151
|
Nakayoshi T, Wanita K, Kato K, Kurimoto E, Oda A. Computational analysis of nonenzymatic deamidation of asparagine residues catalysed by acetic acid. Mol Phys 2021. [DOI: 10.1080/00268976.2020.1827176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Tomoki Nakayoshi
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Kota Wanita
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Koichi Kato
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Department of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | - Eiji Kurimoto
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Akifumi Oda
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
- Institute for Protein Research, Suita, Japan
| |
Collapse
|
152
|
Boukhari SE, Chahid M. Effects of inclusions and the presence of an adsorbing surface on lateral and flip-flop phase transitions in fluid membranes. SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2021.e00736] [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] Open
|
153
|
Chae H, Cho S, Jeong M, Kwon K, Choi D, Lee J, Nam W, Hong J, Lee J, Yoon S, Hong H. Improvement of Biophysical Properties and Affinity of a Human Anti-L1CAM Therapeutic Antibody through Antibody Engineering Based on Computational Methods. Int J Mol Sci 2021; 22:ijms22136696. [PMID: 34206616 PMCID: PMC8268072 DOI: 10.3390/ijms22136696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 01/14/2023] Open
Abstract
The biophysical properties of therapeutic antibodies influence their manufacturability, efficacy, and safety. To develop an anti-cancer antibody, we previously generated a human monoclonal antibody (Ab417) that specifically binds to L1 cell adhesion molecule with a high affinity, and we validated its anti-tumor activity and mechanism of action in human cholangiocarcinoma xenograft models. In the present study, we aimed to improve the biophysical properties of Ab417. We designed 20 variants of Ab417 with reduced aggregation propensity, less potential post-translational modification (PTM) motifs, and the lowest predicted immunogenicity using computational methods. Next, we constructed these variants to analyze their expression levels and antigen-binding activities. One variant (Ab612)—which contains six substitutions for reduced surface hydrophobicity, removal of PTM, and change to the germline residue—exhibited an increased expression level and antigen-binding activity compared to Ab417. In further studies, compared to Ab417, Ab612 showed improved biophysical properties, including reduced aggregation propensity, increased stability, higher purification yield, lower pI, higher affinity, and greater in vivo anti-tumor efficacy. Additionally, we generated a highly productive and stable research cell bank (RCB) and scaled up the production process to 50 L, yielding 6.6 g/L of Ab612. The RCB will be used for preclinical development of Ab612.
Collapse
Affiliation(s)
- Heesu Chae
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Seulki Cho
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea;
| | - Munsik Jeong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Kiyoung Kwon
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Dongwook Choi
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Chungcheongbuk-do, Cheongju-si 28160, Korea;
| | - Jaeyoung Lee
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Woosuk Nam
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Jisu Hong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Jiwoo Lee
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Seonjoo Yoon
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
- Correspondence: (S.Y.); (H.H.); Tel.: +82-10-2305-9704 (S.Y.); +82-10-5430-0480 (H.H.)
| | - Hyojeong Hong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea;
- Correspondence: (S.Y.); (H.H.); Tel.: +82-10-2305-9704 (S.Y.); +82-10-5430-0480 (H.H.)
| |
Collapse
|
154
|
F Nahhas A, F Nahhas A, J Webster T. Nanoscale pathogens treated with nanomaterial-like peptides: a platform technology appropriate for future pandemics. Nanomedicine (Lond) 2021; 16:1237-1254. [PMID: 33988037 PMCID: PMC8120868 DOI: 10.2217/nnm-2020-0447] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/29/2021] [Indexed: 01/13/2023] Open
Abstract
Viral infections are historically very difficult to treat. Although imperfect and time-consuming to develop, we do have some conventional vaccine and therapeutic approaches to stop viral spreading. Most importantly, all of this takes significant time while viruses continue to wreak havoc on our healthcare system. Furthermore, viral infections are accompanied by a weakened immune system which is often overlooked in antiviral drug strategies and requires additional drug development. In this review, for the first time, we touch on some promising alternative approaches to treat viral infections, specifically those focused on the use of platform nanomaterials with antiviral peptides. In doing so, this review presents a timely discussion of how we need to change our old way of treating viruses into one that can quickly meet the demands of COVID-19, as well as future pandemic-causing viruses, which will come.
Collapse
Affiliation(s)
- Alaa F Nahhas
- Biochemistry Department, College of Science, King Abdulaziz University, Jeddah 21589, KSA
| | - Alrayan F Nahhas
- Biochemistry Department, College of Science, King Abdulaziz University, Jeddah 21589, KSA
| | - Thomas J Webster
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
155
|
Pavani P, Kumar K, Rani A, Venkatesu P, Lee MJ. The influence of sodium phosphate buffer on the stability of various proteins: Insights into protein-buffer interactions. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
156
|
Heymich ML, Srirangan S, Pischetsrieder M. Stability and Activity of the Antimicrobial Peptide Leg1 in Solution and on Meat and Its Optimized Generation from Chickpea Storage Protein. Foods 2021; 10:foods10061192. [PMID: 34070446 PMCID: PMC8227015 DOI: 10.3390/foods10061192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
The antimicrobial peptide Leg1 (RIKTVTSFDLPALRFLKL) from chickpea legumin is active against spoilage bacteria, yeast, and mold. The present study tested its effectiveness under food storage conditions and examined options to obtain a food-grade agent. The minimum inhibitory concentration (MIC) of Leg1 against E. coli (62.5 µM) proved stable over seven days at 20 °C or 4 °C. It was not influenced by reduced pH (5.0 vs. 6.8), which is relevant in food such as meat. An incubation temperature of 20 °C vs. 37 °C reduced the MIC to 15.6/7.8 µM against E. coli/B. subtilis. With a minimum bactericidal concentration in meat of 125/15.6 µM against E. coli/B. subtilis, Leg1 is equivalently effective as nisin and 5000–82,000 times more active than sodium benzoate, potassium sorbate, or sodium nitrite. Replacing the counter-ion trifluoroacetate derived from peptide synthesis by the more natural alternatives acetate or chloride did not impair the activity of Leg1. As an alternative to chemical synthesis, an optimized protocol for chymotryptic hydrolysis was developed, increasing the yield from chickpea legumin by a factor of 30 compared to the standard procedure. The present results indicate that food-grade Leg1 could possibly be applicable for food preservation.
Collapse
|
157
|
Wätzig H, Hoffstedt M, Krebs F, Minkner R, Scheller C, Zagst H. Protein analysis and stability: Overcoming trial-and-error by grouping according to physicochemical properties. J Chromatogr A 2021; 1649:462234. [PMID: 34038775 DOI: 10.1016/j.chroma.2021.462234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
Today proteins are possibly the most important class of substances. Yet new tasks for proteins are still often solved by trial-and-error approaches. However, in some areas these euphemistically called "screening approaches" are not suitable. E.g. stability tests just take too long and therefore require a more strategic, target-orientated concept. This concept is available by grouping proteins according to their physicochemical properties and then pulling out the right drawer for new tasks. These properties include size, then charge and hydrophobicity as well as their patchinesses, and the degree of order. In addition, solubility, the content of (free) enthalpy, aromatic-amino-acid- and α/β-frequency as well as helix capping, and corresponding patchiness, the number of specific motifs and domains as well as the typical concentration range can be helpful to discriminate between different groups of proteins. Analyzing correlations will reduce the necessary amount of parameters and additional ones, which may be still undiscovered at the present time, can be identified looking at protein subgroups with similar physicochemical properties which still behave heterogeneously. Step-by-step the methodology will be improved. Possibly protein stability will be the driver of this process, but all other areas such as production, purification and analytics including sample pre-treatment and the choice of appropriate separation conditions for e.g. chromatography and electrophoresis will profit from a rational strategy.
