1
|
Jun T, Shin SH, Won YY. Engineered polymeric excipients for enhancing the stability of protein biologics: Poly(N-isopropylacrylamide)-poly(ethylene glycol) (PNIPAM-PEG) block copolymers. Int J Pharm 2024; 664:124636. [PMID: 39197798 DOI: 10.1016/j.ijpharm.2024.124636] [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: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Protein therapeutics, particularly antibodies, depend on maintaining their native structures for optimal function. Hydrophobic interfaces, such as the air-water interface, can trigger protein aggregation and denaturation. While completely avoiding such interfacial exposures during manufacturing and storage is impractical, minimizing them is crucial for enhancing protein drug stability and extending shelf life. In the biologics industry, surfactants like polysorbates are commonly used as additives (excipients) to mitigate these undesirable interfacial exposures. However, polysorbates, the most prevalent choice, have recognized limitations in terms of polydispersity, purity, and stability, prompting the exploration of alternative excipients. The present study identifies poly(N-isopropylacrylamide)-poly(ethylene glycol) (PNIPAM-PEG) block copolymers as a promising alternative to polysorbates. Due to its stronger affinity for the air-water interface, PNIPAM-PEG significantly outperforms polysorbates in enhancing protein stability. This claim is supported by results from multiple tests. Accelerated dynamic light scattering (DLS) experiments demonstrate PNIPAM-PEG's exceptional efficacy in preserving IgG stability against surface-induced aggregation, surpassing conventional polysorbate excipients (Tween 80 and Tween 20) under high-temperature conditions. Additionally, circular dichroism (CD) spectroscopy results reveal conformational alterations associated with aggregation, with PNIPAM-PEG consistently demonstrates a greater protective effect by mitigating negative shifts at λ ≅ 220 nm, indicative of changes in secondary structure. Overall, this study positions PNIPAM-PEG as a promising excipient for antibody therapeutics, facilitating the development of more stable and effective biopharmaceuticals.
Collapse
Affiliation(s)
- Taesuk Jun
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Sung-Ho Shin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
2
|
Wang Y, Williams HD, Dikicioglu D, Dalby PA. Predictive Model Building for Aggregation Kinetics Based on Molecular Dynamics Simulations of an Antibody Fragment. Mol Pharm 2024. [PMID: 39348223 DOI: 10.1021/acs.molpharmaceut.4c00859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Computational methods including machine learning and molecular dynamics simulations have strong potential to characterize, understand, and ultimately predict the properties of proteins relevant to their stability and function as therapeutics. Such methods would streamline the development pathway by minimizing the current experimental testing required for many protein variants and formulations. The molecular understanding of thermostability and aggregation propensity has advanced significantly along with predictive algorithms based on the sequence-level or structural-level information on a protein. However, these approaches focus largely on a comparison of protein sequence variations to correlate the properties of proteins to their stability, solubility, and aggregation propensity. For therapeutic protein development, it is of equal importance to take into account the impact of the formulation conditions to elucidate and predict the stability of the antibody drugs. At the macroscopic level, changing temperature, pH, ionic strength, and the addition of excipients can significantly alter the kinetics of protein aggregation. The mechanisms controlling aggregation kinetics have been traced back to a combination of molecular features, including conformational stability, partial unfolding to aggregation-prone states, and the colloidal stability governed by surface charges and hydrophobicity. However, very little has been done to evaluate these features in the context of protein dynamics in different formulations. In this work, we have combined a range of molecular features calculated from the Fab A33 protein sequence and molecular dynamics simulations. Using the power of advanced, yet interpretable, statistical tools, it has been possible to uncover greater insights into the mechanisms behind protein stability, validating previous findings, and also develop models that can predict the aggregation kinetics within a range of 49 different solution conditions.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Biochemical Engineering, University College London, London WC1E 6BT, U.K
| | - Hywel D Williams
- Biopharmaceutical Product Development, CSL Ltd., 45 Poplar Road, Parkville 3052, Australia
| | - Duygu Dikicioglu
- Department of Biochemical Engineering, University College London, London WC1E 6BT, U.K
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, London WC1E 6BT, U.K
| |
Collapse
|
3
|
Ramaraju H, Garcia-Gomez E, McAtee AM, Verga AS, Hollister SJ. Shape memory cycle conditions impact human bone marrow stromal cell binding to RGD- and YIGSR-conjugated poly (glycerol dodecanedioate). Acta Biomater 2024; 186:246-259. [PMID: 39111679 DOI: 10.1016/j.actbio.2024.07.057] [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: 05/12/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
Bioresorbable shape memory polymers (SMP) are an emerging class of polymers that can help address several challenges associated with minimally invasive surgery by providing a solution for structural tissue repair. Like most synthetic polymer networks, SMPs require additional biorelevance and modification for biomedical applications. Methodologies used to incorporate bioactive ligands must preserve SMP thermomechanics and ensure biofunctionality following in vivo delivery. We have previously described the development of a novel thermoresponsive bioresorbable SMP, poly (glycerol dodecanedioate) (PGD). In this study, cell-adhesive peptide sequences RGD and YIGSR were conjugated with PGD. We investigated 1) the impact of conjugated peptides on the fixity (Rf), recovery (Rr), and recovery rate (dRr/dT), 2) the impact of conjugated peptides on cell binding, and 3) the impact of the shape memory cycle (Tprog) on conjugated peptide functionality towards binding human bone marrow stromal cells (BMSC). Peptide conjugation conditions impact fixity but not the recovery or recovery rate (p < 0.01). Peptide-conjugated substrates increased cell attachment and proliferation compared with controls (p < 0.001). Using complementary integrin binding cell-adhesive peptides increased proliferation compared with using single peptides (p < 0.05). Peptides bound to PGD substrates exhibited specificity to their respective integrin targets. Following the shape memory cycle, peptides maintained functionality and specificity depending on the shape memory cycle conditions (p < 0.001). The dissipation of strain energy during recovery can drive differential arrangement of conjugated sequences impacting functionality, an important design consideration for functionalized SMPs. STATEMENT OF SIGNIFICANCE: Shape memory elastomers are an emerging class of polymers that are well-suited for minimally invasive repair of soft tissues. Tissue engineering approaches commonly utilize biodegradable scaffolds to deliver instructive cues, including cells and bioactive signals. Delivering these instructive cues on biodegradable shape memory elastomers requires modification with bioactive ligands. Furthermore, it is necessary to ensure the specificity of the ligands to their biological targets when conjugated to the polymer. Moreover, the bioactive ligand functionality must be conserved after completing the shape memory cycle, for applications in tissue engineering.
Collapse
Affiliation(s)
- Harsha Ramaraju
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States.
| | - Elisa Garcia-Gomez
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annabel M McAtee
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Adam S Verga
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Scott J Hollister
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
4
|
Arakawa T, Tomioka Y, Akuta T, Shiraki K. The contrasting roles of co-solvents in protein formulations and food products. Biophys Chem 2024; 312:107282. [PMID: 38944944 DOI: 10.1016/j.bpc.2024.107282] [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: 03/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024]
Abstract
Protein aggregation is a major hurdle in developing biopharmaceuticals, in particular protein formulation area, but plays a pivotal role in food products. Co-solvents are used to suppress protein aggregation in pharmaceutical proteins. On the contrary, aggregation is encouraged in the process of food product making. Thus, it is expected that co-solvents play a contrasting role in biopharmaceutical formulation and food products. Here, we show several examples that utilize co-solvents, e.g., salting-out salts, sugars, polyols and divalent cations in promoting protein-protein interactions. The mechanisms of co-solvent effects on protein aggregation and solubility have been studied on aqueous protein solution and applied to develop pharmaceutical formulation based on the acquired scientific knowledge. On the contrary, co-solvents have been used in food industries based on empirical basis. Here, we will review the mechanisms of co-solvent effects on protein-protein interactions that can be applied to both pharmaceutical and food industries and hope to convey knowledge acquired through research on co-solvent interactions in aqueous protein solution and formulation to those involved in food science and provide those involved in protein solution research with the observations on aggregation behavior of food proteins.
Collapse
Affiliation(s)
- Tsutomu Arakawa
- Alliance Protein Laboratories, 13380 Pantera Road, San Diego, CA 92130, USA.
| | - Yui Tomioka
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd, 3333-26, Aza-Asayama, Kamitezuna Tahahagi, Ibaraki 318-0004, Japan
| | - Teruo Akuta
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd, 3333-26, Aza-Asayama, Kamitezuna Tahahagi, Ibaraki 318-0004, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8573, Japan
| |
Collapse
|
5
|
Groël S, Roncin H, Härdter N, Winter G. Homogeneous Heat Transfer During Freeze-Drying Using Cyclic Olefin Polymer Vials. J Pharm Sci 2024; 113:2947-2951. [PMID: 38906251 DOI: 10.1016/j.xphs.2024.06.015] [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: 03/15/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
In pharmaceutical freeze-drying processes, batch homogeneity is an important quality attribute. In this context, the edge-vial-effect is a challenging phenomenon. Shortly, this effect describes that vials at the edges of the shelf dry faster and at a higher temperature compared to vials in the middle of the shelf. Studies by Ehlers et al. revealed that this effect mainly origins from the number of neighbor vials cooling each other, which is reduced for vials in corners and edges compared to vials in the middle. Due to the reduced heat transfer in cyclic olefin polymer (COP) vials, the adverse edge-vial-effect should be greatly reduced allowing a better batch uniformity. In this focused study, glass and COP vials are compared regarding this effect on a fully loaded shelf. A reference experiment with vials placed at distance using a specially designed frame is presented as well.
Collapse
Affiliation(s)
- Sebastian Groël
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| | - Hugo Roncin
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nicole Härdter
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerhard Winter
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
6
|
Velankar KY, Gawalt ES, Wen Y, Meng WS. Pharmaceutical proteins at the interfaces and the role of albumin. Biotechnol Prog 2024; 40:e3474. [PMID: 38647437 DOI: 10.1002/btpr.3474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/15/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
A critical measure of the quality of pharmaceutical proteins is the preservation of native conformations of the active pharmaceutical ingredients. Denaturation of the active proteins in any step before administration into patients could lead to loss of potency and/or aggregation, which is associated with an increased risk of immunogenicity of the products. Interfacial stress enhances protein instability as their adsorption to the air-liquid and liquid-solid interfaces are implicated in the formation of denatured proteins and aggregates. While excipients in protein formulations have been employed to reduce the risk of aggregation, the roles of albumin as a stabilizer have not been reviewed from practical and theoretical standpoints. The amphiphilic nature of albumin makes it accumulate at the interfaces. In this review, we aim to bridge the knowledge gap between interfacial instability and the influence of albumin as a surface-active excipient in the context of reducing the immunogenicity risk of protein formulations.
Collapse
Affiliation(s)
- Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Yu Y, Chen R, Chen X, Wang J, Liu C. Regulating the bioactivity of non-glycosylated recombinant human bone morphogenetic protein-2 to enhance bone regeneration. Bioact Mater 2024; 38:169-180. [PMID: 38711759 PMCID: PMC11070760 DOI: 10.1016/j.bioactmat.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Recombinant human bone morphogenetic protein-2 (rhBMP-2) is the predominant growth factor that effectively induces osteogenic differentiation in orthopedic procedures. However, the bioactivity and stability of rhBMP-2 are intrinsically associated with its sequence, structure, and storage conditions. In this study, we successfully determined the amino acid sequence and protein secondary structure model of non-glycosylated rhBMP-2 expressed by an E. coli expression system through X-ray crystal structure analysis. Furthermore, we observed that acidic storage conditions enhanced the proliferative and osteoinductive activity of rhBMP-2. Although the osteogenic activity of non-glycosylated rhBMP-2 is relatively weaker compared to glycosylated rhBMP-2; however, this discrepancy can be mitigated by incorporating exogenous chaperone molecules. Overall, such information is crucial for rationalizing the design of stabilization methods and enhancing the bioactivity of rhBMP-2, which may also be applicable to other growth factors.
