@phdthesis{Schultz2015, author = {Schultz, Isabel}, title = {Therapeutic systems for Insulin-like growth factor-1}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119114}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {SUMMARY Insulin-like growth factor I (IGF-I) is a polypeptide with a molecular weight of 7.649 kDa and an anabolic potential. Thereby, IGF-I has a promising therapeutic value e.g. in muscle wasting diseases such as sarcopenia. IGF-I is mainly secreted by the liver in response to growth hormone (GH) stimulation and is rather ubiquitously found within all tissues. The effects of IGF-I are mediated by its respective IGF-I transmembrane tyrosine kinase receptor triggering the stimulation of protein synthesis, glucose uptake and the regulation of cell growth. The actions of IGF-I are modulated by six IGF binding proteins binding and transporting IGF-I in a binary or ternary complex to tissues and receptors and modulating the binding of IGF-I to its receptor. The nature of the formed complexes impacts IGF-I`s half-life, modulating the half-life between 10 minutes (free IGF-I) to 12 - 15 hours when presented in a ternary complex with IGF binding protein 3 and an acid labile subunit (ALS). Therefore, sustained drug delivery systems of free IGF-I are superficially seen as interesting for the development of controlled release profiles, as the rate of absorption is apparently and easily set slower by simple formulation as compared to the rapid rate of elimination. Thereby, one would conclude, the formulation scientist can rapidly develop systems for which the pharmacokinetics of IGF-I are dominated by the formulation release kinetics. However, the in vivo situation is more complex and as mentioned (vide supra), the half-life may easily be prolonged up to hours providing proper IGF-I complexation takes place upon systemic uptake. These and other aspects are reviewed in Chapter I, within which we introduce IGF-I as a promising therapeutic agent detailing its structure and involved receptors along with the resulting signaling pathways. We summarize the control of IGF-I pharmacokinetics in nature within the context of its complex system of 6 binding proteins to control half-life and tissue distribution. Furthermore, we describe IGF-I variants with modulated properties in vivo and originated from alternative splicing. These insights were translated into sophisticated IGF-I delivery systems for therapeutic use. Aside from safety aspects, the challenges and requirements of an effective IGF-I therapy are discussed. Localized and systemic IGF-I delivery strategies, different routes of administration as well as liquid and solid IGF-I formulations are reviewed. Effective targeting of IGF-I by protein decoration is outlined and consequently this chapter provides an interesting guidance for successful IGF-I-delivery. In Chapter II, we firstly outline the stability of IGF-I in liquid formulations with the intention to deliver the biologic through the lung and the impact of buffer type, sodium chloride concentration and pH value on IGF-I stability is presented. IGF-I integrity was preserved in histidine buffer over 4 months at room temperature, but methionine 59 oxidation (Met(o)) along with reducible dimer and trimer formation was observed in an acidic environment (pH 4.5) and using acetate buffer. Strong aggregation resulted in a complete loss of IGF-I bioactivity, whereas the potency was partly maintained in samples showing a slight aggregation and complete IGF-I oxidation. Atomization by air-jet or vibrating-mesh nebulizers yielded in limited Met(o) formation and no aggregation. The results of IGF-I nebulization experiments regarding aerosol output rate, mass median aerodynamic diameter and fine particle fraction were comparable with 0.9\% sodium chloride reference, approving the applicability of liquid IGF-I formulations for pulmonary delivery. In Chapter III we escalated the development to solid delivery systems designed for alveolar landing upon inhalation and by deploying trehalose and the newly introduced for pulmonary application silk-fibroin as carriers. Microparticles were produced using nano spray drying following analyses including IGF-I integrity, IGF-I release profiles and aerodynamic properties. In vitro transport kinetics of IGF-I across pulmonary Calu-3 epithelia were suggesting similar permeability as compared to IGF-I's cognate protein, insulin that has already been successfully administered pulmonary in clinical settings. These in vivo results were translated to an ex vivo human lung lobe model. This work showed the feasibility of pulmonary IGF-I delivery and the advantageous diversification of excipients for pulmonary formulations using silk-fibroin. Chapter IV focuses on an innovative strategy for safe and controllable IGF-I delivery. In that chapter we escalated the development to novel IGF-I analogues. The intention was to provide a versatile biologic into which galenical properties can be engineered through chemical synthesis, e.g. by site directed coupling of polymers to IGF-I. For this purpose we genetically engineered two IGF-I variants containing an unnatural amino acid at two positions, respectively, thereby integrating alkyne functions into the primary sequence of the protein. These allowed linking IGF-I with other molecules in a site specific manner, i.e. via a copper catalyzed azide-alkyne Huisgen cycloaddition (click reaction). In this chapter we mainly introduce the two IGF-I variants, detail the delivery concept and describe the optimization of the expression conditions of the IGF-I variants. In conclusion, we span from simple liquid formulations for aerolization through solid systems for tailored for maximal alveolar landing to novel engineered IGF-I analogues. Thereby, three strategies for advanced IGF-I delivery were addressed and opportunities and limitations of each were outlined. Evidence was provided that sufficiently stable and easy to manufacture formulations can be developed as typically required for first in man studies. Interestingly, solid systems - typically introduced in later stages of pharmaceutical development - were quite promising. By use of silk-fibroin as a new IGF-I carrier for pulmonary administration, a new application was established for this excipient. The demonstrated success using the ex vivo human lung lobe model provided substantial confidence that pulmonary IGF-I delivery is possible in man. Finally, this work describes the expression of two IGF-I variants containing two unnatural amino acids to implement an innovative strategy for IGF-I delivery. This genetic engineering approach was providing the fundament for novel IGF-I analogues. Ideally, the biologic is structurally modified by covalently linked moieties for the control of pharmacokinetics or for targeted delivery, e.g. into sarcopenic muscles. One future scenario is dicussed in the 'conclusion and outlook' section for which IGF-I is tagged to a protease sensitive linker peptide and this linker peptide in return is coupled to a polyethylenglykole (PEG) polymer (required to prolong the half-life). Some proteases may serve as proxy for sarcopenia such that protease upregulation in compromised muscle tissues drives cleavage of IGF-I from the PEG. Thereby, IGF-I is released at the seat of the disease while systemic side effects are minimized.}, subject = {Insulin-like Growth Factor I}, language = {en} }