Collapse
Affiliation(s)
- Hermann Wätzig
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany.
| | - Marc Hoffstedt
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany
| | - Finja Krebs
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany
| | - Robert Minkner
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany
| | - Christin Scheller
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany
| | - Holger Zagst
- Technische Universität Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstraße 55, Braunschweig 38106, Germany
| |
Collapse
|
158
|
Khoeini D, Scott TF, Neild A. Microfluidic enhancement of self-assembly systems. LAB ON A CHIP 2021; 21:1661-1675. [PMID: 33949588 DOI: 10.1039/d1lc00038a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Dynamic, kinetically-controlled, self-assembly processes are commonly observed in nature and are capable of creating intricate, functional architectures from simple precursors. However, notably, much of the research into molecular self-assembly has been performed using conventional bulk techniques where the resultant species are dictated by thermodynamic stability to yield relatively simple assemblies. Whereas, the environmental control offered by microfluidic systems offers methods to achieve non-equilibrium reaction conditions capable of increasingly sophisticated self-assembled structures. Alterations to the immediate microenvironment during the assembly of the molecules is possible, providing the basis for kinetically-controlled assembly. This review examines the key mechanism offered by microfluidic systems and the architectures required to access them. The mechanisms include diffusion-led mixing, shear gradient alignment, spatial and temporal confinement, and structural templates in multiphase systems. The works are selected and categorised in terms of the microfluidic approaches taken rather than the chemical constructs which are formed.
Collapse
Affiliation(s)
- Davood Khoeini
- Laboratory for Micro Systems, Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Timothy F Scott
- Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia and Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Adrian Neild
- Laboratory for Micro Systems, Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
159
|
Zhang C, Codina N, Tang J, Yu H, Chakroun N, Kozielski F, Dalby PA. Comparison of the pH- and thermally-induced fluctuations of a therapeutic antibody Fab fragment by molecular dynamics simulation. Comput Struct Biotechnol J 2021; 19:2726-2741. [PMID: 34093988 PMCID: PMC8131956 DOI: 10.1016/j.csbj.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 05/01/2021] [Indexed: 11/27/2022] Open
Abstract
Successful development of protein therapeutics depends critically on achieving stability under a range of conditions. A deeper understanding of the drivers of instability across different stress conditions, will enable the engineering of more robust protein scaffolds. We compared the impacts of low pH and high temperature stresses on the structure of a humanized antibody fragment (Fab) A33, using atomistic molecular dynamics simulations, using a recent 2.5 Å crystal structure. This revealed that low-pH induced the loss of native contacts in the domain CL. By contrast, thermal stress led to 5-7% loss of native contacts in all four domains, and simultaneous loss of >30% of native contacts in the VL-VH and CL-CH interfaces. This revealed divergent destabilising pathways under the two different stresses. The underlying cause of instability was probed using FoldX and Rosetta mutation analysis, and packing density calculations. These agreed that mutations in the CL domain, and CL-CH1 interface have the greatest potential for stabilisation of Fab A33. Several key salt bridge losses underpinned the conformational change in CL at low pH, whereas at high temperature, salt bridges became more dynamic, thus contributing to an overall destabilization. Lastly, the unfolding events at the two stress conditions exposed different predicted aggregation-prone regions (APR) to solvent, which would potentially lead to different aggregation mechanisms. Overall, our results identified the early stages of unfolding and stability-limiting regions of Fab A33, and the VH and CL domains as interesting future targets for engineering stability to both pH- and thermal-stresses simultaneously.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Nuria Codina
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Jiazhi Tang
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Haoran Yu
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, United Kingdom
| | - Nesrine Chakroun
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Frank Kozielski
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| |
Collapse
|
160
|
Chen Y, Mutukuri TT, Wilson NE, Zhou QT. Pharmaceutical protein solids: Drying technology, solid-state characterization and stability. Adv Drug Deliv Rev 2021; 172:211-233. [PMID: 33705880 PMCID: PMC8107147 DOI: 10.1016/j.addr.2021.02.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 02/22/2021] [Indexed: 01/30/2023]
Abstract
Despite the boom in biologics over the past decade, the intrinsic instability of these large molecules poses significant challenges to formulation development. Almost half of all pharmaceutical protein products are formulated in the solid form to preserve protein native structure and extend product shelf-life. In this review, both traditional and emerging drying techniques for producing protein solids will be discussed. During the drying process, various stresses can impact the stability of protein solids. However, understanding the impact of stress on protein product quality can be challenging due to the lack of reliable characterization techniques for biological solids. Both conventional and advanced characterization techniques are discussed including differential scanning calorimetry (DSC), solid-state Fourier transform infrared spectrometry (ssFTIR), solid-state fluorescence spectrometry, solid-state hydrogen deuterium exchange (ssHDX), solid-state nuclear magnetic resonance (ssNMR) and solid-state photolytic labeling (ssPL). Advanced characterization tools may offer mechanistic investigations into local structural changes and interactions at higher resolutions. The continuous exploration of new drying techniques, as well as a better understanding of the effects caused by different drying techniques in solid state, would advance the formulation development of biological products with superior quality.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Tarun Tejasvi Mutukuri
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Nathan E Wilson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
161
|
Novel formulations and drug delivery systems to administer biological solids. Adv Drug Deliv Rev 2021; 172:183-210. [PMID: 33705873 DOI: 10.1016/j.addr.2021.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in formulation sciences have expanded the previously limited design space for biological modalities, including peptide, protein, and vaccine products. At the same time, the discovery and application of new modalities, such as cellular therapies and gene therapies, have presented formidable challenges to formulation scientists. We explore these challenges and highlight the opportunities to overcome them through the development of novel formulations and drug delivery systems as biological solids. We review the current progress in both industry and academic laboratories, and we provide expert perspectives in those settings. Formulation scientists have made a tremendous effort to accommodate the needs of these novel delivery routes. These include stability-preserving formulations and dehydration processes as well as dosing regimes and dosage forms that improve patient compliance.
Collapse
|
162
|
Schuster J, Mahler HC, Joerg S, Huwyler J, Mathaes R. Analytical Challenges Assessing Protein Aggregation and Fragmentation Under Physiologic Conditions. J Pharm Sci 2021; 110:3103-3110. [PMID: 33933436 DOI: 10.1016/j.xphs.2021.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/01/2021] [Accepted: 04/09/2021] [Indexed: 12/17/2022]
Abstract
Therapeutic proteins are administered by injection or infusion. After administration, the physiologic environment in the desired body compartment - fluid or tissue - can impact protein stability and lead to changes in the safety and/or efficacy profile. For example, protein aggregation and fragmentation are critical quality attributes of the drug product and can occur after administration to patients. In this context, the in vivo stability of therapeutic proteins has gained increasing attention. However, in vivo protein aggregation and fragmentation are difficult to assess and have been rarely investigated. This mini-review summarizes analytical approaches to assess the stability of therapeutic proteins using simulated physiologic conditions. Furthermore, we discuss factors potentially causing in vivo protein aggregation, precipitation, and fragmentation in complex biological fluids. Different analytical approaches are evaluated with respect to their applicability and possible shortcomings when it comes to these degradation events in biological fluids. Tracking protein stability in biological fluids typically requires purifying or labeling the protein of interest to circumvent matrix interference of biological fluids. Improved analytical methods are strongly needed to gain knowledge on in vivo protein aggregation and fragmentation. In vitro models can support the selection of lead candidates and accelerate the pre-clinical development of therapeutic proteins.
