1
|
Wang Y, Shen L, Wang C, Dong Y, Hua H, Xu J, Zhang Y, Huang H, Huang Z, Zhao F, Xu Z, Qiu Y, Lu J, Ju D, Feng J. Lipidation-dimerization platform unlocks treatment potential of fibroblast growth factor 21 for non-alcoholic steatohepatitis. J Control Release 2024; 376:1130-1142. [PMID: 39510256 DOI: 10.1016/j.jconrel.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Optimizing the druggability of both native and AI-designed bioactive proteins is crucial for realizing their therapeutic potential. A key focus in designing protein-based therapeutics is improving their pharmacokinetic properties. However, a significant challenge is to preserve biological activity while implementing long-acting strategies. Fibroblast growth factor 21 (FGF21), an endogenous hormone with potential as a treatment for non-alcoholic steatohepatitis (NASH), exemplifies this challenge. In this study, we present a novel lipidation-dimerization (LiDi) platform that integrates lipidation with a dimeric form of FGF21 connected by a hydrophilic linker. The lipidation enhances albumin binding, enabling sustained release, while the dimeric structure boosts biological activity. In vivo evaluations of the LiDi FGF21 analogs demonstrated that they offer excellent pharmacokinetic properties and superior efficacy compared to other treatments for NASH. This platform effectively extends the therapeutic half-life of proteins without compromising their activity, substantially broadening the application range of proteins as therapeutics.
Collapse
Affiliation(s)
- Yapeng Wang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, 201203 Shanghai, China; National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China
| | - Lei Shen
- Anhui University of Traditional Chinese Medicine School of Pharmacy, 230013 Hefei, China; Yangtze Delta Drug Advanced Research Institute, 226133 Nantong, China; Shanghai Innostar Bio-tech Nantong Co., Ltd., 226133 Nantong, China
| | - Chengcheng Wang
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; School of Pharmacy, Shanghai Jiao Tong University School of Medicine, 200240 Shanghai, China
| | - Yuanzhen Dong
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Shanghai Duomirui Bio-tech Co., Ltd., 201203 Shanghai, China
| | - Haoju Hua
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Shanghai Duomirui Bio-tech Co., Ltd., 201203 Shanghai, China
| | - Jun Xu
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Shanghai Duomirui Bio-tech Co., Ltd., 201203 Shanghai, China
| | - Ying Zhang
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China
| | - Hao Huang
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China
| | - Zongqing Huang
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; School of Pharmacy, Shanghai Jiao Tong University School of Medicine, 200240 Shanghai, China
| | - Fei Zhao
- Shanghai Innostar Bio-tech Nantong Co., Ltd., 226133 Nantong, China
| | - Zhiru Xu
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry Co., Ltd, China State Institute of Pharmaceutical Industry Co., Ltd., 200083 Shanghai, China
| | - Yunliang Qiu
- Shanghai Innostar Bio-tech Nantong Co., Ltd., 226133 Nantong, China
| | - Jianguang Lu
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Shanghai Duomirui Bio-tech Co., Ltd., 201203 Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, 201203 Shanghai, China.
| | - Jun Feng
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry Co., Ltd., 201203 Shanghai, China; Shanghai Duomirui Bio-tech Co., Ltd., 201203 Shanghai, China.
| |
Collapse
|
2
|
Shangguan W, Li X, Wang Y, Huang Z, Dong Y, Feng M, Feng J. Design and Biological Evaluation of the Long-Acting C5-Inhibited Ornithodoros moubata Complement Inhibitor (OmCI) Modified with Fatty Acid. Bioconjug Chem 2024; 35:653-664. [PMID: 38593046 DOI: 10.1021/acs.bioconjchem.4c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.