Collapse
Affiliation(s)
- Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Rui Chen
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xinye Chen
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
8
|
Singh H, Lawanprasert A, Utkarsh, Pimcharoen S, Dewan A, Rahoi D, Kirimanjeswara GS, Medina SH. Decoupling Fluorous Protein Coatings Yield Heat-Stable and Intrinsically Sterile Bioformulations. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38893-38904. [PMID: 39013021 PMCID: PMC11299136 DOI: 10.1021/acsami.4c03724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 07/18/2024]
Abstract
Thermal inactivation is a major bottleneck to the scalable production, storage, and transportation of protein-based reagents and therapies. Failures in temperature control both compromise protein bioactivity and increase the risk of microorganismal contamination. Herein, we report the rational design of fluorochemical additives that promiscuously bind to and coat the surfaces of proteins to enable their stable dispersion within fluorous solvents. By replacing traditional aqueous liquids with fluorinated media, this strategy conformationally rigidifies proteins to preserve their structure and function at extreme temperatures (≥90 °C). We show that fluorous protein formulations resist contamination by bacterial, fungal, and viral pathogens, which require aqueous environments for survival, and display equivalent serum bioavailability to standard saline samples in animal models. Importantly, by designing dispersants that decouple from the protein surface in physiologic solutions, we deliver a fluorochemical formulation that does not alter the pharmacologic function or safety profile of the functionalized protein in vivo. As a result, this nonaqueous protein storage paradigm is poised to open technological opportunities in the design of shelf-stable protein reagents and biopharmaceuticals.
Collapse
Affiliation(s)
- Harminder Singh
- Department
of Biomedical Engineering, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| | - Atip Lawanprasert
- Department
of Biomedical Engineering, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| | - Utkarsh
- Department
of Biomedical Engineering, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| | - Sopida Pimcharoen
- Department
of Biomedical Engineering, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| | - Arshiya Dewan
- Department
of Veterinary and Biomedical Sciences, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| | - Dane Rahoi
- Animal
Diagnostics Laboratory, Pennsylvania State
University, University Park, Pennsylvania 16802-4400, United States
| | - Girish S. Kirimanjeswara
- Department
of Veterinary and Biomedical Sciences, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
- Center
for Infectious Disease Dynamics, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
- Center
for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania 16802-4400, United States
| | - Scott H. Medina
- Department
of Biomedical Engineering, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
- Huck
Institutes
of the Life Sciences, Pennsylvania State
University, University Park, Pennsylvania 16802-4400, United States
| |
Collapse
|
9
|
Hribar-Lee B, Lukšič M. Biophysical Principles Emerging from Experiments on Protein-Protein Association and Aggregation. Annu Rev Biophys 2024; 53:1-18. [PMID: 37906740 DOI: 10.1146/annurev-biophys-030722-111729] [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] [Indexed: 11/02/2023]
Abstract
Protein-protein association and aggregation are fundamental processes that play critical roles in various biological phenomena, from cellular signaling to disease progression. Understanding the underlying biophysical principles governing these processes is crucial for elucidating their mechanisms and developing strategies for therapeutic intervention. In this review, we provide an overview of recent experimental studies focused on protein-protein association and aggregation. We explore the key biophysical factors that influence these processes, including protein structure, conformational dynamics, and intermolecular interactions. We discuss the effects of environmental conditions such as temperature, pH and related buffer-specific effects, and ionic strength and related ion-specific effects on protein aggregation. The effects of polymer crowders and sugars are also addressed. We list the techniques used to study aggregation. We analyze emerging trends and challenges in the field, including the development of computational models and the integration of multidisciplinary approaches for a comprehensive understanding of protein-protein association and aggregation.
Collapse
Affiliation(s)
- Barbara Hribar-Lee
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia;
| | - Miha Lukšič
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia;
| |
Collapse
|
10
|
Manning MC, Holcomb RE, Payne RW, Stillahn JM, Connolly BD, Katayama DS, Liu H, Matsuura JE, Murphy BM, Henry CS, Crommelin DJA. Stability of Protein Pharmaceuticals: Recent Advances. Pharm Res 2024; 41:1301-1367. [PMID: 38937372 DOI: 10.1007/s11095-024-03726-x] [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: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024]
Abstract
There have been significant advances in the formulation and stabilization of proteins in the liquid state over the past years since our previous review. Our mechanistic understanding of protein-excipient interactions has increased, allowing one to develop formulations in a more rational fashion. The field has moved towards more complex and challenging formulations, such as high concentration formulations to allow for subcutaneous administration and co-formulation. While much of the published work has focused on mAbs, the principles appear to apply to any therapeutic protein, although mAbs clearly have some distinctive features. In this review, we first discuss chemical degradation reactions. This is followed by a section on physical instability issues. Then, more specific topics are addressed: instability induced by interactions with interfaces, predictive methods for physical stability and interplay between chemical and physical instability. The final parts are devoted to discussions how all the above impacts (co-)formulation strategies, in particular for high protein concentration solutions.'
Collapse
Affiliation(s)
- Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO, USA.
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA.
| | - Ryan E Holcomb
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Robert W Payne
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
11
|
Ge S, Wang X, Zhao X, Yuan L, Bao X, Sun C, Gong Z, Guo J, Yuan S, Hu D, Yang J, Yuan B, Zhang G. Responsive Multi-Arm PEG-Modified COF Nanocomposites: Dynamic Photothermal, pH/ROS Dual-Responsive, Targeted Carriers for Rheumatoid Arthritis Treatment. Adv Healthc Mater 2024:e2401744. [PMID: 38885286 DOI: 10.1002/adhm.202401744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic immune disease characterized by the infiltration of immune cells and the proliferation of fibroblast-like synoviocytes (FLS) at the joint site, leading to inflammation and joint destruction. However, the available treatment options targeting both inflammatory and proliferative FLS are limited. Herein, this work presents three covalent organic frameworks (COFs) photothermal composite systems modified with multi-armed polyethylene glycols (PEG) for the treatment of RA. These systems exhibit a dual response under low pH and high reactive oxygen species (ROS) conditions at the site of inflammation, with a specific focus on delivering the protein drug ribonuclease A (RNase A). Notably, molecular docking studies reveal the interaction between RNase A and NF-κB p65 protein, and Western blotting confirm its inhibitory effect on NF-κB activity. In vitro and in vivo experiments verify the significant reduction in joint swelling and deformities in adjuvant-induced arthritis (AIA) rats after treatment with RNase A delivered by multi-armed PEG-modified COF ligands, restoring joint morphology to normal. These findings underscore the promising therapeutic potential of COFs for the treatment of RA, highlighting their unique capabilities in addressing both inflammatory and proliferative aspects of the disease and expanding the scope of biomedical applications for COFs.
Collapse
Affiliation(s)
- Saisai Ge
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xinyue Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xinru Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lingling Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xuewei Bao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Caidie Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zehua Gong
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jun Guo
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Siyu Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Danyou Hu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Yang
- Experimental Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Bin Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
12
|
Lynch CC, Khirich G, Lee RT. Quantification of Biopharmaceutically Relevant Nonionic Surfactant Excipients Using Benchtop qNMR. Anal Chem 2024; 96:6746-6755. [PMID: 38632675 DOI: 10.1021/acs.analchem.4c00422] [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: 04/19/2024]
Abstract
Nonionic surfactant excipients (NISEs) are commonly added to biologics formulations to mitigate the effects of stress incurred by the active biotherapeutic during manufacturing, transport, and storage. During manufacturing, NISEs are added by dilution of a stock solution directly into a protein formulation, and their accurate addition is critical in maintaining the quality and integrity of the drug product and thus ensuring patient safety. This is especially true for the common NISEs, polysorbates 20 and 80 (PS20 and PS80, respectively) and poloxamer 188 (P188). With the increasing diversity of biologic modalities within modern pharmaceutical pipelines, there is thus a critical need to develop and deploy convenient and user-accessible analytical techniques that can rapidly and reliably quantify these NISEs under biopharmaceutically relevant conditions. We thus pursued 60 MHz benchtop quantitative NMR (qNMR) as a nondestructive and user-friendly analytical technique for the quantification of PS20, PS80, and P188 under such conditions. We demonstrated the ability of benchtop qNMR (1) to quantify simulated PS20, PS80, and P188 stock solutions representative of those used during the drug substance (DS) formulation step in biomanufacturing and (2) to quantify these NISEs at and below their target concentrations (≤0.025% w/v) directly in biologics formulations containing histidine, sucrose, and one of three biotherapeutic modalities (monoclonal antibody, antibody-drug conjugate, and Fc-fusion protein). Our results demonstrate that benchtop qNMR offers a fit-for-purpose, reliable, user-friendly, and green analytical route by which NISE of interest to the biopharmaceutical industry may be readily and reliably quantified. We conclude that benchtop qNMR has the potential to be applied to other excipient formulation components in the presence of various biological modalities as well as the potential for routine integration within analytical and QC laboratories across pharmaceutical development and manufacturing sites.
Collapse
Affiliation(s)
- Ciarán C Lynch
- Analytical Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Gennady Khirich
- Analytical Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Ryan T Lee
- Analytical Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
13
|
T T Nguyen K, Zillen D, Lasorsa A, van der Wel PCA, Frijlink HW, L J Hinrichs W. Combinations of arginine and pullulan reveal the selective effect of stabilization mechanisms on different lyophilized proteins. Int J Pharm 2024; 654:123938. [PMID: 38408554 DOI: 10.1016/j.ijpharm.2024.123938] [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: 01/11/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
The stability of lactate dehydrogenase (LDH) and β-galactosidase (β-gal), incorporated in arginine/pullulan (A/P) mixtures at various weight ratios by lyophilization, was determined. The physicochemical characteristics of various A/P mixtures were assessed. With decreasing A/P ratios, the glass transition temperature of the formulations increased. Furthermore, arginine crystallization due to high relative humidity (RH) exposure was prevented at an A/P weight ratio of 4/6 or less. When stored at 0 % RH / 60 °C for 4 weeks, arginine was superior to pullulan as stabilizer. During storage at 43 % RH / 30 ℃ for 4 weeks, the enzymatic activity of LDH was best retained at an A/P weight ratio of 2/8, while β-gal activity was relatively well-retained at A/P weight ratios of both 8/2 and 2/8. LDH seemed to be more prone to degradation in the rubbery state. In the glassy state, β-gal degraded faster than LDH. Solid-state nuclear magnetic resonance spectroscopy showed that (labeled) arginine experienced a different interaction in the two protein samples, reflecting a modulation of long-range correlations of the arginine side chain nitrogen atoms (Nε, Nη). In summary, LDH stabilization in the A/P matrix requires vitrification. Further stabilization difference between LDH and β-gal may be dependent on the interaction with arginine.