Collapse
Affiliation(s)
- Joachim Schuster
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland; University of Basel, Pharmacenter, Division of Pharmaceutical Technology, Basel, Switzerland
| | | | - Susanne Joerg
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland
| | - Joerg Huwyler
- University of Basel, Pharmacenter, Division of Pharmaceutical Technology, Basel, Switzerland
| | - Roman Mathaes
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland.
| |
Collapse
|
163
|
Chandrashekar C, Hossain MA, Wade JD. Chemical Glycosylation and Its Application to Glucose Homeostasis-Regulating Peptides. Front Chem 2021; 9:650025. [PMID: 33912539 PMCID: PMC8072350 DOI: 10.3389/fchem.2021.650025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Peptides and proteins are attractive targets for therapeutic drug development due to their exquisite target specificity and low toxicity profiles. However, their complex structures give rise to several challenges including solubility, stability, aggregation, low bioavailability, and poor pharmacokinetics. Numerous chemical strategies to address these have been developed including the introduction of several natural and non-natural modifications such as glycosylation, lipidation, cyclization and PEGylation. Glycosylation is considered to be one of the most useful modifications as it is known to contribute to increasing the stability, to improve solubility, and increase the circulating half-lifves of these biomolecules. However, cellular glycosylation is a highly complex process that generally results in heterogenous glycan structures which confounds quality control and chemical and biological assays. For this reason, much effort has been expended on the development of chemical methods, including by solid phase peptide synthesis or chemoenzymatic processes, to enable the acquisition of homogenous glycopeptides to greatly expand possibilities in drug development. In this mini-review, we highlight the importance of such chemical glycosylation methods for improving the biophysical properties of naturally non-glycosylated peptides as applied to the therapeutically essential insulin and related peptides that are used in the treatment of diabetes.
Collapse
Affiliation(s)
- Chaitra Chandrashekar
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Mohammed Akhter Hossain
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - John D Wade
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,School of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
164
|
Bolje A, Gobec S. Analytical Techniques for Structural Characterization of Proteins in Solid Pharmaceutical Forms: An Overview. Pharmaceutics 2021; 13:pharmaceutics13040534. [PMID: 33920461 PMCID: PMC8070348 DOI: 10.3390/pharmaceutics13040534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/21/2021] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
Therapeutic proteins as biopharmaceuticals have emerged as a very important class of drugs for the treatment of many diseases. However, they are less stable compared to conventional pharmaceuticals. Their long-term stability in solid forms, which is critical for product performance, depends heavily on the retention of the native protein structure during the lyophilization (freeze-drying) process and, thereafter, in the solid state. Indeed, the biological function of proteins is directly related to the tertiary and secondary structure. Besides physical stability and biological activity, conformational stability (three-dimensional structure) is another important aspect when dealing with protein pharmaceuticals. Moreover, denaturation as loss of higher order structure is often a precursor to aggregation or chemical instability. Careful study of the physical and chemical properties of proteins in the dried state is therefore critical during biopharmaceutical drug development to deliver a final drug product with built-in quality that is safe, high-quality, efficient, and affordable for patients. This review provides an overview of common analytical techniques suitable for characterizing pharmaceutical protein powders, providing structural, and conformational information, as well as insights into dynamics. Such information can be very useful in formulation development, where selecting the best formulation for the drug can be quite a challenge.
Collapse
Affiliation(s)
- Aljoša Bolje
- Correspondence: (A.B.); (S.G.); Tel.: +386-147-69500 (A.B.); +386-147-69585 (S.G.)
| | - Stanislav Gobec
- Correspondence: (A.B.); (S.G.); Tel.: +386-147-69500 (A.B.); +386-147-69585 (S.G.)
| |
Collapse
|
165
|
Markwalter CE, Pagels RF, Hejazi AN, Ristroph KD, Wang J, Chen K, Li J, Prud'homme RK. Sustained release of peptides and proteins from polymeric nanocarriers produced by inverse Flash NanoPrecipitation. J Control Release 2021; 334:11-20. [PMID: 33823220 DOI: 10.1016/j.jconrel.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/18/2022]
Abstract
Peptide and protein therapeutics generally exhibit high potency and specificity and are increasingly important segments of the pharmaceutical market. However, their clinical applications are limited by rapid clearance and poor membrane permeability. Encapsulation of the peptide or protein into a nano-scale carrier can modify its pharmacokinetics and biodistribution. This might be employed to promote uptake in desired cell types or tissues, to limit systemic exposure, or to reduce the need for frequent injections. We have recently described inverse Flash NanoPrecipitation (iFNP), a scalable technique to encapsulate water-soluble therapeutics into polymeric nanocarriers, and have demonstrated improvements in therapeutic loading of an order of magnitude over comparable approaches. Here, we describe the formulation parameters that control release rates of encapsulated model therapeutics polymyxin B, lysozyme, and bovine serum albumin from nanocarriers produced using iFNP. Using a neutropenic lung infection mouse model with a multi-drug resistant Acinetobacter baumannii clinical isolate, we demonstrate enhanced therapeutic effect and safety profile afforded by nanocarrier-encapsulated polymyxin B following pulmonary administration. The encapsulated formulation reduced toxicity observed at elevated doses and resulted in up to 2.7-log10 reduction in bacterial burden below that of unencapsulated polymyxin B. These results establish the promise of iFNP as a platform for nanocarrier delivery of water-soluble therapeutics.
Collapse
Affiliation(s)
- Chester E Markwalter
- Dept of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Robert F Pagels
- Dept of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Ava N Hejazi
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Kurt D Ristroph
- Dept of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Jiping Wang
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Ke Chen
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Robert K Prud'homme
- Dept of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
166
|
Souza ALRD, Amorim ACF, Cintra ER, Ferreira NN, Silva LAD, Hayasaki TG, Diniz DGA, Lima EM. Development and validation of a rapid RP-HPLC method for simultaneous quantification of paclitaxel and cetuximab in immunoliposomes. Talanta 2021; 225:121988. [PMID: 33592736 DOI: 10.1016/j.talanta.2020.121988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 11/18/2022]
Abstract
The development of rational therapies against complex diseases, such as cancer, has increased in the past few years due to the advances of 'omics' technologies. Concomitantly, several efforts have been made to design sophisticated drug delivery systems in order to increase specificity and drug accumulation in tumor sites. The complexity of these drug delivery systems highlights the need for suitable analytical methods to determine encapsulation/conjugation efficiency of drugs and molecules responsible for the targeted delivery. Therefore, this study focuses on the development and validation of a RP-HPLC-DAD methodology for concurrent quantification of paclitaxel (PTX) and cetuximab (CTX) in immunoliposomes. Chromatographic separation was achieved using a wide pore C8 column, and a gradient mobile phase consisting of 0.1% trifluoroacetic acid (TFA) in Milli-Q water/acetonitrile/isopropanol with a flow rate of 1 mL min-1. Drug peaks were fully separated and detected at 280 nm using UV detector. The method was validated according to ICH and FDA guidelines in terms of specificity and forced degradation studies, system suitability, linearity, limit of detection, limit of quantification, repeatability, intermediate precision, accuracy, robustness, and short-term stability. The developed method was linear over the concentration range of 37.5-150 μg mL-1 of PTX and 75-300 μg mL-1 of CTX. All parameters evaluated satisfied the acceptance criteria, according to both FDA and ICH guidelines. The applicability of the analytical method was assessed following the development of PTX-loaded immunoliposomes conjugated with CTX. Thus, the present study shows a novel, simple, stability-indicating and suitable method to quantify simultaneously PTX and CTX in immunoliposomes.
