Torring O, Kendler DL, Daizadeh NS, Wang A, O'Malley CD, Wagman RB, Libanati C, Lewiecki EM. Further reductions in nonvertebral fracture rate with long-term denosumab treatment in the FREEDOM open-label extension and influence of hip bone mineral density after 3 years. Osteoporos Int. 2015;26:2763–2771. doi: 10.1007/s00198-015-3179-x.
    1. Cosman F, Crittenden DB, Ferrari S, Khan A, Lane NE, Lippuner K, Matsumoto T, Milmont CE, Libanati C, Grauer A. FRAME study: the foundation effect of building bone with 1 year of romosozumab leads to continued lower fracture risk after transition to denosumab. J Bone Miner Res. 2018;33:1219–1226. doi: 10.1002/jbmr.3427.
    1. Austin M, Yang YC, Vittinghoff E, Adami S, Boonen S, Bauer DC, Bianchi G, Bolognese MA, Christiansen C, Eastell R, Grauer A, Hawkins F, Kendler DL, Oliveri B, McClung MR, Reid IR, Siris ES, Zanchetta J, Zerbini CA, Libanati C, Cummings SR, Trial F. Relationship between bone mineral density changes with denosumab treatment and risk reduction for vertebral and nonvertebral fractures. J Bone Miner Res. 2012;27:687–693. doi: 10.1002/jbmr.1472.
    1. Bouxsein ML, Eastell R, Lui LY, Wu LA, de Papp AE, Grauer A, Marin F, Cauley JA, Bauer DC, Black DM, Project FBQ Change in bone density and reduction in fracture risk: a meta-regression of published trials. J Bone Miner Res. 2019;34:632–642. doi: 10.1002/jbmr.3641.
    1. Recker RR, Delmas PD, Halse J, Reid IR, Boonen S, Garcia-Hernandez PA, Supronik J, Lewiecki EM, Ochoa L, Miller P, Hu H, Mesenbrink P, Hartl F, Gasser J, Eriksen EF. Effects of intravenous zoledronic acid once yearly on bone remodeling and bone structure. J Bone Miner Res. 2008;23:6–16. doi: 10.1359/jbmr.070906.
    1. Zebaze RM, Libanati C, Austin M, Ghasem-Zadeh A, Hanley DA, Zanchetta JR, Thomas T, Boutroy S, Bogado CE, Bilezikian JP, Seeman E. Differing effects of denosumab and alendronate on cortical and trabecular bone. Bone. 2014;59:173–179. doi: 10.1016/j.bone.2013.11.016.
    1. Jiang Y, Zhao JJ, Mitlak BH, Wang O, Genant HK, Eriksen EF. Recombinant human parathyroid hormone (1-34) [teriparatide] improves both cortical and cancellous bone structure. J Bone Miner Res. 2003;18:1932–1941. doi: 10.1359/jbmr.2003.18.11.1932.
    1. Forteo (teriparatide injection) Prescribing Information (2012) Eli Lilly and Company, Indianapolis, IN, US. . Accessed 19 February 2020
    1. Tymlos™ (abaloparatide) Prescribing Information (2017) Radius health, Inc., Waltham, MA, US. . Accessed 19 February 2020
    1. Leder BZ, Tsai JN, Uihlein AV, Wallace PM, Lee H, Neer RM, Burnett-Bowie SA. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA-switch study): extension of a randomised controlled trial. Lancet. 2015;386:1147–1155. doi: 10.1016/S0140-6736(15)61120-5.
    1. Niimi R, Kono T, Nishihara A, Hasegawa M, Kono T, Sudo A. Efficacy of switching from teriparatide to bisphosphonate or denosumab: a prospective, randomized, open-label trial. JBMR Plus. 2018;2:289–294. doi: 10.1002/jbm4.10054.
    1. Bone HG, Cosman F, Miller PD, Williams GC, Hattersley G, Hu MY, Fitzpatrick LA, Mitlak B, Papapoulos S, Rizzoli R, Dore RK, Bilezikian JP, Saag KG. ACTIVExtend: 24 months of alendronate after 18 months of abaloparatide or placebo for postmenopausal osteoporosis. J Clin Endocrinol Metab. 2018;103:2949–2957. doi: 10.1210/jc.2018-00163.
    1. Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, Lindsay R, Foundation NO Clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2014;25:2359–2381. doi: 10.1007/s00198-014-2794-2.
    1. Camacho PM, Petak SM, Binkley N, Clarke BL, Harris ST, Hurley DL, Kleerekoper M, Lewiecki EM, Miller PD, Narula HS, Pessah-Pollack R, Tangpricha V, Wimalawansa SJ, Watts NB (2016) American Association of Clinical Endocrinologists and American College of Endocrinology Clinical Practice Guidelines for the Diagnosis and Treatment of Postmenopausal Osteoporosis - 2016. Endocr Pract 22:1–42
    1. 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. doi: 10.1002/jbmr.173.
    1. Padhi D, Allison M, Kivitz AJ, Gutierrez MJ, Stouch B, Wang C, Jang G. Multiple doses of sclerostin antibody romosozumab in healthy men and postmenopausal women with low bone mass: a randomized, double-blind, placebo-controlled study. J Clin Pharmacol. 2014;54:168–178. doi: 10.1002/jcph.239.
    1. McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP, Diez-Perez A, Langdahl BL, Reginster JY, Zanchetta JR, Wasserman SM, Katz L, Maddox J, Yang YC, Libanati C, Bone HG. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412–420. doi: 10.1056/NEJMoa1305224.