Collapse
Affiliation(s)
- Khanh T T Nguyen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Daan Zillen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Alessia Lasorsa
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, the Netherlands
| | - Patrick C A van der Wel
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, the Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
14
|
Kapre S, Palakurthi SS, Jain A, Palakurthi S. DES-igning the future of drug delivery: A journey from fundamentals to drug delivery applications. J Mol Liq 2024; 400:124517. [DOI: 10.1016/j.molliq.2024.124517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
|
15
|
Bharmoria P, Tietze AA, Mondal D, Kang TS, Kumar A, Freire MG. Do Ionic Liquids Exhibit the Required Characteristics to Dissolve, Extract, Stabilize, and Purify Proteins? Past-Present-Future Assessment. Chem Rev 2024; 124:3037-3084. [PMID: 38437627 PMCID: PMC10979405 DOI: 10.1021/acs.chemrev.3c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
Proteins are highly labile molecules, thus requiring the presence of appropriate solvents and excipients in their liquid milieu to keep their stability and biological activity. In this field, ionic liquids (ILs) have gained momentum in the past years, with a relevant number of works reporting their successful use to dissolve, stabilize, extract, and purify proteins. Different approaches in protein-IL systems have been reported, namely, proteins dissolved in (i) neat ILs, (ii) ILs as co-solvents, (iii) ILs as adjuvants, (iv) ILs as surfactants, (v) ILs as phase-forming components of aqueous biphasic systems, and (vi) IL-polymer-protein/peptide conjugates. Herein, we critically analyze the works published to date and provide a comprehensive understanding of the IL-protein interactions affecting the stability, conformational alteration, unfolding, misfolding, and refolding of proteins while providing directions for future studies in view of imminent applications. Overall, it has been found that the stability or purification of proteins by ILs is bispecific and depends on the structure of both the IL and the protein. The most promising IL-protein systems are identified, which is valuable when foreseeing market applications of ILs, e.g., in "protein packaging" and "detergent applications". Future directions and other possibilities of IL-protein systems in light-harvesting and biotechnology/biomedical applications are discussed.
Collapse
Affiliation(s)
- Pankaj Bharmoria
- CICECO
- Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
- Department
of Smart Molecular, Inorganic and Hybrid Materials, Institute of Materials Science of Barcelona (ICMAB-CSIC), 08193 Bellaterra, Barcelona, Spain
- Department
of Chemistry and Molecular Biology, Wallenberg Centre for Molecular
and Translational Medicine, University of
Gothenburg, SE-412 96 Göteborg, Sweden
| | - Alesia A. Tietze
- Department
of Chemistry and Molecular Biology, Wallenberg Centre for Molecular
and Translational Medicine, University of
Gothenburg, SE-412 96 Göteborg, Sweden
| | - Dibyendu Mondal
- CICECO
- Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
- Institute
of Plant Genetics (IPG), Polish Academy of Sciences, Strzeszyńska 34, 60-479 Poznań, Poland
- Centre
for Nano and Material Sciences, JAIN (Deemed-to-be
University), Jain Global
Campus, Bangalore 562112, India
| | - Tejwant Singh Kang
- Department
of Chemistry, UGC Center for Advance Studies-II,
Guru Nanak Dev University (GNDU), Amritsar 143005, Punjab, India
| | - Arvind Kumar
- Salt
and Marine Chemicals Division, CSIR-Central
Salt and Marine Chemicals Research Institute, G. B. Marg, Bhavnagar 364002, Gujarat, India
| | - Mara G Freire
- CICECO
- Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
16
|
Saurabh S, Zhang Q, Seddon JM, Lu JR, Kalonia C, Bresme F. Unraveling the Microscopic Mechanism of Molecular Ion Interaction with Monoclonal Antibodies: Impact on Protein Aggregation. Mol Pharm 2024; 21:1285-1299. [PMID: 38345400 PMCID: PMC10915798 DOI: 10.1021/acs.molpharmaceut.3c00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
Understanding and predicting protein aggregation represents one of the major challenges in accelerating the pharmaceutical development of protein therapeutics. In addition to maintaining the solution pH, buffers influence both monoclonal antibody (mAb) aggregation in solution and the aggregation mechanisms since the latter depend on the protein charge. Molecular-level insight is necessary to understand the relationship between the buffer-mAb interaction and mAb aggregation. Here, we use all-atom molecular dynamics simulations to investigate the interaction of phosphate (Phos) and citrate (Cit) buffer ions with the Fab and Fc domains of mAb COE3. We demonstrate that Phos and Cit ions feature binding mechanisms, with the protein that are very different from those reported previously for histidine (His). These differences are reflected in distinctive ion-protein binding modes and adsorption/desorption kinetics of the buffer molecules from the mAb surface and result in dissimilar effects of these buffer species on mAb aggregation. While His shows significant affinity toward hydrophobic amino acids on the protein surface, Phos and Cit ions preferentially bind to charged amino acids. We also show that Phos and Cit anions provide bridging contacts between basic amino acids in neighboring proteins. The implications of such contacts and their connection to mAb aggregation in therapeutic formulations are discussed.
Collapse
Affiliation(s)
- Suman Saurabh
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College, London W12 0BZ, U.K.
| | - Qinkun Zhang
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College, London W12 0BZ, U.K.
| | - John M. Seddon
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College, London W12 0BZ, U.K.
| | - Jian R. Lu
- Biological
Physics Group, School of Physics and Astronomy, Faculty of Science
and Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Cavan Kalonia
- Dosage
Form Design and Development, BioPharmaceutical Development, BioPharmaceuticals
R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Fernando Bresme
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College, London W12 0BZ, U.K.
| |
Collapse
|
17
|
Mehrotra S, Kalyan BG P, Nayak PG, Joseph A, Manikkath J. Recent Progress in the Oral Delivery of Therapeutic Peptides and Proteins: Overview of Pharmaceutical Strategies to Overcome Absorption Hurdles. Adv Pharm Bull 2024; 14:11-33. [PMID: 38585454 PMCID: PMC10997937 DOI: 10.34172/apb.2024.009] [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: 11/18/2022] [Revised: 07/04/2023] [Accepted: 08/16/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Proteins and peptides have secured a place as excellent therapeutic moieties on account of their high selectivity and efficacy. However due to oral absorption limitations, current formulations are mostly delivered parenterally. Oral delivery of peptides and proteins (PPs) can be considered the need of the hour due to the immense benefits of this route. This review aims to critically examine and summarize the innovations and mechanisms involved in oral delivery of peptide and protein drugs. Methods Comprehensive literature search was undertaken, spanning the early development to the current state of the art, using online search tools (PubMed, Google Scholar, ScienceDirect and Scopus). Results Research in oral delivery of proteins and peptides has a rich history and the development of biologics has encouraged additional research effort in recent decades. Enzyme hydrolysis and inadequate permeation into intestinal mucosa are the major causes that result in limited oral absorption of biologics. Pharmaceutical and technological strategies including use of absorption enhancers, enzyme inhibition, chemical modification (PEGylation, pro-drug approach, peptidomimetics, glycosylation), particulate delivery (polymeric nanoparticles, liposomes, micelles, microspheres), site-specific delivery in the gastrointestinal tract (GIT), membrane transporters, novel approaches (self-nanoemulsifying drug delivery systems, Eligen technology, Peptelligence, self-assembling bubble carrier approach, luminal unfolding microneedle injector, microneedles) and lymphatic targeting, are discussed. Limitations of these strategies and possible innovations for improving oral bioavailability of protein and peptide drugs are discussed. Conclusion This review underlines the application of oral route for peptide and protein delivery, which can direct the formulation scientist for better exploitation of this route.
Collapse
Affiliation(s)
- Sonal Mehrotra
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pavan Kalyan BG
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pawan Ganesh Nayak
- Department of Pharmacology,Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | | | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
18
|
Mehrdadi S. Lipid-Based Nanoparticles as Oral Drug Delivery Systems: Overcoming Poor Gastrointestinal Absorption and Enhancing Bioavailability of Peptide and Protein Therapeutics. Adv Pharm Bull 2024; 14:48-66. [PMID: 38585451 PMCID: PMC10997935 DOI: 10.34172/apb.2024.016] [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: 11/07/2022] [Revised: 08/09/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
Delivery and formulation of oral peptide and protein therapeutics have always been a challenge for the pharmaceutical industry. The oral bioavailability of peptide and protein therapeutics mainly relies on their gastrointestinal solubility and permeability which are affected by their poor membrane penetration, high molecular weight and proteolytic (chemical and enzymatic) degradation resulting in limited delivery and therapeutic efficacy. The present review article highlights the challenges and limitations of oral delivery of peptide and protein therapeutics focusing on the application, potential and importance of solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) as lipid-based drug delivery systems (LBDDSs) and their advantages and drawbacks. LBDDSs, due to their lipid-based matrix can encapsulate both lipophilic and hydrophilic drugs, and by reducing the first-pass effect and avoiding proteolytic degradation offer improved drug stability, dissolution rate, absorption, bioavailability and controlled drug release. Furthermore, their small size, high surface area and surface modification increase their mucosal adhesion, tissue-targeted distribution, physiological function and half-life. Properties such as simple preparation, high-scale manufacturing, biodegradability, biocompatibility, prolonged half-life, lower toxicity, lower adverse effects, lipid-based structure, higher drug encapsulation rate and various drug release profile compared to other similar carrier systems makes LBDDSs a promising drug delivery system (DDS). Nevertheless, undesired physicochemical features of peptide and protein drug development and discovery such as plasma stability, membrane permeability and circulation half-life remain a serious challenge which should be addressed in future.
Collapse
Affiliation(s)
- Soheil Mehrdadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| |
Collapse
|
19
|
Floyd JA, Gillespie AJ, Nightlinger NS, Siska C, Kerwin BA. The Development of a Novel Aflibercept Formulation for Ocular Delivery. J Pharm Sci 2024; 113:366-376. [PMID: 38042344 DOI: 10.1016/j.xphs.2023.11.022] [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: 07/18/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Aflibercept is a recombinant fusion protein that is commercially available for several ocular diseases impacting millions of people worldwide. Here, we use a case study approach to examine alternative liquid formulations for aflibercept for ocular delivery, utilizing different stabilizers, buffering agents, and surfactants with the goal of improving the thermostability to allow for limited storage outside the cold chain. The formulations were developed by studying the effects of pH changes, substituting amino acids for sucrose and salt, and using polysorbate 80 or poloxamer 188 instead of polysorbate 20. A formulation containing acetate, proline, and poloxamer 188 had lower rates of aggregate formation at 4, 30, and 40°C when compared to the marketed commercial formulation containing phosphate, sucrose, sodium chloride, and polysorbate 20. Further studies examining subvisible particles after exposure to a transport stress and long-term stability at 4°C, post-translational modifications by multi-attribute method, purity by reduced and non-reduced capillary electrophoresis, and potency by cell proliferation also demonstrated a comparable or improved stability for the enhanced formulation of acetate, proline, and poloxamer 188. This enhanced stability could enable limited storage outside of the cold chain, allowing for easier distribution in low to middle income countries.
Collapse
Affiliation(s)
- J Alaina Floyd
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA.
| | | | | | - Christine Siska
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA
| | - Bruce A Kerwin
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA.
| |
Collapse
|
20
|
Gupta MN, Uversky VN. Biological importance of arginine: A comprehensive review of the roles in structure, disorder, and functionality of peptides and proteins. Int J Biol Macromol 2024; 257:128646. [PMID: 38061507 DOI: 10.1016/j.ijbiomac.2023.128646] [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: 11/07/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024]
Abstract
Arginine shows Jekyll and Hyde behavior in several respects. It participates in protein folding via ionic and H-bonds and cation-pi interactions; the charge and hydrophobicity of its side chain make it a disorder-promoting amino acid. Its methylation in histones; RNA binding proteins; chaperones regulates several cellular processes. The arginine-centric modifications are important in oncogenesis and as biomarkers in several cardiovascular diseases. The cross-links involving arginine in collagen and cornea are involved in pathogenesis of tissues but have also been useful in tissue engineering and wound-dressing materials. Arginine is a part of active site of several enzymes such as GTPases, peroxidases, and sulfotransferases. Its metabolic importance is obvious as it is involved in production of urea, NO, ornithine and citrulline. It can form unusual functional structures such as molecular tweezers in vitro and sprockets which engage DNA chains as part of histones in vivo. It has been used in design of cell-penetrating peptides as drugs. Arginine has been used as an excipient in both solid and injectable drug formulations; its role in suppressing opalescence due to liquid-liquid phase separation is particularly very promising. It has been known as a suppressor of protein aggregation during protein refolding. It has proved its usefulness in protein bioseparation processes like ion-exchange, hydrophobic and affinity chromatographies. Arginine is an amino acid, whose importance in biological sciences and biotechnology continues to grow in diverse ways.