Collapse
Affiliation(s)
- Ana Luiza Ribeiro de Souza
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Amanda Cláudia Ferreira Amorim
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Emílio Ramos Cintra
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | | | - Luís Antônio Dantas Silva
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Tacio Gonçalves Hayasaki
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Danielle Guimarães Almeida Diniz
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Eliana Martins Lima
- Laboratory of Pharmaceutical Nanotechnology and Drug Delivery Systems - FarmaTec, School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
167
|
Lenton S, Hervø-Hansen S, Popov AM, Tully MD, Lund M, Skepö M. Impact of Arginine-Phosphate Interactions on the Reentrant Condensation of Disordered Proteins. Biomacromolecules 2021; 22:1532-1544. [PMID: 33730849 PMCID: PMC8045028 DOI: 10.1021/acs.biomac.0c01765] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Re-entrant condensation results in the formation of a condensed protein regime between two critical ion concentrations. The process is driven by neutralization and inversion of the protein charge by oppositely charged ions. Re-entrant condensation of cationic proteins by the polyvalent anions, pyrophosphate and tripolyphosphate, has previously been observed, but not for citrate, which has similar charge and size compared to the polyphosphates. Therefore, besides electrostatic interactions, other specific interactions between the polyphosphate ions and proteins must contribute. Here, we show that additional attractive interactions between arginine and tripolyphosphate determine the re-entrant condensation and decondensation boundaries of the cationic, intrinsically disordered saliva protein, histatin 5. Furthermore, we show by small-angle X-ray scattering (SAXS) that polyvalent anions cause compaction of histatin 5, as would be expected based solely on electrostatic interactions. Hence, we conclude that arginine-phosphate-specific interactions not only regulate solution properties but also influence the conformational ensemble of histatin 5, which is shown to vary with the number of arginine residues. Together, the results presented here provide further insight into an organizational mechanism that can be used to tune protein interactions in solution of both naturally occurring and synthetic proteins.
Collapse
Affiliation(s)
- Samuel Lenton
- Theoretical Chemistry, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | - Stefan Hervø-Hansen
- Theoretical Chemistry, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | - Anton M Popov
- BM29 BIOSAXS, European Synchroton Radiation Facility, 71 avenue des Martyrs, Grenoble, Isère 38043, France
| | - Mark D Tully
- BM29 BIOSAXS, European Synchroton Radiation Facility, 71 avenue des Martyrs, Grenoble, Isère 38043, France
| | - Mikael Lund
- Theoretical Chemistry, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden.,LINXS-Lund Institute of Advanced Neutron and X-ray Science, Scheelevägen 19, SE-223 70 Lund, Sweden
| | - Marie Skepö
- Theoretical Chemistry, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden.,LINXS-Lund Institute of Advanced Neutron and X-ray Science, Scheelevägen 19, SE-223 70 Lund, Sweden
| |
Collapse
|
168
|
ElKassas K, Chullipalliyalil K, McAuliffe M, Vucen S, Crean A. Fluorescence spectroscopy for the determination of reconstitution time of an in-vial lyophilised product. Int J Pharm 2021; 597:120368. [PMID: 33561500 DOI: 10.1016/j.ijpharm.2021.120368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022]
Abstract
Lyophilisation is a prominent technique used to create stabilised, dried forms of biopharmaceutical formulations. Reconstitution of lyophilised parenteral formulations is a key step prior to patient administration. The accurate determination of reconstitution time is a necessity to aid formulation development and support product quality control. Traditional methods for quantifying reconstitution time involve the visual identification of the endpoint, which has led to variable values reported across studies. In this work, the use of ultra-violet (UV) excited fluorescence spectroscopy as an alternative to the visual quantification of the reconstitution time was investigated. Spectrographic information was collected via a bespoke setup that allowed the measurement of the reconstitution time in a standard sealed lyophilisation vial. The spectra were analysed via principal component analysis (PCA) to obtain a time-based representation of the changes in a reconstituting formulation. The analysis was followed by the identification of an endpoint using three techniques ranging from fully automated to manual with regards to the required level of user input. At high protein concentration, the variability of the reconstitution time measurements was reduced from 80.4% relative standard deviation obtained via the traditional method to 8.2% for the instrumental method presented in.
Collapse
Affiliation(s)
- Khaled ElKassas
- SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, T12 YT20, Ireland
| | | | - Michael McAuliffe
- Centre for Advanced Photonics & Process Analysis, Munster Technological University Cork, T12P928, Ireland
| | - Sonja Vucen
- SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, T12 YT20, Ireland
| | - Abina Crean
- SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, T12 YT20, Ireland
| |
Collapse
|
169
|
Zhang ZC, Raab SA, Hales DA, Clemmer DE. Influence of Solvents upon Diketopiperazine Formation of FPG8K. J Phys Chem B 2021; 125:2952-2959. [DOI: 10.1021/acs.jpcb.1c00269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhi-chao Zhang
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, Indiana 47401, United States
| | - Shannon A. Raab
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, Indiana 47401, United States
| | - David A. Hales
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, Indiana 47401, United States
- Department of Chemistry, Hendrix College, Conway, Arkansas 72032, United States
| | - David E. Clemmer
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, Indiana 47401, United States
| |
Collapse
|
170
|
Patient-centric design for peptide delivery: Trends in routes of administration and advancement in drug delivery technologies. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2020.100079] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
171
|
Santra S, Dhurua S, Jana M. Analyzing the driving forces of insulin stability in the basic amino acid solutions: A perspective from hydration dynamics. J Chem Phys 2021; 154:084901. [PMID: 33639734 DOI: 10.1063/5.0038305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Amino acids having basic side chains, as additives, are known to increase the stability of native-folded state of proteins, but their relative efficiency and the molecular mechanism are still controversial and obscure as well. In the present work, extensive atomistic molecular dynamics simulations were performed to investigate the hydration properties of aqueous solutions of concentrated arginine, histidine, and lysine and their comparative efficiency on regulating the conformational stability of the insulin monomer. We identified that in the aqueous solutions of the free amino acids, the nonuniform relaxation of amino acid-water hydrogen bonds was due to the entrapment of water molecules within the amino acid clusters formed in solutions. Insulin, when tested with these solutions, was found to show rigid conformations, relative to that in pure water. We observed that while the salt bridges formed by the lysine as an additive contributed more toward the direct interactions with insulin, the cation-π was more prominent for the insulin-arginine interactions. Importantly, it was observed that the preferentially more excluded arginine, compared to histidine and lysine from the insulin surface, enriches the hydration layer of the protein. Our study reveals that the loss of configurational entropy of insulin in arginine solution, as compared to that in pure water, is more as compared to the entropy loss in the other two amino acid solutions, which, moreover, was found to be due to the presence of motionally bound less entropic hydration water of insulin in arginine solution than in histidine or lysine solution.
Collapse
Affiliation(s)
- Santanu Santra
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela 769008, India
| | - Shakuntala Dhurua
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela 769008, India
| | - Madhurima Jana
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela 769008, India
| |
Collapse
|
172
|
Abstract
Monoclonal antibodies are proteinaceous in nature and are subject to instability issues. Stability testing of monoclonal antibodies is a critical regulatory requirement in their development and commercialization as therapeutic biological molecules. This article reviews the numerous drug manufacturing processes such as: upstream processing, downstream purification and aseptic filling along with physical and chemical factors such as protein concentration, structure, pH, temperature, light, agitation, deamidation, oxidation, glycation leading to instabilities in monoclonal antibodies and it spotlights the variety of analytical techniques employed to investigate and generate information on stability studies and henceforth, helps in developing the stability-indicating methods. In addition, this paper aims to discuss the ICH regulatory guideline (s) for the stability assessment of biological products (Drug Substance and Drug Product).
Collapse
Affiliation(s)
- Harleen Kaur
- Analytical Sciences, Aurobindo Biologics, Hyderabad, India
| |
Collapse
|
173
|
Effect of Concentrated Salts Solutions on the Stability of Immobilized Enzymes: Influence of Inactivation Conditions and Immobilization Protocol. Molecules 2021; 26:molecules26040968. [PMID: 33673063 PMCID: PMC7918437 DOI: 10.3390/molecules26040968] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
This paper aims to investigate the effects of some salts (NaCl, (NH4)2SO4 and Na2SO4) at pH 5.0, 7.0 and 9.0 on the stability of 13 different immobilized enzymes: five lipases, three proteases, two glycosidases, and one laccase, penicillin G acylase and catalase. The enzymes were immobilized to prevent their aggregation. Lipases were immobilized via interfacial activation on octyl agarose or on glutaraldehyde-amino agarose beads, proteases on glyoxyl agarose or glutaraldehyde-amino agarose beads. The use of high concentrations of salts usually has some effects on enzyme stability, but the intensity and nature of these effects depends on the inactivation pH, nature and concentration of the salt, enzyme and immobilization protocol. The same salt can be a stabilizing or a destabilizing agent for a specific enzyme depending on its concentration, inactivation pH and immobilization protocol. Using lipases, (NH4)2SO4 generally permits the highest stabilities (although this is not a universal rule), but using the other enzymes this salt is in many instances a destabilizing agent. At pH 9.0, it is more likely to find a salt destabilizing effect than at pH 7.0. Results confirm the difficulty of foreseeing the effect of high concentrations of salts in a specific immobilized enzyme.
