Single Dose of SHR-1222, a Sclerostin Monoclonal Antibody, in Healthy Men and Postmenopausal Women With Low Bone Mass: A Randomized, Double-Blind, Placebo-Controlled, Dose-Escalation, Phase I Study

Zhijie Dai, Pingfei Fang, Xiang Yan, Ronghua Zhu, Qiong Feng, Qiangyong Yan, Lingfeng Yang, Xiao Fan, Yuting Xie, Lihong Zhuang, Sheng Feng, Yantao Liu, Sheng Zhong, Zeyu Yang, Zhifeng Sheng, Zhiguang Zhou, Zhijie Dai, Pingfei Fang, Xiang Yan, Ronghua Zhu, Qiong Feng, Qiangyong Yan, Lingfeng Yang, Xiao Fan, Yuting Xie, Lihong Zhuang, Sheng Feng, Yantao Liu, Sheng Zhong, Zeyu Yang, Zhifeng Sheng, Zhiguang Zhou

Abstract

SHR-1222 is a humanized monoclonal antibody targeting sclerostin and has the potential to promote bone formation and reduce bone resorption. This study was aimed to assess the safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity of SHR-1222 in healthy men and postmenopausal women with low bone mass (BMD). It was a randomized, double-blind, placebo-controlled, dose-escalation, phase I study. Subjects received SHR-1222 at 50, 100, 200, 300, and 400 mg sequentially or matching placebo subcutaneously. Totally, 50 subjects with low BMD were enrolled and randomly assigned; 10 received placebo and 40 received SHR-1222 (50 mg, n = 4; 100, 200, 300, or 400 mg, n = 9). The most common adverse events that occurred at least 10% higher in subjects with SHR-1222 treatment than those with placebo were decreased blood calcium, blood urine present, increased blood cholesterol, electrocardiogram T wave abnormal, urinary tract infection, increased blood pressure diastolic, and positive bacterial test. All the above adverse events were mild in severity and well resolved except one of increased blood cholesterol in a subject lost to follow-up. The serum SHR-1222 concentration increased in a dose-dependent manner. Administration of SHR-1222 upregulated the bone-formation markers N-terminal propeptide of type 1 procollagen, osteocalcin, and bone-specific alkaline phosphatase, while downregulated the bone-resorption marker β-C-telopeptide. The BMD at the lumbar spine notably rose after a single dose of SHR-1222. The largest increase occurred in the 400 mg cohort (3.8, 6.7, and 6.1% on day 29, 57, and 85, respectively; compared with 1.4, 0.8, and 1.0% in the placebo group). Although 10.0% of subjects receiving SHR-1222 tested positive for anti-SHR-1222 antibodies, no obvious effects of antibody formation were found on pharmacokinetics. Overall, SHR-1222 was well tolerated at doses from 50 to 400 mg and is a promising new remedy for osteoporosis. Clinical Trial Registration: http://www.clinicaltrials.gov, NCT03870100.

Keywords: SHR-1222; bone formation; bone resorption; low bone mass; sclerostin.

Conflict of interest statement

Authors LZ, SF, YL, SZ and ZY were employed by Jiangsu Hengrui Pharmaceuticals Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this study received funding from Jiangsu Hengrui Pharmaceuticals Co., Ltd.. The funder had the following involvement with the study: design, data collection and analysis.

Copyright © 2021 Dai, Fang, Yan, Zhu, Feng, Yan, Yang, Fan, Xie, Zhuang, Feng, Liu, Zhong, Yang, Sheng and Zhou.

Figures

FIGURE 1
FIGURE 1
Study design. (A) Framework for the dose-escalation; (B) Dosing and assessment schema.
FIGURE 2
FIGURE 2
Mean serum concentration-time profiles of SHR-1222.
FIGURE 3
FIGURE 3
The effect of SHR-1222 on bone turnover markers. P1NP, N-terminal propeptide of type 1 procollagen; OST, osteocalcin; BSAP, bone-specific alkaline phosphatase; CTx, C-telopeptide.
FIGURE 4
FIGURE 4
The effect of SHR-1222 on bone mineral density at lumbar spine L1-L4.