Collapse
Affiliation(s)
- Wenwen Shangguan
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Xiaowan Li
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yandan Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 310014 Hangzhou, China
| | - Zongqing Huang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Yuanzhen Dong
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| |
Collapse
|
3
|
Wang XD, Su ZH, Du J, Yu WJ, Sun WL. Site-selective fatty acid chain conjugation of the N-terminus of the recombinant human granulocyte colony-stimulating factor. Front Bioeng Biotechnol 2024; 12:1360506. [PMID: 38576447 PMCID: PMC10993259 DOI: 10.3389/fbioe.2024.1360506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
The clinical application of the recombinant human granulocyte colony-stimulating factor (rhG-CSF) is restricted by its short serum half-life. Herein, site-selective modification of the N-terminus of rhG-CSF with PAL-PEG3-Ph-CHO was used to develop a long-acting rhG-CSF. The optimized conditions for rhG-CSF modification with PAL-PEG3-Ph-CHO were: reaction solvent system of 3% (w/v) Tween 20 and 30 mM NaCNBH3 in acetate buffer (20 mmol/L, pH 5.0), molar ratio of PAL-PEG3-Ph-CHO to rhG-CSF of 6:1, temperature of 20°C, and reaction time of 12 h, consequently, achieving a PAL-PEG3-Ph-rhG-CSF product yield of 70.8%. The reaction mixture was purified via preparative liquid chromatography, yielding the single-modified product PAL-PEG3-Ph-rhG-CSF with a HPLC purity exceeding 95%. The molecular weight of PAL-PEG3-Ph-rhG-CSF was 19297 Da by MALDI-TOF-MS, which was consistent with the theoretical value. The circular dichroism analysis revealed no significant change in its secondary structure compared to unmodified rhG-CSF. The PAL-PEG3-Ph-rhG-CSF retained 82.0% of the in vitro biological activity of unmodified rhG-CSF. The pharmacokinetic analyses showed that the serum half-life of PAL-PEG3-Ph-rhG-CSF was 7.404 ± 0.777 h in mice, 4.08 times longer than unmodified rhG-CSF. Additionally, a single subcutaneous dose of PAL-PEG3-Ph-rhG-CSF presented comparable in vivo efficacy to multiple doses of rhG-CSF. This study demonstrated an efficacious strategy for developing long-acting rhG-CSF drug candidates.
Collapse
Affiliation(s)
- Xu-Dong Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Zhi-Hao Su
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jie Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Wei-Jia Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Wen-Long Sun
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, China
| |
Collapse
|
4
|
Wang Y, Kim M, Buckley C, Maynard HD, Langley RJ, Perry JK. Growth hormone receptor agonists and antagonists: From protein expression and purification to long-acting formulations. Protein Sci 2023; 32:e4727. [PMID: 37428391 PMCID: PMC10443362 DOI: 10.1002/pro.4727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
Recombinant human growth hormone (rhGH) and GH receptor antagonists (GHAs) are used clinically to treat a range of disorders associated with GH deficiency or hypersecretion, respectively. However, these biotherapeutics can be difficult and expensive to manufacture with multiple challenges from recombinant protein generation through to the development of long-acting formulations required to improve the circulating half-life of the drug. In this review, we summarize methodologies and approaches used for making and purifying recombinant GH and GHA proteins, and strategies to improve pharmacokinetic and pharmacodynamic properties, including PEGylation and fusion proteins. Therapeutics that are in clinical use or are currently under development are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- Liggins Institute, University of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
| | - Minah Kim
- Liggins Institute, University of AucklandAucklandNew Zealand
| | - Chantal Buckley
- Liggins Institute, University of AucklandAucklandNew Zealand
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry and the California NanoSystems InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Ries J. Langley
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Jo K. Perry
- Liggins Institute, University of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryAucklandNew Zealand
| |
Collapse
|
5
|
Villarruel LA, Brie B, Municoy S, Becú-Villalobos D, Desimone MF, Catalano PN. Silica-collagen nanoformulations with extended human growth hormone release. Int J Pharm 2023; 634:122662. [PMID: 36736675 DOI: 10.1016/j.ijpharm.2023.122662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Growth hormone deficiency has been treated by the daily administration of recombinant human growth hormone (hGH) for decades. Patient compliance to this treatment is generally incomplete due to challenges including dose frequency and lack of perceived benefits. This stimulates the research on new formulations to reduce the number of periodic administrations. In this study silica nanoparticles and silica-collagen nanocomposites were evaluated for hGH loading and release. Bare nanoparticles showed higher hGH adsorption capacity than thiol- and isobutyl-bearing particles of similar diameters. Monitoring of bound protein conformation changes indicated hGH structure retention when adsorbed on bare silica nanoparticles and suggested no alterations on protein activity. Protein-loaded particles incorporated into collagen matrices (silica-collagen nanocomposites) showed a progressive protein release profile different from the observed for hGH-loaded silica nanoparticles and hGH-loaded collagen matrices. While both the collagen and the silica nanoparticle systems reached a 100 % release after 4 and 7 days respectively, silica-collagen nanocomposites showed a bi-phasic prolonged hGH release reaching approximately an 80 % after 15 days. These findings suggest that biocompatible silica-collagen nanocomposites could be used as vehicles for the prolonged delivery of hGH which could lead to a potential reduction in the number of periodic administrations.