Collapse
|
174
|
Erfani A, Zarrintaj P, Seaberg J, Ramsey JD, Aichele CP. Zwitterionic poly(carboxybetaine) microgels for enzyme (chymotrypsin) covalent immobilization with extended stability and activity. J Appl Polym Sci 2021. [DOI: 10.1002/app.50545] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Amir Erfani
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma USA
| | - Payam Zarrintaj
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma USA
| | - Joshua Seaberg
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma USA
| | - Joshua D. Ramsey
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma USA
| | - Clint P. Aichele
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma USA
| |
Collapse
|
175
|
Meyer RM, Berger L, Nerkamp J, Scheler S, Nehring S, Friess W. Identification of monoclonal antibody variants involved in aggregate formation - Part 2: Hydrophobicity variants. Eur J Pharm Biopharm 2021; 160:134-142. [PMID: 33524536 DOI: 10.1016/j.ejpb.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 11/29/2022]
Abstract
Monoclonal antibodies (mAbs) are valuable tools both in therapy and in diagnostic. Their tendency to aggregate is a serious concern. Since a mAb drug substance (DS) is composed of different variants, it is important for manufacturers to know the behavior and stability not only of the mAb as a whole, but also of the variants contained in the product. We present a method to separate hydrophobicity variants of a mAb and subsequently analyzed these variants for stability and aggregation propensity. We identified a potentially aggregation prone hydrophilic variant which is interrelated with another previously identified aggregation prone acidic charge variant. Additionally, we assessed the risk posed by the aggregation prone variant to the DS by spiking hydrophobicity variants into DS and did not observe an enhanced aggregation propensity. Thus we present an approach to separate, characterize and analyze the criticality of aggregation prone variants in protein DS which is a step forward to further assure drug safety.
Collapse
Affiliation(s)
- Robina M Meyer
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, University of Munich, Butenandtstr. 5, 81377 Munich, Germany
| | - Lukas Berger
- Sandoz Biopharmaceutics, Biochemiestr. 10, 6336 Langkampfen, Austria
| | - Joerg Nerkamp
- Sandoz Biopharmaceutics, Biochemiestr. 10, 6336 Langkampfen, Austria
| | - Stefan Scheler
- Sandoz Biopharmaceutics, Biochemiestr. 10, 6336 Langkampfen, Austria
| | - Sebastian Nehring
- Sandoz Biopharmaceutics, Biochemiestr. 10, 6336 Langkampfen, Austria
| | - Wolfgang Friess
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, University of Munich, Butenandtstr. 5, 81377 Munich, Germany.
| |
Collapse
|
176
|
Economidou SN, Pissinato Pere CP, Okereke M, Douroumis D. Optimisation of Design and Manufacturing Parameters of 3D Printed Solid Microneedles for Improved Strength, Sharpness, and Drug Delivery. MICROMACHINES 2021; 12:mi12020117. [PMID: 33499301 PMCID: PMC7912255 DOI: 10.3390/mi12020117] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/20/2022]
Abstract
3D printing has emerged as a powerful manufacturing technology and has attracted significant attention for the fabrication of microneedle (MN)-mediated transdermal systems. In this work, we describe an optimisation strategy for 3D-printed MNs, ranging from the design to the drug delivery stage. The key relationships between design and manufacturing parameters and quality and performance are systematically explored. The printing and post-printing set parameters were found to influence quality and material mechanical properties, respectively. It was demonstrated that the MN geometry affected piercing behaviour, fracture, and coating morphology. The delivery of insulin in porcine skin by inkjet-coated MNs was shown to be influenced by MN design.
Collapse
Affiliation(s)
- Sophia N. Economidou
- Medway School of Pharmacy, University of Kent, Medway Campus, Central Avenue, Chatham Maritime, Chatham, Kent ME4 4TB, UK;
- Correspondence: (S.N.E.); (D.D.)
| | - Cristiane P. Pissinato Pere
- Medway School of Pharmacy, University of Kent, Medway Campus, Central Avenue, Chatham Maritime, Chatham, Kent ME4 4TB, UK;
| | - Michael Okereke
- Department of Engineering Science, University of Greenwich, Kent ME4 4TB, UK;
| | - Dennis Douroumis
- CIPER Centre for Innovation and Process Engineering Research, Kent ME4 4TB, UK
- Correspondence: (S.N.E.); (D.D.)
| |
Collapse
|
177
|
Khodabakhsh F, Salimian M, Hedayati MH, Ahangari Cohan R, Norouzian D. Challenges and advancements in the pharmacokinetic enhancement of therapeutic proteins. Prep Biochem Biotechnol 2021; 51:519-529. [PMID: 33459157 DOI: 10.1080/10826068.2020.1839907] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nowadays, proteins are frequently administered as therapeutic agents in human diseases. However, the main challenge regarding the clinical application of therapeutic proteins is short circulating plasma half-life that leads to more frequent injections for maintaining therapeutic plasma levels, increased therapy costs, immunogenic reactions, and low patient compliance. So, the development of novel strategies to enhance the pharmacokinetic profile of therapeutic proteins has attracted great attention in pharmaceuticals. So far, several techniques, each with their pros and cons, have been developed including chemical bonding to polymers, hyper glycosylation, Fc fusion, human serum albumin fusion, and recombinant PEG mimetics. These techniques mainly classify into three strategies; (i) the endosomal recycling of neonatal Fc receptor which is observed for immunoglobulins and albumin, (ii) decrease in receptor-mediated clearance, and (iii) increase in hydrodynamic radius through chemical and genetic modifications. Recently, novel PEG mimetic peptides like proline/alanine/serine repeat sequences are designed to overcome pitfalls associated with the previous technologies. Biodegradability, lack of or low immunogenicity, product homogeneity, and a simple production process, currently make these polypeptides as the preferred technology for plasma half-life extension of therapeutic proteins. In this review, challenges and pitfalls in the pharmacokinetic enhancement of therapeutic proteins using PEG-mimetic peptides will be discussed in detail.
Collapse
Affiliation(s)
- Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Morteza Salimian
- Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Hedayati
- Department of Quality Control, Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Dariush Norouzian
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
178
|
Guckeisen T, Hosseinpour S, Peukert W. Effect of pH and urea on the proteins secondary structure at the water/air interface and in solution. J Colloid Interface Sci 2021; 590:38-49. [PMID: 33524719 DOI: 10.1016/j.jcis.2021.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 01/09/2023]
Abstract
HYPOTHESIS The secondary structure of proteins affects their functionality and performance in physiological environments or industrial applications. Change of the solution pH or the presence of protein denaturants are the main chemical means that can alter the secondary structure of proteins or lead to protein denaturation. Since proteins in the bulk solution and those residing at the solution/air interface experience different local environments, their response to chemical denaturation can be different. EXPERIMENTS We utilize circular dichroism and chiral/achiral sum frequency generation spectroscopy to study the secondary structure of selected proteins as a function of the solution pH or in the presence of 8 M urea in the bulk solution and at the solution/air interface, respectively. FINDINGS The liquid/air interface can enhance or decrease protein conformation stability. The change in the secondary structure of the surface adsorbed proteins in alkaline solutions occurs at pH values lower than those denaturing the studied proteins in the bulk solution. In contrast, while 8 M urea completely denatures the studied proteins in the bulk solution, the liquid/air interface prevents the urea-induced denaturation of the surface adsorbed proteins by limiting the access of urea to the hydrophobic side chains of proteins protruding to air.