Collapse
Affiliation(s)
- Munishwar Nath Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
21
|
Gupta T, Seshadri S. Highly purified charge variants of a proposed biosimilar to Omalizumab: impact on in vitro potency and stability under thermal stress. Bioprocess Biosyst Eng 2024; 47:57-64. [PMID: 38156991 DOI: 10.1007/s00449-023-02944-8] [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: 08/22/2023] [Accepted: 11/08/2023] [Indexed: 01/03/2024]
Abstract
Biosimilars are highly complex and similar biological drugs are developed with different manufacturing processes which are not similar to originator manufacturing process. Due to this, biosimilar products inherently have quality differences in comparison to innovator molecule which may be related to size, charge and glycosylation. Despite these differences they are supposed to demonstrate similar behaviour in safety and efficacy profile to the reference product and these differences should not be clinically meaningful. Charge variants are one of the critical quality attributes and sources of heterogeneity. In this study, highly purified charge variants cluster (acidic, main peak and basic) of biosimilar product of Xolair were assessed for their impact on in vitro potency and stability at different thermal stress conditions (2-8 °C and - 20 °C). The study data indicating purified charge variants (> 90%) have no impact on in vitro potency and are stable at different thermal stress conditions up to a week.
Collapse
Affiliation(s)
- Tarun Gupta
- Institute of Science, Nirma University, Sarkhej Gandhinagar Highway, Ahmedabad, Gujarat, India
- Downstream Process Development, Kashiv BioSciences Pvt Ltd, Ahmedabad, Gujarat, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, Sarkhej Gandhinagar Highway, Ahmedabad, Gujarat, India.
| |
Collapse
|
22
|
Vieillard V, Le Guyader G, Jallades A, Astier A. Extended physicochemical stability of cetuximab in opened vials and infusion bags when stored at 4°C and 25°C. J Oncol Pharm Pract 2024; 30:142-150. [PMID: 37078110 PMCID: PMC10804814 DOI: 10.1177/10781552231170583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION This study aimed to determine the stability of cetuximab: (1) under "in-use" conditions after dilution to 1 mg/mL in 0.9% sodium chloride in polyolefin bags and (2) as an undiluted solution (5 mg/mL) repackaged in polypropylene bags or kept in the vial after opening. METHODS Ready-to-use 500 mg/100 mL vials of cetuximab solution were diluted to 1 mg/mL in 100 mL bags of 0.9% sodium chloride or repackaged as a 5 mg/mL solution into empty 100 mL bags. Bags and vials were stored at 4°C for 90 days and 25°C for 3 days. A syringe sample of 7 mL was taken from each bag for the initial determinations. The sampled bags were weighed to determine their initial weight and placed under the planned storage conditions. The physicochemical stability of cetuximab was estimated using validated methods. RESULTS No changes in turbidity, no protein loss, and no changes in cetuximab tertiary structure were observed after 30 days of storage or when subjected to a temperature excursion of 3 days at 25°C and when stored at 4°C for up to 90 days, regardless of the concentrations and batches. The colligative parameters did not change under any of the tested conditions. No evidence of microbial growth was found in bags after 90 days of storage at 4°C. CONCLUSION These results support the extended in-use shelf-life of cetuximab vials and bags, which can be cost-effective for healthcare providers.
Collapse
Affiliation(s)
- Victoire Vieillard
- Department of Pharmacy, AP-HP, Hôpitaux Universitaires Henri-Mondor, Créteil, France
| | - Guillaume Le Guyader
- Department of Pharmacy, AP-HP, Hôpitaux Universitaires Henri-Mondor, Créteil, France
| | - Alice Jallades
- Merck Santé S.A.S., Lyon, France, an affiliate of Merck KGaA
| | - Alain Astier
- Department of Pharmacy, AP-HP, Hôpitaux Universitaires Henri-Mondor, Créteil, France
- Academie Nationale de Pharmacie, Paris, France
| |
Collapse
|
23
|
Glücklich N, Carle S, Diederichs T, Buske J, Mäder K, Garidel P. How enzymatic hydrolysis of polysorbate 20 influences colloidal protein stability. Eur J Pharm Sci 2023; 191:106597. [PMID: 37770006 DOI: 10.1016/j.ejps.2023.106597] [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: 05/13/2023] [Revised: 08/26/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Polysorbates (PS) are esters of ethoxylated sorbitol anhydrides of different composition and are widely used surfactants in biologics. PSs are applied to increase protein stability and concomitant shelf-life via shielding against e.g., interfacial stresses. Due to the presence of specific lipolytic host cell protein (HCP) contaminations in the drug substance, PSs can be degraded via enzymatic hydrolysis. Surfactant hydrolysis leads to the formation of degradants, such as free fatty acids that might form fatty acid particles. In addition, PS degradation may reduce surfactant functionality and thus reduce the protection of the active pharmaceutical ingredient (API). Although enzymatic degradation was observed and reported in the last years, less is known about the relationship between certain polysorbate degradation patterns and the increase of mechanical and interfacial stress towards the API. In this study, the impact of specifically hydrolyzed polysorbate 20 (PS20) towards the stabilization of two monoclonal antibodies (mAbs) during accelerated shaking stress conditions was investigated. The results show that a specific enzymatic degradation pattern of PS20 can influence the colloidal stability of biopharmaceutical formulations. Furthermore, the kinetics of the appearance of visual phenomena, opalescence, and particle formation depended on the polysorbate degradation fingerprint as induced via the presence of surrogate enzymes. The current case study shows the importance of focusing on specific polysorbate ester fractions to understand the overall colloidal protein stabilizing effect. The performed study gives first insight into the functional properties of PS and helps to evaluate the impact of PS degradation in the formulation development of biopharmaceuticals in general.
Collapse
Affiliation(s)
- Nils Glücklich
- Institute of Pharmacy, Faculty of Biosciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, Halle (Saale) 06120, Germany
| | - Stefan Carle
- Innovation Unit, PDB, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, Biberach an der Riss 88397, Germany
| | - Tim Diederichs
- Innovation Unit, PDB, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, Biberach an der Riss 88397, Germany
| | - Julia Buske
- Innovation Unit, PDB, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, Biberach an der Riss 88397, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty of Biosciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, Halle (Saale) 06120, Germany
| | - Patrick Garidel
- Innovation Unit, PDB, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, Biberach an der Riss 88397, Germany; Institute of Chemistry, Faculty of Physical and Theoretical Chemistry, Martin-Luther-University Halle-Wittenberg, Von-Danckelmann-Platz 4, Halle (Saale) 06120, Germany.
| |
Collapse
|
24
|
Groël S, Menzen T, Winter G. Possibilities and limitations of α-relaxation data of amorphous freeze-dried cakes to predict long term IgG 1 antibody stability. Int J Pharm 2023; 646:123445. [PMID: 37748632 DOI: 10.1016/j.ijpharm.2023.123445] [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: 06/05/2023] [Revised: 08/30/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
The value of correlating global α-relaxations with long term protein stability after freeze-drying is inconsistently reported. This study aims to clarify whether and to what extend the long term stability of a freeze-dried protein formulation can be predicted with this method. For this purpose, the α-relaxation parameter τβ [h] of freshly prepared freeze-dried products is obtained by isothermal microcalorimetry. The concept is, that molecular movements in the amorphous matrix are strongly reduced in cakes with longer relaxation time and the product should therefore be more resistant against aggregation. To increase τβ in comparison to a conventional freeze-drying cycle, aggressive drying cycles including structural collapse of the product as well as tempering protocols after freeze-drying are applied. The τβ values are correlated with the aggregation rate of a freeze-dried IgG1 monoclonal antibody measured with high performance size exclusion chromatography. The antibody was used in its market formulation and 6 further compositions. A weak correlation between α-relaxation times and IgG1 aggregation was found. A higher mobility level through increased residual moisture helped to improve the correlation.
Collapse
Affiliation(s)
- Sebastian Groël
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| | | | - Gerhard Winter
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
25
|
Kopp KT, Saerens L, Voorspoels J, Van den Mooter G. Solidification and oral delivery of biologics to the colon- A review. Eur J Pharm Sci 2023; 190:106523. [PMID: 37429482 DOI: 10.1016/j.ejps.2023.106523] [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: 02/01/2023] [Revised: 06/16/2023] [Accepted: 07/08/2023] [Indexed: 07/12/2023]
Abstract
The oral delivery of biologics such as therapeutic proteins, peptides and oligonucleotides for the treatment of colon related diseases has been the focus of increasing attention over the last years. However, the major disadvantage of these macromolecules is their degradation propensity in liquid state which can lead to the undesirable and complete loss of function. Therefore, to increase the stability of the biologic and reduce their degradation propensity, formulation techniques such as solidification can be performed to obtain a stable solid dosage form for oral administration. Due to their fragility, stress exerted on the biologic during solidification has to be reduced with the incorporation of stabilizing excipients into the formulation. This review focuses on the state-of-the-art solidification techniques required to obtain a solid dosage form for the oral delivery of biologics to the colon and the use of suitable excipients for adequate stabilization upon solidification. The solidifying processes discussed within this review are spray drying, freeze drying, bead coating and also other techniques such as spray freeze drying, electro spraying, vacuum- and supercritical fluid drying. Further, the colon as site of absorption in both healthy and diseased state is critically reviewed and possible oral delivery systems for biologics are discussed.
Collapse
Affiliation(s)
- Katharina Tatjana Kopp
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium; Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium
| | - Lien Saerens
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Jody Voorspoels
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Guy Van den Mooter
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
26
|
Sattler S, Gollomp S, Curry A. A Narrative Literature Review of the Established Safety of Human Serum Albumin Use as a Stabilizer in Aesthetic Botulinum Toxin Formulations Compared to Alternatives. Toxins (Basel) 2023; 15:619. [PMID: 37888650 PMCID: PMC10610632 DOI: 10.3390/toxins15100619] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Despite more than 80 years of use in a number of conditions, including in critically ill patients, comments have recently arisen regarding the safety and efficacy of human serum albumin (HSA) as a therapeutic product and stabilizer/excipient in botulinum neurotoxins. This review summarizes the literature on the safety of HSA. Beyond decades of safe use, the largest clinical dataset of HSA safety is a large meta-analysis of HSA supplier data, which found only an extremely remote risk of serious adverse events across millions of doses of therapeutic concentrations of HSA. There is a paucity of literature identifying HSA-specific adverse events when used as a stabilizer/excipient; however, studies of HSA-containing botulinum neurotoxins (BoNTs) suggest that adverse events are not related to HSA. Polysorbates, which are synthetically produced and not physiologically inert, are contained in pending or new-to-market BoNT formulations. In contrast to HSA, evidence exists to suggest that polysorbates (particularly PS20/PS80) can cause serious adverse events (e.g., hypersensitivity, anaphylaxis, and immunogenicity).