References

    1. Armas L. A., Recker R. R. (2012). Pathophysiology of Osteoporosis: New Mechanistic Insights. Endocrinol. Metab. Clin. North. Am. 41 (3), 475–486. 10.1016/j.ecl.2012.04.006
    1. Burr D. B., Miller L., Grynpas M., Li J., Boyde A., Mashiba T., et al. (2003). Tissue Mineralization Is Increased Following 1-year Treatment with High Doses of Bisphosphonates in Dogs. Bone 33 (6), 960–969. 10.1016/j.bone.2003.08.004
    1. Cano A., Chedraui P., Goulis D. G., Lopes P., Mishra G., Mueck A., et al. (2018). Calcium in the Prevention of Postmenopausal Osteoporosis: EMAS Clinical Guide. Maturitas 107, 7–12. 10.1016/j.maturitas.2017.10.004
    1. Cosman F., Crittenden D. B., Adachi J. D., Binkley N., Czerwinski E., Ferrari S., et al. (2016). Romosozumab Treatment in Postmenopausal Women with Osteoporosis. N. Engl. J. Med. 375 (16), 1532–1543. 10.1056/NEJMoa1607948
    1. Coughlan T., Dockery F. (2014). Osteoporosis and Fracture Risk in Older People. Clin. Med. (Lond) 14 (2), 187–191. 10.7861/clinmedicine.14-2-187
    1. Drake M. T., Clarke B. L., Khosla S. (2008). Bisphosphonates: Mechanism of Action and Role in Clinical Practice. Mayo Clin. Proc. 83 (9), 1032–1045. 10.4065/83.9.1032
    1. EMA (2019). Evenity (Romosozumab) an Overview of Evenity and Why it Is Authorised in the EU. Available at: (Accessed September 25, 2021).
    1. FDA (2019). Prescribing Information for Romosozumab. Available at: (Accessed May 25, 2021).
    1. Fulzele K., Lai F., Dedic C., Saini V., Uda Y., Shi C., et al. (2017). Osteocyte-Secreted Wnt Signaling Inhibitor Sclerostin Contributes to Beige Adipogenesis in Peripheral Fat Depots. J. Bone Miner Res. 32 (2), 373–384. 10.1002/jbmr.3001
    1. Hangartner T. N. (2007). A Study of the Long-Term Precision of Dual-Energy X-ray Absorptiometry Bone Densitometers and Implications for the Validity of the Least-Significant-Change Calculation. Osteoporos. Int. 18 (4), 513–523. 10.1007/s00198-006-0280-1
    1. Hodsman A. B., Bauer D. C., Dempster D. W., Dian L., Hanley D. A., Harris S. T., et al. (2005). Parathyroid Hormone and Teriparatide for the Treatment of Osteoporosis: a Review of the Evidence and Suggested Guidelines for its Use. Endocr. Rev. 26 (5), 688–703. 10.1210/er.2004-0006
    1. Kanis J. A. (2002). Diagnosis of Osteoporosis and Assessment of Fracture Risk. Lancet 359 (9321), 1929–1936. 10.1016/S0140-6736(02)08761-5
    1. Kim S. P., Frey J. L., Li Z., Kushwaha P., Zoch M. L., Tomlinson R. E., et al. (2017). Sclerostin Influences Body Composition by Regulating Catabolic and Anabolic Metabolism in Adipocytes. Proc. Natl. Acad. Sci. U S A. 114, E11238. 10.1073/pnas.1707876115
    1. Li X., Zhang Y., Kang H., Liu W., Liu P., Zhang J., et al. (2005). Sclerostin Binds to LRP5/6 and Antagonizes Canonical Wnt Signaling. J. Biol. Chem. 280 (20), 19883–19887. 10.1074/jbc.M413274200