Collapse
Affiliation(s)
- Luis A Villarruel
- Instituto de Nanociencia y Nanotecnología (CNEA - CONICET), Nodo Constituyentes, Av. Gral.Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Departamento de Micro y Nanotecnología, Gerencia de Desarrollo Tecnológico y Proyectos Especiales, Gerencia de Área de Investigación y Aplicaciones No Nucleares, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica, Av. Gral. Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Belén Brie
- Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490 (C1428ADN), Buenos Aires, Argentina
| | - Sofía Municoy
- Universidad de Buenos Aires (UBA), CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina
| | - Damasia Becú-Villalobos
- Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490 (C1428ADN), Buenos Aires, Argentina
| | - Martín F Desimone
- Universidad de Buenos Aires (UBA), CONICET, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina.
| | - Paolo N Catalano
- Instituto de Nanociencia y Nanotecnología (CNEA - CONICET), Nodo Constituyentes, Av. Gral.Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Departamento de Micro y Nanotecnología, Gerencia de Desarrollo Tecnológico y Proyectos Especiales, Gerencia de Área de Investigación y Aplicaciones No Nucleares, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica, Av. Gral. Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 954 (1113), Buenos Aires, Argentina.
| |
Collapse
|
6
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
7
|
Helleberg H, Lindecrona RH, Thygesen P, Bjelke M. Structure identification of circulating metabolites from somapacitan, a long-acting growth hormone derivative, and pharmacokinetics after single and multiple subcutaneous dosing in rats. Eur J Pharm Sci 2022; 168:106032. [PMID: 34610450 DOI: 10.1016/j.ejps.2021.106032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/31/2021] [Accepted: 09/30/2021] [Indexed: 11/03/2022]
Abstract
Somapacitan is a growth hormone derivative approved for once-weekly treatment of growth hormone deficiency in adults and currently in clinical development for once-weekly dosing in children. The purpose of this study was to obtain non-clinical data from rats to support the safety evaluation of the most abundant metabolites of somapacitan in humans. The aims were to identify somapacitan metabolites and their relative proportions in rat plasma, identify the structure of abundant metabolites and measure the systemic metabolite exposure at the no-observed-adverse-effect level in the rat. After a single dose of radiolabelled somapacitan and analysis by high-performance liquid chromatography with radiochemical detection, seven somapacitan-related metabolites were detected in plasma from male rats, of which six were seen in plasma from female rats. The three most abundant metabolites (M1, M2 and M3) were structurally identified from liquid chromatography and mass spectrometry data, and a fourth metabolite (P1) was characterised from its specific retention time (lacking retention to the stationary phase) in plasma analysis with reversed-phase liquid chromatography and radiochemical detection. The metabolites were products from proteolysis of the peptide backbone in somapacitan. A deamidation product of the M1 metabolite (M1B) was also identified. Following multiple, twice-weekly dosing for 4 weeks, somapacitan was the principal plasma component up to 36 h after dosing. After 36 h, metabolites M1+M1B were the most abundant plasma components. Pharmacokinetic models were developed for somapacitan and metabolite P1 and used for steady-state assessment in the rat. Comparison of our data generated from rats with data from the parallel human study demonstrated that the most abundant metabolites were present in rats at higher levels than in humans. This study has provided non-clinical safety data that contribute to an overall safety assessment of somapacitan.