    1. Chavassieux P, Portero-Muzy N, Roux JP, Horlait S, Dempster DW, Wang A, Wagman RB, Chapurlat R. Reduction of cortical bone turnover and erosion depth after 2 and 3 years of denosumab: iliac bone histomorphometry in the FREEDOM trial. J Bone Miner Res. 2019;34:626–631. doi: 10.1002/jbmr.3631.
    1. Ominsky MS, Brown DL, Van G, Cordover D, Pacheco E, Frazier E, Cherepow L, Higgins-Garn M, Aguirre JI, Wronski TJ, Stolina M, Zhou L, Pyrah I, Boyce RW. Differential temporal effects of sclerostin antibody and parathyroid hormone on cancellous and cortical bone and quantitative differences in effects on the osteoblast lineage in young intact rats. Bone. 2015;81:380–391. doi: 10.1016/j.bone.2015.08.007.
    1. McClung MR, Brown JP, Diez-Perez A, Resch H, Caminis J, Meisner P, Bolognese MA, Goemaere S, Bone HG, Zanchetta JR, Maddox J, Bray S, Grauer A. Effects of 24 months of treatment with romosozumab followed by 12 months of denosumab or placebo in postmenopausal women with low bone mineral density: a randomized, double-blind, phase 2, parallel group study. J Bone Miner Res. 2018;33:1397–1406. doi: 10.1002/jbmr.3452.
    1. Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, Zerbini CA, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375:1532–1543. doi: 10.1056/NEJMoa1607948.
    1. Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, Maddox J, Fan M, Meisner PD, Grauer A. Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med. 2017;377:1417–1427. doi: 10.1056/NEJMoa1708322.
    1. Langdahl BL, Libanati C, Crittenden DB, Bolognese MA, Brown JP, Daizadeh NS, Dokoupilova E, Engelke K, Finkelstein JS, Genant HK, Goemaere S, Hyldstrup L, Jodar-Gimeno E, Keaveny TM, Kendler D, Lakatos P, Maddox J, Malouf J, Massari FE, Molina JF, Ulla MR, Grauer A. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial. Lancet. 2017;390:1585–1594. doi: 10.1016/S0140-6736(17)31613-6.
    1. EVENITY® (romosozumab-aqqg) US Prescribing Information (2019) Amgen Inc., Thousand Oaks, CA, US. . Accessed 24 February 2020
    1. EVENITY® (romosozumab) EU Summary of Product Characteristics (2019) Amgen Inc., Thousand Oaks, CA, US. . Accessed 24 February 2020
    1. Kendler DL, Bone HG, Massari FE, Gielen E, Palacios S, Maddox J, Yan C, Yue S, Dinavahi RV, Libanati C, Grauer A. Bone mineral density gains with a second 12-month course of romosozumab therapy following placebo or denosumab. Osteoporos Int. 2019;30:2437–2448. doi: 10.1007/s00198-019-05146-9.
    1. Anastasilakis AD, Papapoulos SE, Polyzos SA, Appelman-Dijkstra NM, Makras P. Zoledronate for the prevention of bone loss in women discontinuing denosumab treatment: a prospective 2-year clinical trial. J Bone Miner Res. 2019;34:2220–2228. doi: 10.1002/jbmr.3853.
    1. Grey A, Bolland MJ, Horne A, Mihov B, Gamble G, Reid IR. Duration of antiresorptive activity of zoledronate in postmenopausal women with osteopenia: a randomized, controlled multidose trial. CMAJ. 2017;189:e1130–e1136. doi: 10.1503/cmaj.161207.
    1. McClung M, Miller P, Recknor C, Mesenbrink P, Bucci-Rechtweg C, Benhamou CL. Zoledronic acid for the prevention of bone loss in postmenopausal women with low bone mass: a randomized controlled trial. Obstet Gynecol. 2009;114:999–1007. doi: 10.1097/AOG.0b013e3181bdce0a.
    1. Ascott-Evans BH, Guanabens N, Kivinen S, Stuckey BG, Magaril CH, Vandormael K, Stych B, Melton ME. Alendronate prevents loss of bone density associated with discontinuation of hormone replacement therapy: a randomized controlled trial. Arch Intern Med. 2003;163:789–794. doi: 10.1001/archinte.163.7.789.
    1. Freemantle N, Satram-Hoang S, Tang ET, Kaur P, Macarios D, Siddhanti S, Borenstein J, Kendler DL, DAPS Investigators (2012) Final results of the DAPS (Denosumab Adherence Preference Satisfaction) study: a 24-month, randomized, crossover comparison with alendronate in postmenopausal women. Osteoporos Int 23:317–326
    1. Reid IR, Horne AM, Mihov B, Gamble GD. Bone loss after denosumab: only partial protection with zoledronate. Calcif Tissue Int. 2017;101:371–374. doi: 10.1007/s00223-017-0288-x.
    1. Kendler DL, Marin F, Zerbini CAF, Russo LA, Greenspan SL, Zikan V, Bagur A, Malouf-Sierra J, Lakatos P, Fahrleitner-Pammer A, Lespessailles E, Minisola S, Body JJ, Geusens P, Moricke R, Lopez-Romero P. Effects of teriparatide and risedronate on new fractures in post-menopausal women with severe osteoporosis (VERO): a multicentre, double-blind, double-dummy, randomised controlled trial. Lancet. 2018;391:230–240. doi: 10.1016/S0140-6736(17)32137-2.
    1. Cosman F, Nieves JW, Dempster DW. Treatment sequence matters: anabolic and antiresorptive therapy for osteoporosis. J Bone Miner Res. 2017;32:198–202. doi: 10.1002/jbmr.3051.
    1. McClung MR. Using osteoporosis therapies in combination. Curr Osteoporos Rep. 2017;15:343–352. doi: 10.1007/s11914-017-0376-x.

Source: PubMed

3
Subscribe