Collapse
Affiliation(s)
- Tobias Guckeisen
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| | - Saman Hosseinpour
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| | - Wolfgang Peukert
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| |
Collapse
|
179
|
Mi F, Guan M, Hu C, Peng F, Sun S, Wang X. Application of lectin-based biosensor technology in the detection of foodborne pathogenic bacteria: a review. Analyst 2021; 146:429-443. [DOI: 10.1039/d0an01459a] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Foodborne diseases caused by pathogenic bacteria pose a serious threat to human health.
Collapse
Affiliation(s)
- Fang Mi
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
- Xinjiang bingtuan Xingxin Vocational and Technical College
| | - Ming Guan
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
| | - Cunming Hu
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
| | - Fei Peng
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
| | - Shijiao Sun
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
| | - Xiaomei Wang
- College of Chemistry and Chemical Engineering
- Xinjiang normal University
- Urumqi
- China
| |
Collapse
|
180
|
Meyer RM, Berger L, Nerkamp J, Scheler S, Nehring S, Friess W. Identification of monoclonal antibody variants involved in aggregate formation – Part 1: Charge variants. Eur J Pharm Biopharm 2021; 158:123-131. [DOI: 10.1016/j.ejpb.2020.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 11/27/2022]
|
181
|
Rahman MS, Roy R, Jadhav B, Hossain MN, Halim MA, Raynie DE. Formulation, structure, and applications of therapeutic and amino acid-based deep eutectic solvents: An overview. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114745] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
182
|
Sadiki A, Vaidya SR, Abdollahi M, Bhardwaj G, Dolan ME, Turna H, Arora V, Sanjeev A, Robinson TD, Koid A, Amin A, Zhou ZS. Site-specific conjugation of native antibody. Antib Ther 2020; 3:271-284. [PMID: 33644685 PMCID: PMC7906296 DOI: 10.1093/abt/tbaa027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traditionally, non-specific chemical conjugations, such as acylation of amines on lysine or alkylation of thiols on cysteines, are widely used; however, they have several shortcomings. First, the lack of site-specificity results in heterogeneous products and irreproducible processes. Second, potential modifications near the complementarity-determining region may reduce binding affinity and specificity. Conversely, site-specific methods produce well-defined and more homogenous antibody conjugates, ensuring developability and clinical applications. Moreover, several recent side-by-side comparisons of site-specific and stochastic methods have demonstrated that site-specific approaches are more likely to achieve their desired properties and functions, such as increased plasma stability, less variability in dose-dependent studies (particularly at low concentrations), enhanced binding efficiency, as well as increased tumor uptake. Herein, we review several standard and practical site-specific bioconjugation methods for native antibodies, i.e., those without recombinant engineering. First, chemo-enzymatic techniques, namely transglutaminase (TGase)-mediated transamidation of a conserved glutamine residue and glycan remodeling of a conserved asparagine N-glycan (GlyCLICK), both in the Fc region. Second, chemical approaches such as selective reduction of disulfides (ThioBridge) and N-terminal amine modifications. Furthermore, we list site-specific antibody–drug conjugates in clinical trials along with the future perspectives of these site-specific methods.
Collapse
Affiliation(s)
- Amissi Sadiki
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Shefali R Vaidya
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Mina Abdollahi
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Gunjan Bhardwaj
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Michael E Dolan
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA.,Downstream Development, Biologics Process Development, Millennium Pharmaceuticals, Inc., (a wholly-owned subsidiary of Takeda Pharmaceuticals Company Limited), Cambridge, Massachusetts 02139, USA
| | - Harpreet Turna
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Varnika Arora
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Athul Sanjeev
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Timothy D Robinson
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Andrea Koid
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Aashka Amin
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Zhaohui Sunny Zhou
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| |
Collapse
|
183
|
Cotrina EY, Vilà M, Nieto J, Arsequell G, Planas A. Preparative Scale Production of Recombinant Human Transthyretin for Biophysical Studies of Protein-Ligand and Protein-Protein Interactions. Int J Mol Sci 2020; 21:ijms21249640. [PMID: 33348885 PMCID: PMC7766448 DOI: 10.3390/ijms21249640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
Human transthyretin (hTTR), a serum protein with a main role in transporting thyroid hormones and retinol through binding to the retinol-binding protein, is an amyloidogenic protein involved in familial amyloidotic polyneuropathy (FAP), familial amyloidotic cardiomyopathy, and central nervous system selective amyloidosis. hTTR also has a neuroprotective role in Alzheimer disease, being the major Aβ binding protein in human cerebrospinal fluid (CSF) that prevents amyloid-β (Aβ) aggregation with consequent abrogation of toxicity. Here we report an optimized preparative expression and purification protocol of hTTR (wt and amyloidogenic mutants) for in vitro screening assays of TTR ligands acting as amyloidogenesis inhibitors or acting as molecular chaperones to enhance the TTR:Aβ interaction. Preparative yields were up to 660 mg of homogenous protein per L of culture in fed-batch bioreactor. The recombinant wt protein is mainly unmodified at Cys10, the single cysteine in the protein sequence, whereas the highly amyloidogenic Y78F variant renders mainly the S-glutathionated form, which has essentially the same amyloidogenic behavior than the reduced protein with free Cys10. The TTR production protocol has shown inter-batch reproducibility of expression and protein quality for in vitro screening assays.
Collapse
Affiliation(s)
- Ellen Y. Cotrina
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.Y.C.); (M.V.); (J.N.)
- Institut de Química Avançada de Catalunya, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), 08034 Barcelona, Spain;
| | - Marta Vilà
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.Y.C.); (M.V.); (J.N.)
| | - Joan Nieto
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.Y.C.); (M.V.); (J.N.)
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), 08034 Barcelona, Spain;
| | - Antoni Planas
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.Y.C.); (M.V.); (J.N.)
- Correspondence:
| |
Collapse
|
184
|
Mutukuri TT, Wilson NE, Taylor LS, Topp EM, Zhou QT. Effects of drying method and excipient on the structure and physical stability of protein solids: Freeze drying vs. spray freeze drying. Int J Pharm 2020; 594:120169. [PMID: 33333176 DOI: 10.1016/j.ijpharm.2020.120169] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 11/25/2022]
Abstract
This study aims to determine the impacts of drying method and excipient on changes in protein structure and physical stability of model protein solids. Protein solids containing one of two model proteins (lysozyme or myoglobin) were produced with or without excipients (sucrose or mannitol) using freeze drying or spray freeze drying (SFD). The protein powders were then characterized using solid-state Fourier transform infrared spectroscopy (ssFTIR), differential scanning calorimetry (DSC), circular dichroism spectrometry (CD), size exclusion chromatography (SEC), BET surface area measurements and solid-state hydrogen deuterium exchange with mass spectrometry (ssHDX-MS). ssFTIR and CD could identify little to no difference in structure of the proteins in the formulation. ssHDX-MS was able to identify the population heterogeneity, which was undetectable by conventional characterization techniques of ssFTIR and CD. ssHDX-MS metrics such as Dmax and peak area showed a good correlation with the protein physical instability (loss of the monomeric peak area by size exclusion chromatography) in 90-day stability studies conducted at 40 °C for lysozyme. Higher specific surface area was associated with greater loss in monomer content for myoglobin-mannitol formulations as compared to myoglobin-only formulations. Spray freeze drying seems a viable manufacturing technique for protein solids with appropriate optimization of formulations. The differences observed within the formulations and between the processes using ssHDX-MS, BET surface area measurements and SEC in this study provide an insight into the influence of drying methods and excipients on protein physical stability.