Collapse
Affiliation(s)
- Hans Helleberg
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark.
| | | | - Peter Thygesen
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | - Mads Bjelke
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
8
|
Ruan S, Yang G, Dong Y, Shangguan W, Lu W. Discovery of a Long-Acting Parathyroid Hormone 1-34 Analogue to Treat Hypoparathyroidism. Mol Pharm 2021; 18:3260-3271. [PMID: 34482698 DOI: 10.1021/acs.molpharmaceut.1c00149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hypoparathyroidism (HP) is a rare disease with clinical manifestations of hypocalcemia and hyperphosphatemia, resulting from deficient or absent parathyroid hormone (PTH) secretion. Conventional treatment for patients with HP involves extensive calcium and vitamin D supplementation. In 2015, PTH1-84 was approved by the United States Food and Drug Administration as an adjunct for HP patients who cannot be well-controlled on conventional treatment. However, PTH1-84 therapy requires a daily injection, leading to poor patient compliance. The purpose of this study was to develop a long-acting PTH1-34 analogue by increasing its affinity to albumin. Three PTH1-34 variants were generated by substituting two of the three lysine (Lys) residues with arginine, reserving a single Lys as the modification site in each sequence. A series of side chains, containing fatty acid, deoxycholic acid, or biotin groups, were synthesized to modify these PTH1-34 variants by using a solid-liquid phase synthesis approach. In vitro bioactivity and albumin affinity tests were used to screen these new PTH1-34 analogues. Finally, Lys27-AAPC was selected from 69 synthesized analogues as a candidate therapeutic compound because it retained potency and exhibited a high albumin-binding capacity. In pharmacodynamic experiments, Lys27-AAPC demonstrated enhanced and prolonged efficacy in serum calcium elevating relative to PTH1-84. Moreover, a lyophilized powder for injection containing Lys27-AAPC was developed for further testing and represented a potential long-acting HP treatment.
Collapse
Affiliation(s)
- Sida Ruan
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Guiying Yang
- Shanghai Duomirui Biotechnology Ltd., Shanghai 201203, China
| | - Yuanzhen Dong
- Shanghai Duomirui Biotechnology Ltd., Shanghai 201203, China
| | - Wenwen Shangguan
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Weigen Lu
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
9
|
Ruan SD, Dong YZ, Lu JG, Zhao MJ, Lu WG, Feng J. Synthesis of a Novel PTH1–34 Analog with Increased Human Serum Albumin Affinity. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1731299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Parathyroid hormone (PTH)1–34 is an effective peptide drug for osteoporosis therapy. However, the half-life of PTH1–34 in vivo is short, leading to the need for frequent injections of this drug during its treatment. To prolong the half-life of PTH1–34, a novel PTH1–34 analog was generated based on fatty acid generation, and its synthesis process included recombinant protein expression, side-chain modification, and peptide decoration. The PTH1–34 variant was expressed in Escherichia coli, with a single Lys (position 27) retained as a modification site. The side chain, –AEEA-γGlu-C18 diacid, was synthesized using 2-chlorotrityl chloride resin as a solid support, and then was conjugated to the PTH1-34 variant to form PTH-Lys27-AGC. Reversed-phase chromatography confirmed a high final purity (>98%) of the target compound; in vitro bioactivity tests showed that PTH-1 receptor potency of PTH-Lys27-AGC was comparable to that of the native PTH1–34. A competitive human serum albumin binding test demonstrated a high albumin affinity of PTH-Lys27-AGC in comparison to PTH1–34. In summary, we developed a novel PTH1–34 analog, PTH-Lys27-AGC, which may be a long-acting agent for osteoporosis treatment in the future.
Collapse
Affiliation(s)
- Si-Da Ruan
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Yuan-Zhen Dong
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jian-Guang Lu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Meng-Jia Zhao
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Wei-Gen Lu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Cho J, Park J, Kim S, Kim JC, Tae G, Jin MS, Kwon I. Intramolecular distance in the conjugate of urate oxidase and fatty acid governs FcRn binding and serum half-life in vivo. J Control Release 2020; 321:49-58. [DOI: 10.1016/j.jconrel.2020.01.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/17/2022]
|
11
|
The Minimal Effect of Linker Length for Fatty Acid Conjugation to a Small Protein on the Serum Half-Life Extension. Biomedicines 2020; 8:biomedicines8050096. [PMID: 32357510 PMCID: PMC7277390 DOI: 10.3390/biomedicines8050096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 01/20/2023] Open
Abstract
Conjugation of serum albumin or one of its ligands (such as fatty acid) has been an effective strategy to prolong the serum half-lives of drugs via neonatal Fc receptor (FcRn)–mediated recycling of albumin. So far, fatty acid (FA) has been effective in prolonging the serum half-lives for therapeutic peptides and small proteins, but not for large therapeutic proteins. Very recently, it was reported a large protein conjugated to FA competes with the binding of FcRn with serum albumin, leading to limited serum half-life extension, because primary FA binding sites in serum albumin partially overlap with FcRn binding sites. In order to prevent such competition, longer linkers between FA and the large proteins were required. Herein, we hypothesized that small proteins do not cause substantial competition for FcRn binding to albumin, resulting in the extended serum half-life. Using a small protein (28 kDa), we investigated whether the intramolecular distance in FA-protein conjugate affects the FcRn binding with albumin and serum half-life using linkers with varying lengths. Unlike with the FA-conjugated large protein, all FA-conjugated small proteins with different linkers exhibited comparable the FcRn binding to albumin and extended serum half-life.
