Affinity targeting of therapeutic proteins to the bone surface-local delivery of sclerostin-neutralizing antibody enhances efficacy

Boya Zhang,William Benton Swanson,Margaret Durdan, Heather N. Livingston, Michaela Dodd, Sachith M. Vidanapathirana,Alec Desai, Lindsey Douglas,Yuji Mishina,Megan Weivoda,Colin F. Greineder

JOURNAL OF BONE AND MINERAL RESEARCH(2024)

引用 0|浏览0
暂无评分
摘要
Currently available biotherapeutics for the treatment of osteoporosis lack explicit mechanisms for bone localization, potentially limiting efficacy and inducing off-target toxicities. While various strategies have been explored for targeting the bone surface, critical aspects remain poorly understood, including the optimal affinity ligand, the role of binding avidity and circulation time, and, most importantly, whether or not this strategy can enhance the functional activity of clinically relevant protein therapeutics. To investigate, we generated fluorescent proteins (eg, mCherry) with site-specifically attached small molecule (bisphosphonate) or peptide (deca-aspartate, D-10) affinity ligands. While both affinity ligands successfully anchored fluorescent protein to the bone surface, quantitative radiotracing revealed only modest femoral and vertebral accumulation and suggested a need for enhanced circulation time. To achieve this, we fused mCherry to the Fc fragment of human IgG(1) and attached D-10 peptides to each C-terminus. The mCherry-Fc-D-10 demonstrated an similar to 80-fold increase in plasma exposure and marked increases in femoral and vertebral accumulation (13.6% +/- 1.4% and 11.4% +/- 1.3% of the injected dose/g [%ID/g] at 24 h, respectively). To determine if bone surface targeting could enhance the efficacy of a clinically relevant therapeutic, we generated a bone-targeted sclerostin-neutralizing antibody, anti-sclerostin-D-10. The targeted antibody demonstrated marked increases in bone accumulation and retention (20.9 +/- 2.5% and 19.5 +/- 2.5% ID/g in femur and vertebrae at 7 days) and enhanced effects in a murine model of ovariectomy-induced bone loss (bone volume/total volume, connectivity density, and structure model index all increased [P < .001] vs untargeted anti-sclerostin). Collectively, our results indicate the importance of both bone affinity and circulation time in achieving robust targeting of therapeutic proteins to the bone surface and suggest that this approach may enable lower doses and/or longer dosing intervals without reduction in biotherapeutic efficacy. Future studies will be needed to determine the translational potential of this strategy and its potential impact on off-site toxicities.
更多
查看译文
关键词
osteoporosis,diseases and disorders of/related to bone,preclinical studies,animal models,wnt/beta-catenin/lrps,cell/tissue signaling-paracrine pathways,therapeutics,anabolics,therapeutics
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要