Collapse
|
27
|
Kozuch B, Weber J, Buske J, Mäder K, Garidel P, Diederichs T. Comparative Stability Study of Polysorbate 20 and Polysorbate 80 Related to Oxidative Degradation. Pharmaceutics 2023; 15:2332. [PMID: 37765302 PMCID: PMC10537708 DOI: 10.3390/pharmaceutics15092332] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The surfactants polysorbate 20 (PS20) and polysorbate 80 (PS80) are utilized to stabilize protein drugs. However, concerns have been raised regarding the degradation of PSs in biologics and the potential impact on product quality. Oxidation has been identified as a prevalent degradation mechanism under pharmaceutically relevant conditions. So far, a systematic stability comparison of both PSs under pharmaceutically relevant conditions has not been conducted and little is known about the dependence of oxidation on PS concentration. Here, we conducted a comparative stability study to investigate (i) the different oxidative degradation propensities between PS20 and PS80 and (ii) the impact of PS concentration on oxidative degradation. PS20 and PS80 in concentrations ranging from 0.1 mg⋅mL-1 to raw material were stored at 5, 25, and 40 °C for 48 weeks in acetate buffer pH 5.5 and water, respectively. We observed a temperature-dependent oxidative degradation of the PSs with strong (40 °C), moderate (25 °C), and weak/no degradation (5 °C). Especially at elevated temperatures such as 40 °C, fast oxidative PS degradation processes were detected. In this case study, a stronger degradation and earlier onset of oxidation was observed for PS80 in comparison to PS20, detected via the fluorescence micelle assay. Additionally, degradation was found to be strongly dependent on PS concentration, with significantly less oxidative processes at higher PS concentrations. Iron impurities, oxygen in the vial headspaces, and the pH values of the formulations were identified as the main contributing factors to accelerate PS oxidation.
Collapse
Affiliation(s)
- Benedykt Kozuch
- PDB-TIP, Innovation Unit, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Johanna Weber
- Institute of Pharmacy, Faculty of Biosciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle, Germany
| | - Julia Buske
- PDB-TIP, Innovation Unit, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty of Biosciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle, Germany
| | - Patrick Garidel
- PDB-TIP, Innovation Unit, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Tim Diederichs
- PDB-TIP, Innovation Unit, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| |
Collapse
|
28
|
Li J, Sonje J, Suryanarayanan R. Role of Poloxamer 188 in Preventing Ice-Surface-Induced Protein Destabilization during Freeze-Thawing. Mol Pharm 2023; 20:4587-4596. [PMID: 37535010 DOI: 10.1021/acs.molpharmaceut.3c00312] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
The phase behavior of poloxamer 188 (P188) in aqueous solutions, characterized by differential scanning calorimetry (DSC) and synchrotron X-ray diffractometry, revealed solute crystallization during both freezing and thawing. Sucrose and trehalose inhibited P188 crystallization during freeze-thawing (FT). While trehalose inhibited P188 crystallization only during cooling, sucrose completely suppressed P188 crystallization during both cooling and heating. Lactate dehydrogenase (LDH) served as a model protein to evaluate the stabilizing effect of P188. The ability of P188, over a concentration range of 0.003-0.800% w/v, to prevent LDH (10 μg/mL) destabilization was evaluated. After five FT cycles, the aggregation behavior (by dynamic light scattering) and activity recovery were evaluated. While LDH alone was sensitive to interfacial stress, P188 at concentrations of ≥0.100% w/v stabilized the protein. However, as the surfactant concentration decreased, protein aggregation after FT increased. The addition of sugar (1.0% w/v; sucrose or trehalose) improved the stabilizing function of P188 at lower concentrations (≤0.010% w/v), possibly due to the inhibition of surfactant crystallization. Based on a comparison with the stabilization effect of polysorbate (both 20 and 80), it was evident that P188 could be a promising alternative surfactant in frozen protein formulations. However, when the surfactant concentration is low, the potential for P188 crystallization and the consequent compromise in its functionality warrant careful consideration.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jayesh Sonje
- Pfizer Biotherapeutics, Pfizer Inc., Andover, Massachusetts 01810, United States
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
29
|
Papež P, Merzel F, Praprotnik M. Rotational Dynamics of a Protein under Shear Flow Studied by the Eckart Frame Formalism. J Phys Chem B 2023; 127:7231-7243. [PMID: 37556834 PMCID: PMC10461304 DOI: 10.1021/acs.jpcb.3c02324] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/20/2023] [Indexed: 08/11/2023]
Abstract
Proteins are natural polymers that play an essential role in both living organisms and biotechnological applications. During certain bioprocessing steps, they can be exposed to significant mechanical stress induced by, for example, shear flow or sonication, resulting in reduced therapeutic efficacy, aggregation, or even a loss of activity. For this reason, there is a need to understand and determine the susceptibility of the protein activity to the experienced mechanical stress. To acquire this knowledge, it is necessary to study the rotational dynamics of the protein. Commonly, the rotational dynamics of soft molecules is interpreted based on a theoretical analysis performed in an inertial laboratory frame. However, the obtained angular velocity mixes pure rotations and vibrations with angular momentum, consequently lacking a clear dynamical interpretation. On the other hand, the use of the noninertial internal Eckart frame allows the determination of pure angular velocity as it minimizes the coupling between the rotational and vibrational degrees of freedom. In the present work, by conducting open-boundary molecular dynamics simulations and exploiting the Eckart frame formalism, we study the rotational dynamics of a small protein under the shear flow of various strengths. Our results show that the angular velocity increases nonlinearly with increasing shear rate. Furthermore, the protein gains vibrational angular momentum at higher shear rates, which is reflected in the higher angular velocity computed by employing the Eckart frame formalism and confirmed by analysis of the contributions to the total kinetic energy of the biomolecule.
Collapse
Affiliation(s)
- Petra Papež
- Theory
Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- Department
of Physics, Faculty of Mathematics and Physics, University of Ljubljana, Jadranska 19, SI-1000 Ljubljana, Slovenia
| | - Franci Merzel
- Theory
Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Matej Praprotnik
- Theory
Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- Department
of Physics, Faculty of Mathematics and Physics, University of Ljubljana, Jadranska 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
30
|
Shi M, McHugh KJ. Strategies for overcoming protein and peptide instability in biodegradable drug delivery systems. Adv Drug Deliv Rev 2023; 199:114904. [PMID: 37263542 PMCID: PMC10526705 DOI: 10.1016/j.addr.2023.114904] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
The global pharmaceutical market has recently shifted its focus from small molecule drugs to peptide, protein, and nucleic acid drugs, which now comprise a majority of the top-selling pharmaceutical products on the market. Although these biologics often offer improved drug specificity, new mechanisms of action, and/or enhanced efficacy, they also present new challenges, including an increased potential for degradation and a need for frequent administration via more invasive administration routes, which can limit patient access, patient adherence, and ultimately the clinical impact of these drugs. Controlled-release systems have the potential to mitigate these challenges by offering superior control over in vivo drug levels, localizing these drugs to tissues of interest (e.g., tumors), and reducing administration frequency. Unfortunately, adapting controlled-release devices to release biologics has proven difficult due to the poor stability of biologics. In this review, we summarize the current state of controlled-release peptides and proteins, discuss existing techniques used to stabilize these drugs through encapsulation, storage, and in vivo release, and provide perspective on the most promising opportunities for the clinical translation of controlled-release peptides and proteins.
Collapse
Affiliation(s)
- Miusi Shi
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; Department of Chemistry, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
31
|
Mould R, Sargent PW, Huang Y, Fields AL, Zhang L, Herbert FC, Stewart SL, Wang T. Impact of Primary Container Closure System on PS80 Oxidation and the Mechanistic Understanding. Pharm Res 2023; 40:1965-1976. [PMID: 37434039 DOI: 10.1007/s11095-023-03556-3] [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: 03/21/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE Polysorbate oxidation can potentially lead to protein degradation and loss of potency, which has been a challenge for the pharmaceutical industry for decades. Many factors have been reported to impact polysorbate oxidation rate, including types of elemental impurities, peroxide content, pH, light exposure, grades of polysorbate, etc. Even though there are many publications in this field, the impact of primary container closure system on PS80 oxidation has not been systematically studied or reported. The purpose of the current study is to close this gap. METHODS Placebo PS80 formulations were prepared and filled into different container-closure systems (CCS), including different types of glass vials and polymer vials. Oleic acid content was monitored on stability as a surrogate value for PS80 content, which will decline upon oxidation. ICP-MS analysis and metal spiking studies were carried out to correlate the PS80 oxidation rate with metals leached from primary containers. RESULTS PS80 degrades via oxidation at the fastest rate in glass vials with high coefficient of expansion (COE), followed by glass vials with low coefficient of expansion, while polymer vials minimized the oxidation of PS80 in most formulation conditions explored in this paper. ICP-MS analysis demonstrated that 1) 51 COE glass has more metal leachables than 33 COE glass in this study; and 2) More metal leachables correlates with faster PS80 oxidation. Metal spiking studies confirmed the hypothesis that aluminum and iron have a synergistic catalysis effect on PS80 oxidation. CONCLUSIONS Primary containers of drug products play a significant role in the rate of PS80 oxidation. This study revealed a new major contributor to PS80 oxidation and potential mitigation strategy for biological drug products.
Collapse
Affiliation(s)
- Ryan Mould
- Lilly Research Laboratories: Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Yining Huang
- Lilly Research Laboratories: Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Allison L Fields
- Lilly Research Laboratories: Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Lin Zhang
- Lilly Research Laboratories: Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | - Tingting Wang
- Lilly Research Laboratories: Eli Lilly and Company, Indianapolis, Indiana, USA.
| |
Collapse
|
32
|
Sarcina L, Scandurra C, Di Franco C, Caputo M, Catacchio M, Bollella P, Scamarcio G, Macchia E, Torsi L. A stable physisorbed layer of packed capture antibodies for high-performance sensing applications. JOURNAL OF MATERIALS CHEMISTRY. C 2023; 11:9093-9106. [PMID: 37457868 PMCID: PMC10341389 DOI: 10.1039/d3tc01123b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/10/2023] [Indexed: 07/18/2023]
Abstract
Antibody physisorption at a solid interface is a very interesting phenomenon that has important effects on applications such as the development of novel biomaterials and the rational design and fabrication of high-performance biosensors. The strategy selected to immobilize biorecognition elements can determine the performance level of a device and one of the simplest approaches is physical adsorption, which is cost-effective, fast, and compatible with printing techniques as well as with green-chemistry processes. Despite its huge advantages, physisorption is very seldom adopted, as there is an ingrained belief that it does not lead to high performance because of its lack of uniformity and long-term stability, which, however, have never been systematically investigated, particularly for bilayers of capture antibodies. Herein, the homogeneity and stability of an antibody layer against SARS-CoV-2-Spike1 (S1) protein physisorbed onto a gold surface have been investigated by means of multi-parametric surface plasmon resonance (MP-SPR). A surface coverage density of capture antibodies as high as (1.50 ± 0.06) × 1012 molecules per cm-2 is measured, corresponding to a thickness of 12 ± 1 nm. This value is compatible with a single monolayer of homogeneously deposited antibodies. The effect of the ionic strength (is) of the antibody solution in controlling physisorption of the protein was thoroughly investigated, demonstrating an enhancement in surface coverage at lower ionic strength. An atomic force microscopy (AFM) investigation shows a globular structure attributed to is-related aggregations of antibodies. The long-term stability over two weeks of the physisorbed proteins was also assessed. High-performance sensing was proven by evaluating figures of merit, such as the limit of detection (2 nM) and the selectivity ratio between a negative control and the sensing experiment (0.04), which is the best reported performance for an SPR S1 protein assay. These figures of merit outmatch those measured with more sophisticated biofunctionalization procedures involving chemical bonding of the capture antibodies to the gold surface. The present study opens up interesting new pathways toward the achievement of a cost-effective and scalable biofunctionalization protocol, which could guarantee the prolonged stability of the biolayer and easy handling of the biosensing system.