Collapse
|
12
|
Petersen M, Gandhi PS, Buchardt J, Alanentalo T, Fels JJ, Johansen NL, Helding-Kvist P, Vad K, Thygesen P. Tissue Distribution and Receptor Activation by Somapacitan, a Long Acting Growth Hormone Derivative. Int J Mol Sci 2020; 21:ijms21041181. [PMID: 32053994 PMCID: PMC7072805 DOI: 10.3390/ijms21041181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 11/16/2022] Open
Abstract
Somapacitan is a long-acting, once-weekly, albumin-binding growth hormone (GH) derivative. The reversible albumin-binding properties leads to prolonged circulation half-life. Here, we investigated and compared somapacitan with human GH on downstream receptor signaling in primary hepatocytes and hepatocellular models and using isothermal titration calorimetry to characterize receptor binding of somapacitan in the presence or absence of human serum albumin (HSA). With non-invasive fluorescence imaging we quantitatively visualize and compare the temporal distribution and examine the tissue-specific growth hormone receptor (GHR) activation at distribution sites. We found that signaling kinetics were slightly more rapid and intense for GH compared with somapacitan. Receptor binding isotherms were characterized by a high and a low affinity interaction site with or without HSA. Using in vivo optical imaging we found prolonged systemically biodistribution of somapacitan compared with GH, which correlated with plasma pharmacokinetics. Ex vivo mouse organ analysis revealed that the temporal fluorescent intensity in livers dosed with somapacitan was significantly increased compared with GH-dosed livers and correlated with the degree of downstream GHR activation. Finally, we show that fluorescent-labeled analogs distributed to the hypertrophic zone in the epiphysis of proximal tibia of hypophysectomized rats and that somapacitan and GH activate the GHR signaling in epiphyseal tissues.
Collapse
Affiliation(s)
- Maj Petersen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Jens Buchardt
- Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Tomas Alanentalo
- Umeå Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | | | | | | | - Knud Vad
- Global Development, Novo Nordisk A/S, 2860 Søborg, Denmark
| | - Peter Thygesen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
- Correspondence: ; Tel.: +45-3075-4617
| |
Collapse
|
13
|
Sikder S, Gote V, Alshamrani M, Sicotte J, Pal D. Long-term delivery of protein and peptide therapeutics for cancer therapies. Expert Opin Drug Deliv 2019; 16:1113-1131. [DOI: 10.1080/17425247.2019.1662785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Sadia Sikder
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Vrinda Gote
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Meshal Alshamrani
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Jeff Sicotte
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Dhananjay Pal
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| |
Collapse
|
14
|
Holt RIG, Ho KKY. The Use and Abuse of Growth Hormone in Sports. Endocr Rev 2019; 40:1163-1185. [PMID: 31180479 DOI: 10.1210/er.2018-00265] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/27/2019] [Indexed: 12/18/2022]
Abstract
GH is banned by the World Anti-Doping Agency as a performance-enhancing anabolic agent. Doping with GH likely began in the early 1980s and became more prevalent with the advent of recombinant technology well before any scientific evidence of benefit. The expectation that GH improves physical function stems from its anabolic and lipolytic properties. Athletic performance depends on muscle strength and the energy required to power muscle function. In recreational athletes, GH selectively improves anaerobic sprint capacity but has not been proven to significantly enhance muscle strength, power, or maximum rate of oxygen consumption. GH is secreted as a family of isoform peptides in a pulsatile manner reflecting intermittent secretion and rapid clearance. Its anabolic actions are largely mediated by IGF-I, which stimulates whole-body protein synthesis, including skeletal muscle and collagen proteins. Two methods have been validated for detecting GH abuse in athletes. The first (the isoform method) is based on distinguishing pure recombinant 22-kDa GH from the heterogeneous isoforms secreted from the pituitary. The second (the marker method) is based on measuring blood levels of GH-responsive proteins, specifically IGF-I and the N-terminal propeptide of type III collagen (P-III-NP). Only a handful of athletes have been caught since the implementation of GH doping tests in 2004. The low rate likely reflects the limitation of in-competition testing using current methods. Improved detection rates may be achieved by more out-of-competition testing, introducing athletes' biological passports, and the development of novel methods. Governance, operational, technical, and political factors influence the effectiveness of an anti-doping program.