Collapse
Affiliation(s)
- Tarun Tejasvi Mutukuri
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Nathan E Wilson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Elizabeth M Topp
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; National Institute for Bioprocessing Research and Training, Belfield, Blackrock, Co. Dublin A94 X099, Ireland
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
185
|
Tian Z, Qian F. Adenosine Triphosphate-Induced Rapid Liquid-Liquid Phase Separation of a Model IgG1 mAb. Mol Pharm 2020; 18:267-274. [PMID: 33307701 DOI: 10.1021/acs.molpharmaceut.0c00905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adenosine triphosphate (ATP) is amphiphilic in nature and has the characteristics of a hydrotrope because of the charged triphosphate moiety and the large aromatic ring located on each end of its structure. Previous studies revealed that ATP can effectively maintain the solubility and prevent liquid-liquid phase separation (LLPS) of some biological proteins. In this study, we assessed the impact of ATP on the stability of a model therapeutic IgG1 antibody (MA1) to evaluate its potential application in protein formulation design. In our system, ATP promotes rapid LLPS of MA1 and we demonstrate that the ATP-MA1 static interaction drives phase separation of MA1. The attractive protein-protein interaction increased exclusively in the presence of ATP but not in the presence of other ATP analogues, such as adenosine diphosphate, adenosine monophosphate, and adenine. Through an intrinsic fluorescence quenching study, we revealed that ATP bound to MA1 electrostatically and formed static interactions; furthermore, such static ATP-MA1 interactions significantly altered the surface property of the protein and the protein-protein interactions and subsequently induced LLPS of MA1. This ATP-induced LLPS could be effectively eliminated by Mg2+, which chelated with ATP and thus negated ATP-MA1 static interaction. Our results revealed the unique molecular mechanism of ATP-induced rapid LLPS of MA1.
Collapse
Affiliation(s)
- Zhou Tian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, P. R. China
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
186
|
Bui TNN, Sandar S, Luna G, Beaman J, Sunderland B, Czarniak P. An investigation of reconstituted terlipressin infusion stability for use in hepatorenal syndrome. Sci Rep 2020; 10:21037. [PMID: 33273555 PMCID: PMC7712657 DOI: 10.1038/s41598-020-78044-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/19/2020] [Indexed: 12/26/2022] Open
Abstract
Hepatorenal syndrome (HRS) is a fatal complication of renal dysfunction associated with ascites, liver failure and advanced cirrhosis. Although the best option for long-term survival is liver transplantation, in the critical acute phase, vasoconstrictors are considered first-line supportive agents. Terlipressin is the most widely used vasoconstrictor globally but owing to its short elimination half-life, it is usually administered six hourly by slow intravenous bolus injection. This requires patients to remain in hospital, increasing hospital bed costs and affecting their quality of life. An alternative option for administration of terlipressin is as a continuous infusion using an elastomeric infusor device in the patient’s home. However, stability data on terlipressin in elastomeric infusor devices is lacking. This research aimed to evaluate the stability of terlipressin reconstituted in infusor devices for up to 7 days at 2–8 °C and subsequently at 22.5 °C for 24 h, to mimic home storage and administration temperatures. We report that terlipressin was physically and chemically stable under these conditions; all reconstituted infusor concentrations retained above 90% of the original concentration over the test conditions. No colour change or precipitation in the solutions were evident.
Collapse
Affiliation(s)
- Thi Ngoc Nhieu Bui
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Su Sandar
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Giuseppe Luna
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Jasmine Beaman
- Pharmacy Department, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
| | - Bruce Sunderland
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Petra Czarniak
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Kent St, Bentley, WA, 6102, Australia.
| |
Collapse
|
187
|
Butreddy A, Janga KY, Ajjarapu S, Sarabu S, Dudhipala N. Instability of therapeutic proteins - An overview of stresses, stabilization mechanisms and analytical techniques involved in lyophilized proteins. Int J Biol Macromol 2020; 167:309-325. [PMID: 33275971 DOI: 10.1016/j.ijbiomac.2020.11.188] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 01/06/2023]
Abstract
Solid-state is the preferred choice for storage of protein therapeutics to improve stability and preserve the biological activity by decreasing the physical and chemical degradation associated with liquid protein formulations. Lyophilization or freeze-drying is an effective drying method to overcome the instability problems of proteins. However, the processing steps (freezing, primary drying and secondary drying) involved in the lyophilization process can expose the proteins to various stress and harsh conditions, leading to denaturation, aggregation often a loss in activity of protein therapeutics. Stabilizers such as sugars and surfactants are often added to protect the proteins against physical stress associated with lyophilization process and storage conditions. Another way to curtail the degradation of proteins due to process related stress is by modification of the lyophilization process. Slow freezing, high nucleation temperature, decreasing the extent of supercooling, and annealing can minimize the formation of the interface (ice-water) by producing large ice crystals with less surface area, thereby preserving the native structure and stability of the proteins. Hence, a thorough understanding of formulation composition, lyophilization process parameters and the choice of analytical methods to characterize and monitor the protein instability is crucial for development of stable therapeutic protein products. This review provides an overview of various stress conditions that proteins might encounter during lyophilization process, mechanisms to improve the stability and analytical techniques to tackle the proteins instability during both freeze-drying and storage.
Collapse
Affiliation(s)
- Arun Butreddy
- Formulation R&D, Biological E. Limited, IKP Knowledge Park, Shameerpet, Hyderabad, Telangana State 500078, India; Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Karthik Yadav Janga
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Srinivas Ajjarapu
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Sandeep Sarabu
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Narendar Dudhipala
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India; Department of Pharmaceutics, Vaagdevi College of Pharmacy, Warangal, Telangana State 506 005, India..
| |
Collapse
|
188
|
Mass spectrometric and kinetics characterization of modified species of Growth Hormone Releasing Hexapeptide generated under thermal stress in different pH and buffers. J Pharm Biomed Anal 2020; 194:113776. [PMID: 33272786 DOI: 10.1016/j.jpba.2020.113776] [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: 05/13/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/22/2022]
Abstract
Growth Hormone Releasing Peptide-6 (GHRP-6) is a promising molecule (H-His1-d-Trp- Ala-Trp-d-Phe-Lys6-NH2) for the treatment of several diseases. Studies on the degradation pathways of this molecule under stressed conditions are needed to develop appropriate formulations. Degradation products (DPs) of GHRP-6, generated by heating in the dark at 60 °C with pH ranging from 3.0 to 8.0 and in presence of common buffers, were isolated by RP-HPLC and characterized by ESI-MS/MS. C-terminal deamidation of GHRP-6 was generated preferentially at pH 3.0 and 8.0. Hydrolysis and head-to-tail cyclization were favored at pH ranging from 6.0 to 7.0 in phosphate containing buffers. A DP with +12 Da molecular mass was presumably originated by the reaction with formaldehyde derived from some of the additives and/or elastomeric closures. Certain DPs derived from the acylation reaction of the tri- and di-carboxylic buffering species were favored at pH 3.0-6.0 and indicate that buffer components, including those "Generally Recognized as Safe", may potentially introduce chemical modifications and product heterogeneity. Nano LC-MS/MS analysis revealed GHRP-6 was also detected as a low-abundance species with Trp oxidized to 5-hydroxy, kynurenine, and N-formylkynurenine. The kinetics for the formation of the major degradation products was also studied by RP-HPLC.