Collapse
Affiliation(s)
- Lucia Sarcina
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4 70125 Bari Italy
| | - Cecilia Scandurra
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4 70125 Bari Italy
| | - Cinzia Di Franco
- CNR - Institute of Photonics and Nanotechnologies 70126 Bari Italy
| | - Mariapia Caputo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro 70126 Bari Italy
| | - Michele Catacchio
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro 70126 Bari Italy
| | - Paolo Bollella
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4 70125 Bari Italy
- CSGI (Centre for Colloid and Surface Science), Via E. Orabona 4 70125 Bari Italy
| | - Gaetano Scamarcio
- Dipartimento Interateneo di Fisica "M. Merlin", Università degli Studi di Bari Aldo Moro 70126 Bari Italy
- CSGI (Centre for Colloid and Surface Science), Via E. Orabona 4 70125 Bari Italy
| | - Eleonora Macchia
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro 70126 Bari Italy
- CSGI (Centre for Colloid and Surface Science), Via E. Orabona 4 70125 Bari Italy
- The Faculty of Science and Engineering, Åbo Akademi University 20500 Turku Finland
| | - Luisa Torsi
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4 70125 Bari Italy
- CSGI (Centre for Colloid and Surface Science), Via E. Orabona 4 70125 Bari Italy
- The Faculty of Science and Engineering, Åbo Akademi University 20500 Turku Finland
| |
Collapse
|
33
|
Mutukuri TT, Ling J, Du Y, Su Y, Zhou QT. Effect of Buffer Salts on Physical Stability of Lyophilized and Spray-Dried Protein Formulations Containing Bovine Serum Albumin and Trehalose. Pharm Res 2023; 40:1355-1371. [PMID: 35764755 PMCID: PMC9794634 DOI: 10.1007/s11095-022-03318-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/14/2022] [Indexed: 12/30/2022]
Abstract
This study examined the effect of buffer salts on the physical stability of spray-dried and lyophilized formulations of a model protein, bovine serum albumin (BSA). BSA formulations with various buffers were dried by either lyophilization or spray drying. The protein powders were then characterized using solid-state Fourier transform infrared spectroscopy (ssFTIR), powder X-ray diffraction (PXRD), size exclusion chromatography (SEC), solid-state hydrogen/deuterium exchange with mass spectrometry (ssHDX-MS), and solid-state nuclear magnetic resonance spectroscopy (ssNMR). Particle characterizations such as Brunauer-Emmett-Teller (BET) surface area, particle size distribution, and particle morphology were also performed. Results from conventional techniques such as ssFTIR did not exhibit correlations with the physical stability of studied formulations. Deconvoluted peak areas of deuterated samples from the ssHDX-MS study showed a satisfactory correlation with the loss of the monomeric peak area measured by SEC (R2 of 0.8722 for spray-dried formulations and 0.8428 for lyophilized formulations) in the 90-day accelerated stability study conducted at 40°C. mDSC and PXRD was unable to measure phase separation in the samples right after drying. In contrast, ssNMR successfully detected the occurrence of phase separation between the succinic buffer component and protein in the lyophilized formulation, which results in a distribution of microenvironmental acidity and the subsequent loss of long-term stability. Moreover, our results suggested that buffer salts have less impact on physical stability for the spray-dried formulations than the lyophilized solids.
Collapse
Affiliation(s)
- Tarun Tejasvi Mutukuri
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana, 47907, USA
| | - Jing Ling
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., South San Francisco, California, 94080, USA
| | - Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey, 07065, USA
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey, 07065, USA.
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana, 47907, USA.
| |
Collapse
|
34
|
Lisina S, Inam W, Huhtala M, Howaili F, Zhang H, Rosenholm JM. Nano Differential Scanning Fluorimetry as a Rapid Stability Assessment Tool in the Nanoformulation of Proteins. Pharmaceutics 2023; 15:pharmaceutics15051473. [PMID: 37242715 DOI: 10.3390/pharmaceutics15051473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The development and production of innovative protein-based therapeutics is a complex and challenging avenue. External conditions such as buffers, solvents, pH, salts, polymers, surfactants, and nanoparticles may affect the stability and integrity of proteins during formulation. In this study, poly (ethylene imine) (PEI) functionalized mesoporous silica nanoparticles (MSNs) were used as a carrier for the model protein bovine serum albumin (BSA). To protect the protein inside MSNs after loading, polymeric encapsulation with poly (sodium 4-styrenesulfonate) (NaPSS) was used to seal the pores. Nano differential scanning fluorimetry (NanoDSF) was used to assess protein thermal stability during the formulation process. The MSN-PEI carrier matrix or conditions used did not destabilize the protein during loading, but the coating polymer NaPSS was incompatible with the NanoDSF technique due to autofluorescence. Thus, another pH-responsive polymer, spermine-modified acetylated dextran (SpAcDEX), was applied as a second coating after NaPSS. It possessed low autofluorescence and was successfully evaluated with the NanoDSF method. Circular dichroism (CD) spectroscopy was used to determine protein integrity in the case of interfering polymers such as NaPSS. Despite this limitation, NanoDSF was found to be a feasible and rapid tool to monitor protein stability during all steps needed to create a viable nanocarrier system for protein delivery.
Collapse
Affiliation(s)
- Sofia Lisina
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Wali Inam
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Mikko Huhtala
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, 20500 Turku, Finland
| | - Fadak Howaili
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland
| |
Collapse
|
35
|
Kulakova A, Augustijn D, El Bialy I, Gentiluomo L, Greco ML, Hervø-Hansen S, Indrakumar S, Mahapatra S, Martinez Morales M, Pohl C, Polimeni M, Roche A, Svilenov HL, Tosstorff A, Zalar M, Curtis R, Derrick JP, Frieß W, Golovanov AP, Lund M, Nørgaard A, Khan TA, Peters GHJ, Pluen A, Roessner D, Streicher WW, van der Walle CF, Warwicker J, Uddin S, Winter G, Bukrinski JT, Rinnan Å, Harris P. Chemometrics in Protein Formulation: Stability Governed by Repulsion and Protein Unfolding. Mol Pharm 2023. [PMID: 37146162 DOI: 10.1021/acs.molpharmaceut.3c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Therapeutic proteins can be challenging to develop due to their complexity and the requirement of an acceptable formulation to ensure patient safety and efficacy. To date, there is no universal formulation development strategy that can identify optimal formulation conditions for all types of proteins in a fast and reliable manner. In this work, high-throughput characterization, employing a toolbox of five techniques, was performed on 14 structurally different proteins formulated in 6 different buffer conditions and in the presence of 4 different excipients. Multivariate data analysis and chemometrics were used to analyze the data in an unbiased way. First, observed changes in stability were primarily determined by the individual protein. Second, pH and ionic strength are the two most important factors determining the physical stability of proteins, where there exists a significant statistical interaction between protein and pH/ionic strength. Additionally, we developed prediction methods by partial least-squares regression. Colloidal stability indicators are important for prediction of real-time stability, while conformational stability indicators are important for prediction of stability under accelerated stress conditions at 40 °C. In order to predict real-time storage stability, protein-protein repulsion and the initial monomer fraction are the most important properties to monitor.
Collapse
Affiliation(s)
- Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Dillen Augustijn
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Inas El Bialy
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Lorenzo Gentiluomo
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | - Maria Laura Greco
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Stefan Hervø-Hansen
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | | | - Marcello Martinez Morales
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Christin Pohl
- Novozymes A/S, Krogshoejvej 36, Bagsvaerd 2880, Denmark
| | - Marco Polimeni
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Aisling Roche
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Hristo L Svilenov
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Andreas Tosstorff
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Matja Zalar
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Robin Curtis
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Alexander P Golovanov
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Mikael Lund
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | | | - Tarik A Khan
- Pharmaceutical Development & Supplies, Pharma Technical Development Biologics Europe, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Alain Pluen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, U.K
| | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | | | - Christopher F van der Walle
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Jim Warwicker
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Shahid Uddin
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | | | - Åsmund Rinnan
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| |
Collapse
|
36
|
Park H, Ha E, Kim J, Kim M. Injectable sustained-release poly(lactic-co-glycolic acid) (PLGA) microspheres of exenatide prepared by supercritical fluid extraction of emulsion process based on a design of experiment approach. Bioeng Transl Med 2023; 8:e10485. [PMID: 37206215 PMCID: PMC10189459 DOI: 10.1002/btm2.10485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/18/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
This study aimed to develop an improved sustained-release (SR) PLGA microsphere of exenatide using supercritical fluid extraction of emulsions (SFEE). As a translational research, we investigated the effect of various process parameters on the fabrication of exenatide-loaded PLGA microspheres by SFEE (ELPM_SFEE) using the Box-Behnken design (BBD), a design of experiment approach. Further, ELPM obtained under optimized conditions and satisfying all the response criteria were compared with PLGA microspheres prepared using the conventional solvent evaporation (ELPM_SE) method through various solid-state characterizations and in vitro and in vivo evaluations. The four process parameters selected as independent variables were pressure (X 1), temperature (X 2), stirring rate (X 3), and flow ratio (X 4). The effects of these independent variables on five responses, namely the particle size, its distribution (SPAN value), encapsulation efficiency (EE), initial drug burst release (IBR), and residual organic solvent, were evaluated using BBD. Based on the experimental results, a desirable range of combinations of various variables in the SFEE process was determined by graphical optimization. Solid-state characterization and in vitro evaluation revealed that ELPM_SFEE improved properties, including a smaller particle size and SPAN value, higher EE, lower IBR, and lower residual solvent. Furthermore, the pharmacokinetic and pharmacodynamic study results indicated better in vivo efficacy with desirable SR properties, including a reduction in blood glucose levels, weight gain, and food intake, for ELPM_SFEE than those generated using SE. Therefore, the potential drawback of conventional technologies such as the SE for the preparation of injectable SR PLGA microspheres could be improved by optimizing the SFEE process.
Collapse
Affiliation(s)
- Heejun Park
- College of PharmacyDuksung Women's UniversitySeoulSouth Korea
| | - Eun‐Sol Ha
- College of PharmacyPusan National UniversityBusanSouth Korea
| | - Jeong‐Soo Kim
- Dong‐A ST Research InstituteDong‐A ST Co. Ltd.Giheung‐guYongin‐siGyeonggiSouth Korea
| | - Min‐Soo Kim
- College of PharmacyPusan National UniversityBusanSouth Korea
| |
Collapse
|
37
|
Jha R, Mayanovic RA. A Review of the Preparation, Characterization, and Applications of Chitosan Nanoparticles in Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13081302. [PMID: 37110887 PMCID: PMC10140956 DOI: 10.3390/nano13081302] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 06/12/2023]
Abstract
Chitosan is a fibrous compound derived from chitin, which is the second most abundant natural polysaccharide and is produced by crustaceans, including crabs, shrimps, and lobsters. Chitosan has all of the important medicinal properties, including biocompatibility, biodegradability, and hydrophilicity, and it is relatively nontoxic and cationic in nature. Chitosan nanoparticles are particularly useful due to their small size, providing a large surface-to-volume ratio, and physicochemical properties that may differ from that of their bulk counterparts; thus, chitosan nanoparticles (CNPs) are widely used in biomedical applications and, particularly, as contrast agents for medical imaging and as vehicles for drug and gene delivery into tumors. Because CNPs are formed from a natural biopolymer, they can readily be functionalized with drugs, RNA, DNA, and other molecules to target a desired result in vivo. Furthermore, chitosan is approved by the United States Food and Drug Administration as being Generally Recognized as Safe (GRAS). This paper reviews the structural characteristics and various synthesis methods used to produce chitosan nanoparticles and nanostructures, such as ionic gelation, microemulsion, polyelectrolyte complexing, emulsification solvent diffusion, and the reverse micellar method. Various characterization techniques and analyses are also discussed. In addition, we review drug delivery applications of chitosan nanoparticles, including for ocular, oral, pulmonary, nasal, and vaginal methodologies, and applications in cancer therapy and tissue engineering.