Collapse
Affiliation(s)
- Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ken K Y Ho
- Garvan Institute of Medical Research, St. Vincent's Hospital, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Iyengar ARS, Gupta S, Jawalekar S, Pande AH. Protein Chimerization: A New Frontier for Engineering Protein Therapeutics with Improved Pharmacokinetics. J Pharmacol Exp Ther 2019; 370:703-714. [DOI: 10.1124/jpet.119.257063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
|
16
|
Menacho-Melgar R, Decker JS, Hennigan JN, Lynch MD. A review of lipidation in the development of advanced protein and peptide therapeutics. J Control Release 2018; 295:1-12. [PMID: 30579981 DOI: 10.1016/j.jconrel.2018.12.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 12/22/2022]
Abstract
The use of biologics (peptide and protein based drugs) has increased significantly over the past few decades. However, their development has been limited by their short half-life, immunogenicity and low membrane permeability, restricting most therapies to extracellular targets and administration by injection. Lipidation is a clinically-proven post-translational modification that has shown great promise to address these issues: improving half-life, reducing immunogenicity and enabling intracellular uptake and delivery across epithelia. Despite its great potential, lipidation remains an underutilized strategy in the clinical translation of lead biologics. We review how lipidation can overcome common challenges in biologics development as well as highlight gaps in our understanding of the effect of lipidation on therapeutic efficacy, where increased research and development efforts may lead to next-generation drugs.
Collapse
Affiliation(s)
| | - John S Decker
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Michael D Lynch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
17
|
Frich CK, Krüger F, Walther R, Domar C, Andersen AHF, Tvilum A, Dagnæs-Hansen F, Denton PW, Tolstrup M, Paludan SR, Münch J, Zelikin AN. Non-covalent hitchhiking on endogenous carriers as a protraction mechanism for antiviral macromolecular prodrugs. J Control Release 2018; 294:298-310. [PMID: 30552954 DOI: 10.1016/j.jconrel.2018.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/26/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022]
Abstract
Albumin is a highly successful tool of drug delivery providing drastically extended body and blood residence time for the associated cargo, but it only traffics single drug copies at a time. In turn, macromolecular prodrugs (MP) are advantaged in carrying a high drug payload but offering only a modest extension of residence time to the conjugated drugs. In this work, we engineer MP to contain terminal groups that bind to albumin via non-covalent association and reveal that this facile measure affords a significant protraction for the associated polymers. This methodology is applied to MP of acyclovir, a successful drug against herpes simplex virus infection but with poor pharmacokinetics. Resulting albumin-affine MP were efficacious agents against herpes simplex virus type 2 (HSV-2) both in vitro and in vivo. In the latter case, sub-cutaneous administration of MP resulted in local (vaginal) antiviral effects and a systemic protection. Presented benefits of non-covalent association with albumin are readily transferrable to a wide variety of MP in development for drug delivery as anticancer, anti-inflammatory, and anti-viral measures.
Collapse
Affiliation(s)
| | - Franziska Krüger
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Raoul Walther
- Department of Chemistry, Aarhus University, 8000 Aarhus C, Denmark
| | - Cecilie Domar
- Department of Chemistry, Aarhus University, 8000 Aarhus C, Denmark
| | - Anna H F Andersen
- Department of Infectious Diseases, Aarhus University Hospital, 8000 Aarhus C, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus N, Denmark
| | - Anne Tvilum
- Department of Chemistry, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Paul W Denton
- Department of Infectious Diseases, Aarhus University Hospital, 8000 Aarhus C, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus N, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, 8000 Aarhus C, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus N, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany.
| | | |
Collapse
|