    1. Lindsay R., Krege J. H., Marin F., Jin L., Stepan J. J. (2016). Teriparatide for Osteoporosis: Importance of the Full Course. Osteoporos. Int. 27 (8), 2395–2410. 10.1007/s00198-016-3534-6
    1. Padhi D., Allison M., Kivitz A. J., Gutierrez M. J., Stouch B., Wang C., et al. (2014). 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. 54 (2), 168–178. 10.1002/jcph.239
    1. Padhi D., Jang G., Stouch B., Fang L., Posvar E. (2011). Single-dose, Placebo-Controlled, Randomized Study of AMG 785, a Sclerostin Monoclonal Antibody. J. Bone Miner Res. 26 (1), 19–26. 10.1002/jbmr.173
    1. Paschalis E. P., Gamsjaeger S., Hassler N., Fahrleitner-Pammer A., Dobnig H., Stepan J. J., et al. (2017). Vitamin D and Calcium Supplementation for Three Years in Postmenopausal Osteoporosis Significantly Alters Bone mineral and Organic Matrix Quality. Bone 95, 41–46. 10.1016/j.bone.2016.11.002
    1. Paszty C., Turner C. H., Robinson M. K. (2010). Sclerostin: a Gem from the Genome Leads to Bone-Building Antibodies. J. Bone Miner Res. 25 (9), 1897–1904. 10.1002/jbmr.161
    1. Poole K. E., van Bezooijen R. L., Loveridge N., Hamersma H., Papapoulos S. E., Löwik C. W., et al. (2005). Sclerostin Is a Delayed Secreted Product of Osteocytes that Inhibits Bone Formation. FASEB J. 19 (13), 1842–1844. 10.1096/fj.05-4221fje
    1. Saag K. G., Petersen J., Brandi M. L., Karaplis A. C., Lorentzon M., Thomas T., et al. (2017). Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N. Engl. J. Med. 377 (15), 1417–1427. 10.1056/NEJMoa1708322
    1. Semënov M., Tamai K., He X. (2005). SOST Is a Ligand for LRP5/LRP6 and a Wnt Signaling Inhibitor. J. Biol. Chem. 280 (29), 26770–26775. 10.1074/jbc.M504308200
    1. Sølling A. S. K., Harsløf T., Langdahl B. (2019). Current Status of Bone-Forming Therapies for the Management of Osteoporosis. Drugs Aging 36 (7), 625–638. 10.1007/s40266-019-00675-8
    1. Weaver C. M., Alexander D. D., Boushey C. J., Dawson-Hughes B., Lappe J. M., LeBoff M. S., et al. (2016). Calcium Plus Vitamin D Supplementation and Risk of Fractures: an Updated Meta-Analysis from the National Osteoporosis Foundation. Osteoporos. Int. 27 (1), 367–376. 10.1007/s00198-015-3386-5
    1. Wijenayaka A. R., Kogawa M., Lim H. P., Bonewald L. F., Findlay D. M., Atkins G. J. (2011). Sclerostin Stimulates Osteocyte Support of Osteoclast Activity by a RANKL-dependent Pathway. PLoS One 6 (10), e25900. 10.1371/journal.pone.0025900
    1. Winkler D. G., Sutherland M. K., Geoghegan J. C., Yu C., Hayes T., Skonier J. E., et al. (2003). Osteocyte Control of Bone Formation via Sclerostin, a Novel BMP Antagonist. EMBO J. 22 (23), 6267–6276. 10.1093/emboj/cdg599
    1. Zeng Q., Li N., Wang Q., Feng J., Sun D., Zhang Q., et al. (2019). The Prevalence of Osteoporosis in China, a Nationwide, Multicenter DXA Survey. J. Bone Miner Res. 34 (10), 1789–1797. 10.1002/jbmr.3757

Source: PubMed

3
구독하다