Collapse
|
38
|
Diederichs T, Mittag JJ, Humphrey J, Voss S, Carle S, Buske J, Garidel P. Existence of a superior polysorbate fraction in respect to protein stabilization and particle formation? Int J Pharm 2023; 635:122660. [PMID: 36740078 DOI: 10.1016/j.ijpharm.2023.122660] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/02/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Biologicals including monoclonal antibodies are the current flagships in pharmaceutical industry. However, they are exposed to a multitude of destabilization conditions like for instance hydrophobic interfaces, leading to reduced biological activity. Polysorbates are commonly applied to effectively stabilize these active pharmaceutical ingredients against colloidal stress. Nevertheless, chemical instability of polysorbate via hydrolysis or oxidation results in degradation products that might form particles via phase separation. Polysorbates are mixtures of hundreds of individual components, and recently purer quality grades with reduced variations in the fatty acid composition are available. As the protective function of polysorbate itself is not completely understood, even less is known about its individual components, raising the question of the existence of a superior polysorbate species in respect to protein stabilization or degradation susceptibility. Here, we evaluated the protective function of four main fractions of polysorbate 20 (PS20) in agitation studies with monoclonal antibodies, followed by particle analysis as well as protein and polysorbate content determination. The commercially-available inherent mixtures PS20 high purity and PS20 all-laurate, as well as the fraction isosorbide-POE-monolaurate showed superior protection against mechanical-induced stress (visual inspection and turbidity) at the air-water interface in comparison to sole sorbitan-POE-monolaurate, -dilaurate, and -trilaurate. Fractions composed mainly of higher-order esters like sorbitan-POE-dilaurate and sorbitan-POE-trilaurate indicated high turbidities as indication for subvisible and small particles accompanied by a reduced protein monomer content after agitation. For the isosorbide-POE-monolaurates as well as for the inherent polysorbate mixtures no obvious differences in protein content and protein aggregation (SEC) were observed, reflecting the observations from visual appearance. However, absolute polysorbate concentrations vary drastically between different species in the actual formulations. As there are still open questions in respect to protein specificity or regarding mixtures versus individual components of PS20, further studies must be performed, to gain a better understanding of a "generalized" stabilizing effect of polysorbates on monoclonal antibodies. The knowledge of the characteristics of individual polysorbate species can have the potential to pave the way to superior detergents in respect to protein stabilization and/or degradation susceptibility.
Collapse
Affiliation(s)
- Tim Diederichs
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany.
| | - Judith J Mittag
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - James Humphrey
- Croda Europe Ltd, Cowick Hall, DN14 9AA, Snaith, United Kingdom
| | - Söhnke Voss
- Croda Europe Ltd, Cowick Hall, DN14 9AA, Snaith, United Kingdom
| | - Stefan Carle
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Julia Buske
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany.
| |
Collapse
|
39
|
Zäh M, Brandenbusch C, Winter G, Sadowski G. Predicting the amorphous-phase composition during lyophilization. Int J Pharm 2023; 636:122836. [PMID: 36940838 DOI: 10.1016/j.ijpharm.2023.122836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
The glass-transition temperature and the composition of the amorphous phase/maximally concentrated solution (classically referred to as Tg' and wg', respectively) as function of added excipients are crucial for the design of lyophilization processes. Whereas the determination of Tg' can be accomplished easily using mDSC, the determination of wg' poses challenges, since the experimental effort needs to be redone for each new excipient mixture (limited transferability of the results possible). In this work, an approach was developed which allows to predict wg' for (1) single excipients, (2) given compositions of a binary excipient mixture, and (3) single excipients in aqueous (model) protein solutions using the thermodynamic model PC-SAFT and one experimental data point of Tg'. Sucrose, trehalose, fructose, sorbitol, and lactose were considered as single excipients. The binary excipient mixture consisted of sucrose and ectoine. The model protein was bovine serum albumin in combination with sucrose. The results reveal that the new approach can precisely predict wg' in the systems considered, including the non-linear course of wg' identified for different sucrose/ectoine ratios. The same applies to the course of wg' as function of the protein concentration. This newly developed approach allows for the reduction of the experimental effort to a minimum.
Collapse
Affiliation(s)
- Maximilian Zäh
- TU Dortmund University, Laboratory of Thermodynamics, Department of Biochemical and Chemical Engineering, Emil-Figge-Str. 70, 44227 Dortmund, Germany.
| | - Christoph Brandenbusch
- TU Dortmund University, Laboratory of Thermodynamics, Department of Biochemical and Chemical Engineering, Emil-Figge-Str. 70, 44227 Dortmund, Germany.
| | - Gerhard Winter
- LMU Munich, Chair of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Butenandtstr. 5, 81377 Munich, Germany.
| | - Gabriele Sadowski
- TU Dortmund University, Laboratory of Thermodynamics, Department of Biochemical and Chemical Engineering, Emil-Figge-Str. 70, 44227 Dortmund, Germany.
| |
Collapse
|
40
|
Zhou X, Sinkjær AW, Zhang M, Pinholt HD, Nielsen HM, Hatzakis NS, van de Weert M, Foderà V. Heterogeneous and Surface-Catalyzed Amyloid Aggregation Monitored by Spatially Resolved Fluorescence and Single Molecule Microscopy. J Phys Chem Lett 2023; 14:912-919. [PMID: 36669144 DOI: 10.1021/acs.jpclett.2c03400] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Amyloid aggregation is associated with many diseases and may also occur in therapeutic protein formulations. Addition of co-solutes is a key strategy to modulate the stability of proteins in pharmaceutical formulations and select inhibitors for drug design in the context of diseases. However, the heterogeneous nature of this multicomponent system in terms of structures and mechanisms poses a number of challenges for the analysis of the chemical reaction. Using insulin as protein system and polysorbate 80 as co-solute, we combine a spatially resolved fluorescence approach with single molecule microscopy and machine learning methods to kinetically disentangle the different contributions from multiple species within a single aggregation experiment. We link the presence of interfaces to the degree of heterogeneity of the aggregation kinetics and retrieve the rate constants and underlying mechanisms for single aggregation events. Importantly, we report that the mechanism of inhibition of the self-assembly process depends on the details of the growth pathways of otherwise macroscopically identical species. This information can only be accessed by the analysis of single aggregate events, suggesting our method as a general tool for a comprehensive physicochemical characterization of self-assembly reactions.
Collapse
Affiliation(s)
- Xin Zhou
- Drug Delivery and Biophysics of Biopharmaceuticals and Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Anders Wilgaard Sinkjær
- Drug Delivery and Biophysics of Biopharmaceuticals and Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Min Zhang
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
- Nano-Science Center, University of Copenhagen Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Henrik Dahl Pinholt
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Hanne Mørck Nielsen
- Drug Delivery and Biophysics of Biopharmaceuticals and Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Nano-Science Center, University of Copenhagen Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Nikos S Hatzakis
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
- Nano-Science Center, University of Copenhagen Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Marco van de Weert
- Drug Delivery and Biophysics of Biopharmaceuticals and Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Nano-Science Center, University of Copenhagen Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Vito Foderà
- Drug Delivery and Biophysics of Biopharmaceuticals and Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Nano-Science Center, University of Copenhagen Universitetsparken 5, 2100 Copenhagen, Denmark
| |
Collapse
|
41
|
Vieillard V, Paul M. Physicochemical stability study of a biosimilar of Bevacizumab in vials and after dilution in 0.9% NaCl in polyolefin intravenous bags. PHARMACEUTICAL TECHNOLOGY IN HOSPITAL PHARMACY 2023. [DOI: 10.1515/pthp-2022-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Abstract
Objectives
Bevacizumab was first marketed in 2005. Since then, its stability has been extensively studied. The arrival of numerous biosimilars on the market has called into question these stabilities and organisation within reconstitution units. To study the stability of the Bevacizumab biosimilar Alymsys® marketed by Zentiva laboratory in ready-to-use vials at a concentration of 25 mg/mL and following dilution to obtain final concentrations of 1.4 and 16.5 mg/mL and storage in polyolefin IV bags at 4 °C. In parallel, the impact of a storage temperature excursion at 25 °C for three days and storage of the vial before opening at room temperature (25 ± 2 °C) and after opening at 4 °C was studied.
Methods
The vials were supplied by Zentiva laboratory. The vials (three batches) were diluted to the final concentrations of 1.4 or 16.5 mg/mL in 100 mL IV bags of NaCl. The IV bags and vials were stored at 4 °C and at room temperature throughout the duration of the study. The physico-chemical stability was tested using the following methods: turbidimetry, UV spectrometry and fluorescence, dynamic light scattering, ion exchange and steric exclusion chromatography, pH, osmolality and density.
Results
Out of all the parameters studied, for the two concentrations and standard storage conditions (90 days at +4 °C) or after a three-day temperature excursion at +25 °C, no modification was detected for the three batches tested with respect to physical and chemical stability. Hence, no signs of physical instability were observed, with, in particular, the absence of formation of submicron or micron sized aggregates and particles. The steric exclusion chromatography profiles did not demonstrate any oligomer formation or molecular structure rupture. Ion exchange chromatography did not demonstrate any significant modification in the distribution of charge variants. Derivative UV and fluorescence spectral analysis did not demonstrate any modification. The thermal denaturation curves were identical, suggesting the absence of thermodynamic destabilisation. Identical results were observed for the vials stored for 60 days at 4 °C after opening. Finally, only ion exchange chromatography demonstrated a slight change after 45 days of storage at 25 °C for vials before opening.
Conclusions
After dilution in sterile conditions with 0.9% NaCl in polyolefin IV bags, at the usual concentrations of 1.4 and 16.5 mg/mL, the Bevacizumab biosimilar Alymsys® is stable for at least three months at 4 °C protected from light and after a three-day temperature excursion at +25 °C. The same conclusions can be reached for the 25 mg/mL vials stored for 60 days at +4 °C after opening. However, the stability of vials stored at 25 °C before opening is no longer guaranteed beyond 15 days.
Collapse
Affiliation(s)
| | - Muriel Paul
- Service Pharmacie , CHU Henri Mondor , Créteil , France
| |
Collapse
|
42
|
Strofaldi A, Quinn MK, Seddon AM, McManus JJ. Polymorphic protein phase transitions driven by surface anisotropy. J Chem Phys 2023; 158:014905. [PMID: 36610968 DOI: 10.1063/5.0125452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phase transitions of proteins are strongly influenced by surface chemical modifications or mutations. Human γD-crystallin (HGD) single-mutants have been extensively studied because they are associated with the onset of juvenile cataract. However, they have also provided a rich library of molecules to examine how specific inter-protein interactions direct protein assembly, providing new insights and valuable experimental data for coarse-grained patchy-particle models. Here, we demonstrate that the addition of new inter-protein interactions by mutagenesis is additive and increases the number and variety of condensed phases formed by proteins. When double mutations incorporating two specific single point mutations are made, the properties of both single mutations are retained in addition to the formation of a new condensed phase. We find that the HGD double-mutant P23VC110M self-assembles into spherical particles with retrograde solubility, orthorhombic crystals, and needle/plate shape crystals, while retaining the ability to undergo liquid-liquid phase separation. This rich polymorphism is only partially predicted by the experimental data on the constituent single mutants. We also report a previously un-characterized amorphous protein particle, with unique properties that differ from those of protein spherulites, protein particulates previously described. The particles we observe are amorphous, reversible with temperature, tens of microns in size, and perfectly spherical. When they are grown on pristine surfaces, they appear to form by homogeneous nucleation, making them unique, and we believe a new form of protein condensate. This work highlights the challenges in predicting protein behavior, which has frustrated rational assembly and crystallization but also provides rich data to develop new coarse-grained models to explain the observed polymorphism.
