Your browser doesn't support javascript.
loading
Affinity targeting of therapeutic proteins to the bone surface-local delivery of sclerostin-neutralizing antibody enhances efficacy.
Zhang, Boya; Swanson, William Benton; Durdan, Margaret; Livingston, Heather N; Dodd, Michaela; Vidanapathirana, Sachith M; Desai, Alec; Douglas, Lindsey; Mishina, Yuji; Weivoda, Megan; Greineder, Colin F.
Afiliación
  • Zhang B; Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA.
  • Swanson WB; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
  • Durdan M; Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA.
  • Livingston HN; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
  • Dodd M; Department of Hematology, Mayo Clinic, Rochester, MN 55905, USA.
  • Vidanapathirana SM; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
  • Desai A; Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
  • Douglas L; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
  • Mishina Y; Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
  • Weivoda M; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
  • Greineder CF; Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
J Bone Miner Res ; 39(6): 717-728, 2024 Jul 23.
Article en En | MEDLINE | ID: mdl-38526976
ABSTRACT
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, D10) 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 IgG1 and attached D10 peptides to each C-terminus. The mCherry-Fc-D10 demonstrated an ~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-D10. 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.
Several biologic therapies have been approved for osteoporosis, but they lack means of localization to bone tissue, potentially limiting their efficacy and leading to off-target toxicities. This manuscript investigates strategies for targeting biotherapeutics to the bone surface and asks the question of whether or not this approach can enhance functional activity and allow for lower or less frequent dosing. To define the key determinants of bone surface targeting, we begin by synthesizing fluorescent model proteins with different bone targeting tags. We show that even 1 tag is enough to make the surface of the femur and vertebrae fluorescent following systemic administration. The results are relatively modest at first, but when we combine the bone targeting tag with a second modification that makes the protein circulate in the body for a longer period of time, we observe a huge increase in bone surface delivery. We then synthesize a bone surface targeted version of a sclerostin-inhibiting antibody and show that it is more effective than the untargeted antibody and provides near complete protection of bone density despite relatively low dose. Our findings could have translational implications for existing bone therapies and help guide design of future strategies for optimized bone surface targeting.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Anticuerpos Neutralizantes Límite: Animals / Female / Humans Idioma: En Revista: J Bone Miner Res Asunto de la revista: METABOLISMO / ORTOPEDIA Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Anticuerpos Neutralizantes Límite: Animals / Female / Humans Idioma: En Revista: J Bone Miner Res Asunto de la revista: METABOLISMO / ORTOPEDIA Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos