학술논문

Sustained plasma hepcidin suppression and iron elevation by Anticalin-derived hepcidin antagonist in cynomolgus monkey.
Document Type
Journal Article
Source
British Journal of Pharmacology. Apr2018, Vol. 175 Issue 7, p1054-1065. 12p. 2 Charts, 6 Graphs.
Subject
*HEPCIDIN
*IRON deficiency anemia
*KRA
*IMMUNOSUPPRESSION
*PHARMACOKINETICS
*PHARMACODYNAMICS
*ANIMAL experimentation
*BIOLOGICAL models
*COMPARATIVE studies
*IRON
*RESEARCH methodology
*MEDICAL cooperation
*PEPTIDES
*PRIMATES
*RESEARCH
*EVALUATION research
*CHEMICAL inhibitors
Language
ISSN
0007-1188
Abstract
Background and Purpose: Anaemia of chronic disease (ACD) has been linked to iron-restricted erythropoiesis imposed by high circulating levels of hepcidin, a 25 amino acid hepatocyte-derived peptide that controls systemic iron homeostasis. Here, we report the engineering of the human lipocalin-derived, small protein-based anticalin PRS-080 hepcidin antagonist with high affinity and selectivity.Experimental Approach: Anticalin- and hepcidin-specific pharmacokinetic (PK)/pharmacodynamic modelling (PD) was used to design and select the suitable drug candidate based on t1/2 extension and duration of hepcidin suppression. The development of a novel free hepcidin assay enabled accurate analysis of bioactive hepcidin suppression and elucidation of the observed plasma iron levels after PRS-080-PEG30 administration in vivo.Key Results: PRS-080 had a hepcidin-binding affinity of 0.07 nM and, after coupling to 30 kD PEG (PRS-080-PEG30), a t1/2 of 43 h in cynomolgus monkeys. Dose-dependent iron mobilization and hepcidin suppression were observed after a single i.v. dose of PRS-080-PEG30 in cynomolgus monkeys. Importantly, in these animals, suppression of free hepcidin and subsequent plasma iron elevation were sustained during repeated s.c. dosing. After repeated dosing and followed by a treatment-free interval, all iron parameters returned to pre-dose values.Conclusions and Implications: In conclusion, we developed a dose-dependent and safe approach for the direct suppression of hepcidin, resulting in prolonged iron mobilization to alleviate iron-restricted erythropoiesis that can address the root cause of ACD. PRS-080-PEG30 is currently in early clinical development. [ABSTRACT FROM AUTHOR]