Collapse
Affiliation(s)
| | - Michelle K Quinn
- Department of Chemistry Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Annela M Seddon
- HH Wills Physics Laboratory, School of Physics, University of Bristol, Bristol BS8 1TL, United Kingdom
| | - Jennifer J McManus
- HH Wills Physics Laboratory, School of Physics, University of Bristol, Bristol BS8 1TL, United Kingdom
| |
Collapse
|
43
|
Development and optimization of a LC-MS based multi-attribute method (MAM) workflow for characterization of therapeutic Fc-fusion protein. Anal Biochem 2023; 660:114969. [PMID: 36343663 DOI: 10.1016/j.ab.2022.114969] [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: 06/23/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The growing complexity of novel biopharmaceutical formats, such as Fc-fusion proteins, in increasingly competitive environment has highlighted the need of high-throughput analytical platforms. Multi-attribute method (MAM) is an emerging analytical technology that utilizes liquid chromatography coupled with mass spectrometry to monitor critical quality attributes (CQAs) in biopharmaceuticals. MAM is intended to supplement or replace the conventional chromatographic and electrophoretic approaches used for quality control and drug release purpose. In this investigation, we have developed an agile sample preparation approach for deploying MAM workflow for a complex VEGFR-targeted therapeutic Fc-fusion protein. Initially, a systematic time course evaluation of tryptic digestion step was performed to achieve maximum amino acid sequence coverage of >96.5%, in a short duration of 2 h, with minimum assay artifacts. This approach facilitated precise identification of five sites of N-glycosylation with successful monitoring of other CQAs such as deamidation, oxidation, etc. Subsequently, the developed MAM workflow with suitable tryptic digestion time was qualified according to the International council for harmonisation (i.e. ICH) Q2R1 guidelines for method validation. Post-validation, the analytical workflow was also evaluated for its capability to identify unknown moieties, termed as 'New Peak Detection' (i.e. NPD), and assess fold change between the reference and non-reference samples, in a representative investigation of pH stress study. The study, thus, demonstrated the suitability of the MAM workflow for characterization of heavily glycosylated Fc-fusion proteins. Moreover, its NPD feature could offer an all-encompassing view if applied for forced degradation and stability studies.
Collapse
|
44
|
Ghosh I, Gutka H, Krause ME, Clemens R, Kashi RS. A systematic review of commercial high concentration antibody drug products approved in the US: formulation composition, dosage form design and primary packaging considerations. MAbs 2023; 15:2205540. [PMID: 37243580 DOI: 10.1080/19420862.2023.2205540] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/29/2023] Open
Abstract
Three critical aspects that define high concentration antibody products (HCAPs) are as follows: 1) formulation composition, 2) dosage form, and 3) primary packaging configuration. HCAPs have become successful in the therapeutic sector due to their unique advantage of allowing subcutaneous self-administration. Technical challenges, such as physical and chemical instability, viscosity, delivery volume limitations, and product immunogenicity, can hinder successful development and commercialization of HCAPs. Such challenges can be overcome by robust formulation and process development strategies, as well as rational selection of excipients and packaging components. We compiled and analyzed data from US Food and Drug Administration-approved and marketed HCAPs that are ≥100 mg/mL to identify trends in formulation composition and quality target product profile. This review presents our findings and discusses novel formulation and processing technologies that enable the development of improved HCAPs at ≥200 mg/mL. The observed trends can be used as a guide for further advancements in the development of HCAPs as more complex antibody-based modalities enter biologics product development.
Collapse
Affiliation(s)
- Indrajit Ghosh
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Hiten Gutka
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Mary E Krause
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Ryan Clemens
- College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Ramesh S Kashi
- Sterile Product Development, Bristol Myers Squibb, Summit, NJ, USA
| |
Collapse
|
45
|
Jakob LA, Mesurado T, Jungbauer A, Lingg N. Increase in cysteine-mediated multimerization under attractive protein-protein interactions. Prep Biochem Biotechnol 2022; 53:891-905. [PMID: 36576211 DOI: 10.1080/10826068.2022.2158471] [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] [Indexed: 12/29/2022]
Abstract
The CASPON enzyme became an interesting enzyme for fusion protein processing because it generates an authentic N-terminus. However, the high cysteine content of the CASPON enzyme may induce aggregation via disulfide-bond formation, which can reduce enzymatic activity and be considered a critical quality attribute. Different multimerization states of the CASPON enzyme were isolated by preparative size exclusion chromatography and analyzed with respect to multimerization propensity and enzymatic activity. The impact of co-solutes on multimerization was studied in solution and in adsorbed state. Furthermore, protein-protein interactions in the presence of different co-solutes were measured by self-interaction chromatography and were then correlated to the multimerization propensity. The dimer was the most stable and active species with 50% higher enzymatic activity than the tetramer. Multimerization was mainly governed by a cysteine-mediated pathway, as indicated by DTT-induced reduction of most caspase multimers. In the presence of ammonium sulfate, attractive protein-protein interactions were consistent with those observed for higher multimerization when the cysteine-mediated pathway was followed. Multimerization was also observed under attractive conditions on a chromatographic stationary phase. These findings corroborate common rules to perform protein purification with low residence time to avoid disulfide bond formation and conformational change of the protein upon adsorption.
Collapse
Affiliation(s)
- Leo A Jakob
- Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Tomás Mesurado
- Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alois Jungbauer
- Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Nico Lingg
- Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| |
Collapse
|
46
|
Nadri S, Rahmani A, Hosseini SH, Habibizadeh M, Araghi M, Mostafavi H. Prevention of peritoneal adhesions formation by core-shell electrospun ibuprofen-loaded PEG/silk fibrous membrane. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:40-48. [PMID: 35296208 DOI: 10.1080/21691401.2021.1883043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/15/2021] [Accepted: 01/24/2021] [Indexed: 06/14/2023]
Abstract
Adhesion bands are pathological fibrous tissues that create in the middle of tissues and organs, often reasons of intestinal obstruction, and female infertility. Here, we explored the anti-adhesive and inflammatory capacities of PEG/silk and Ibuprofen-loaded PEG/Silk core-shell nanofibrous membranes, respectively. The ibuprofen-loaded Silk Fibroin-Poly ethylene Glycol (SF-PEG) core-shell membrane was fabricated by electrospinning and considered in terms of morphology, surface wettability, drug release, and degradation. To reveal the membrane capability for adhesion bands inhibition, the membrane was stitched among the abdominal partition and peritoneum and then evaluated using two scoring adhesion systems. According to results, the fibrous membrane hindered cell proliferation, and the scoring systems and pathology showed that in a rat model, Ibuprofen-loaded PEG/Silk core-shell membrane caused a lightening in post-operative adhesion bands and the low-grade inflammatory reaction in animal models. Collectively, we fabricated new ibuprofen-loaded PEG/SF membranes with anti-adhesion and anti-inflammation properties. Moreover, this core-shell electrospun fibrous membrane has not even now been used to prevent peritendinous adhesion generation.
Collapse
Affiliation(s)
- Samad Nadri
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Rahmani
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Hojjat Hosseini
- Department of Pharmacology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mina Habibizadeh
- Department of Pharmacy Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahmood Araghi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Mostafavi
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
47
|
Castañeda Ruiz AJ, Shetab Boushehri MA, Phan T, Carle S, Garidel P, Buske J, Lamprecht A. Alternative Excipients for Protein Stabilization in Protein Therapeutics: Overcoming the Limitations of Polysorbates. Pharmaceutics 2022; 14:2575. [PMID: 36559072 PMCID: PMC9781097 DOI: 10.3390/pharmaceutics14122575] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Given their safety and efficiency in protecting protein integrity, polysorbates (PSs) have been the most widely used excipients for the stabilization of protein therapeutics for years. In recent decades, however, there have been numerous reports about visible or sub-visible particles in PS-containing biotherapeutic products, which is a major quality concern for parenteral drugs. Alternative excipients that are safe for parenteral administration, efficient in protecting different protein drugs against various stress conditions, effective in protein stabilization in high-concentrated liquid formulations, stable under the storage conditions for the duration of the product's shelf-life, and compatible with other formulation components and the primary packaging are highly sought after. The aim of this paper is to review potential alternative excipients from different families, including surfactants, carbohydrate- and amino acid-based excipients, synthetic amphiphilic polymers, and ionic liquids that enable protein stabilization. For each category, important characteristics such as the ability to stabilize proteins against thermal and mechanical stresses, current knowledge related to the safety profile for parenteral administration, potential interactions with other formulation components, and primary packaging are debated. Based on the provided information and the detailed discussion thereof, this paper may pave the way for the identification or development of efficient excipients for biotherapeutic protein stabilization.
Collapse
Affiliation(s)
- Angel J. Castañeda Ruiz
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, 53121 Bonn, Germany
| | | | - Tamara Phan
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Stefan Carle
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Julia Buske
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, 53121 Bonn, Germany
| |
Collapse
|
48
|
Akbarian M, Chen SH. Instability Challenges and Stabilization Strategies of Pharmaceutical Proteins. Pharmaceutics 2022; 14:2533. [PMID: 36432723 PMCID: PMC9699111 DOI: 10.3390/pharmaceutics14112533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Maintaining the structure of protein and peptide drugs has become one of the most important goals of scientists in recent decades. Cold and thermal denaturation conditions, lyophilization and freeze drying, different pH conditions, concentrations, ionic strength, environmental agitation, the interaction between the surface of liquid and air as well as liquid and solid, and even the architectural structure of storage containers are among the factors that affect the stability of these therapeutic biomacromolecules. The use of genetic engineering, side-directed mutagenesis, fusion strategies, solvent engineering, the addition of various preservatives, surfactants, and additives are some of the solutions to overcome these problems. This article will discuss the types of stress that lead to instabilities of different proteins used in pharmaceutics including regulatory proteins, antibodies, and antibody-drug conjugates, and then all the methods for fighting these stresses will be reviewed. New and existing analytical methods that are used to detect the instabilities, mainly changes in their primary and higher order structures, are briefly summarized.
Collapse
Affiliation(s)
| | - Shu-Hui Chen
- Department of Chemistry, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
49
|
Ullah A, Kwon HT, Lim SI. Albumin: A Multi-talented Clinical and Pharmaceutical Player. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-022-0104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
50
|
Young KA, Mancera RL. Review: Investigating the aggregation of amyloid beta with surface plasmon resonance: Do different approaches yield different results? Anal Biochem 2022; 654:114828. [PMID: 35931183 DOI: 10.1016/j.ab.2022.114828] [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: 03/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022]
Abstract
Aggregation of amyloid beta into amyloid plaques in the brain is a hallmark characteristic of Alzheimer's disease. Therapeutics aimed at preventing or retarding amyloid formation often rely on detailed characterization of the underlying mechanism and kinetics of protein aggregation. Surface plasmon resonance (SPR) spectroscopy is a robust technique used to determine binding affinity and kinetics of biomolecular interactions. This approach has been used to characterize the mechanism of aggregation of amyloid beta but there are multiple pitfalls that need to be addressed when working with this and other amyloidogenic proteins. The choice of method for analyte preparation and ligand immobilization to a sensor chip can lead to different theoretical and practical implications in terms of the mathematical modelling of binding data, different mechanisms of binding and the presence of different interacting species. This review examines preparation methods for SPR characterisation of the aggregation of amyloid beta and their influence on the findings derived from such studies.
Collapse
Affiliation(s)
- Kimberly A Young
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia.
| |
Collapse
|