Collapse
|
189
|
Thomas SL, Thacker JB, Schug KA, Maráková K. Sample preparation and fractionation techniques for intact proteins for mass spectrometric analysis. J Sep Sci 2020; 44:211-246. [DOI: 10.1002/jssc.202000936] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Shannon L. Thomas
- Department of Chemistry & Biochemistry The University of Texas Arlington Arlington Texas USA
| | - Jonathan B. Thacker
- Department of Chemistry & Biochemistry The University of Texas Arlington Arlington Texas USA
| | - Kevin A. Schug
- Department of Chemistry & Biochemistry The University of Texas Arlington Arlington Texas USA
| | - Katarína Maráková
- Department of Pharmaceutical Analysis and Nuclear Pharmacy Faculty of Pharmacy Comenius University in Bratislava Bratislava Slovakia
| |
Collapse
|
190
|
Köhn B, Kovermann M. All atom insights into the impact of crowded environments on protein stability by NMR spectroscopy. Nat Commun 2020; 11:5760. [PMID: 33188202 PMCID: PMC7666220 DOI: 10.1038/s41467-020-19616-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 10/23/2020] [Indexed: 01/16/2023] Open
Abstract
The high density of macromolecules affecting proteins due to volume exclusion has been discussed in theory but numerous in vivo experiments cannot be sufficiently understood taking only pure entropic stabilization into account. Here, we show that the thermodynamic stability of a beta barrel protein increases equally at all atomic levels comparing crowded environments with dilute conditions by applying multidimensional high-resolution NMR spectroscopy in a systematic manner. Different crowding agents evoke a pure stabilization cooperatively and do not disturb the surface or integrity of the protein fold. The here developed methodology provides a solid base that can be easily expanded to incorporate e.g. binding partners to recognize functional consequences of crowded conditions. Our results are relevant to research projects targeting soluble proteins in vivo as it can be anticipated that their thermodynamic stability increase comparably and has consequently to be taken into account to coherently understand intracellular processes.
Collapse
Affiliation(s)
- Birgit Köhn
- Department of Chemistry, University of Konstanz, Universitätsstrasse. 10, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology KoRS-CB, University of Konstanz, Universitätsstrasse. 10, 78457, Konstanz, Germany
| | - Michael Kovermann
- Department of Chemistry, University of Konstanz, Universitätsstrasse. 10, 78457, Konstanz, Germany.
- Konstanz Research School Chemical Biology KoRS-CB, University of Konstanz, Universitätsstrasse. 10, 78457, Konstanz, Germany.
| |
Collapse
|
191
|
Xiao W, Jakimowicz MD, Zampetakis I, Neely S, Scarpa F, Davis SA, Williams DS, Perriman AW. Biopolymeric Coacervate Microvectors for the Delivery of Functional Proteins to Cells. ACTA ACUST UNITED AC 2020; 4:e2000101. [PMID: 33166084 DOI: 10.1002/adbi.202000101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/29/2020] [Indexed: 11/07/2022]
Abstract
The extent to which biologic payloads can be effectively delivered to cells is a limiting factor in the development of new therapies. Limitations arise from the lack of pharmacokinetic stability of biologics in vivo. Encapsulating biologics in a protective delivery vector has the potential to improve delivery profile and enhance performance. Coacervate microdroplets are developed as cell-mimetic materials with established potential for the stabilization of biological molecules, such as proteins and nucleic acids. Here, the development of biodegradable coacervate microvectors (comprising synthetically modified amylose polymers) is presented, for the delivery of biologic payloads to cells. Amylose-based coacervate microdroplets are stable under physiological conditions (e.g., temperature and ionic strength), are noncytotoxic owing to their biopolymeric structure, spontaneously interacted with the cell membrane, and are able to deliver and release proteinaceous payloads beyond the plasma membrane. In particular, myoglobin, an oxygen storage and antioxidant protein, is successfully delivered into human mesenchymal stem cells (hMSCs) within 24 h. Furthermore, coacervate microvectors are implemented for the delivery of human bone morphogenetic protein 2 growth factor, inducing differentiation of hMSCs into osteoprogenitor cells. This study demonstrates the potential of coacervate microdroplets as delivery microvectors for biomedical research and the development of new therapies.
Collapse
Affiliation(s)
- Wenjin Xiao
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Monika D Jakimowicz
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
- HH Wills Physics Laboratory, University of Bristol, Bristol, BS8 1TL, UK
- Centre for Organized Matter Chemistry and Centre for Protolife Research School of Chemistry, University of Bristol, Bristol, BS8 1TS, UK
| | - Ioannis Zampetakis
- Bristol Composites Institute (ACCIS), Department of Aerospace Engineering, University of Bristol, Bristol, BS8 1TF, UK
| | - Sarah Neely
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), Department of Aerospace Engineering, University of Bristol, Bristol, BS8 1TF, UK
| | - Sean A Davis
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
| | - David S Williams
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
192
|
Fayter AE, Hasan M, Congdon TR, Kontopoulou I, Gibson MI. Ice recrystallisation inhibiting polymers prevent irreversible protein aggregation during solvent-free cryopreservation as additives and as covalent polymer-protein conjugates. Eur Polym J 2020; 140:110036. [PMID: 33311718 PMCID: PMC7709485 DOI: 10.1016/j.eurpolymj.2020.110036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 01/11/2023]
Abstract
Protein storage and transport is essential to deliver therapies (biologics), enzymes for biotechnological applications, and underpins fundamental structural and molecular biology. To enable proteins to be stored and transported it is often essential to freeze them, requiring cryoprotectants such as glycerol or trehalose. Here we explore the mechanisms by which poly(vinyl alcohol), PVA, a potent ice recrystallisation inhibitor protects proteins during freeze/thaw to enable solvent-free cryopreservation with a focus on comparing mixing, verses polymer-protein conjugation. A panel of poly(vinyl alcohol)s are investigated including commercial, well-defined (from RAFT), and PVA-protein conjugates, to map out PVA's efficacy. Enzymatic activity recovery of lactate dehydrogenase was found to correlate with post-thaw aggregation state (less aggregated protein had greater activity), which was modulated by PVA's ice recrystallisation inhibition activity. This macromolecular cryoprotectant matched the performance of glycerol, but at lower additive concentrations (as low as 1 mg.mL-1). It was also demonstrated that storage at -20 °C, rather than -80 °C was possible using PVA as a cryoprotectant, which is not possible with glycerol storage. A second protein, green-fluorescent protein (GFP), was used to enable screening of molecular weight effects and to obtain PVA-GFP bioconjugates. It was observed that covalent attachment of RAFT-derived PVA showed superior cryoprotectant activity compared to simple mixing of the polymer and protein. These results show that PVA is a real alternative to solvent-based protein storage with potential in biotechnology, food and therapeutics. PVA is already approved for many biomedical applications, is low cost and available on a large scale, making it an ideal cryoprotectant formulation enhancer.
Collapse
Affiliation(s)
- Alice E.R. Fayter
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Muhammad Hasan
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Thomas R. Congdon
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | | | - Matthew I. Gibson
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
193
|
Beloqui A, Mane SR, Langer M, Glassner M, Bauer DM, Fruk L, Barner‐Kowollik C, Delaittre G. Hetero‐Diels‐Alder‐Cycloaddition mit RAFT‐Polymeren als Biokonjugationsplattform. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ana Beloqui
- Institute of Biological and Chemical Systems (IBCS) Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
- Department of Applied Chemistry (UPV/EHU) Avda. Manuel de Lardizabal 3 E-20018 Donostia – San Sebastian Spanien
- IKERBASQUE Basque Foundation for Science Maria Diaz de Haro 3 E-48013 Bilbao Spanien
| | - Shivshankar R. Mane
- Institute of Biological and Chemical Systems (IBCS) Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
| | - Marcel Langer
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
| | - Mathias Glassner
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
| | - Dennis M. Bauer
- Center for Functional Nanostructures (CFN) Karlsruhe Institute of Technology (KIT) Wolfgang-Gaede-Straße 1a 76131 Karlsruhe Deutschland
| | - Ljiljana Fruk
- Center for Functional Nanostructures (CFN) Karlsruhe Institute of Technology (KIT) Wolfgang-Gaede-Straße 1a 76131 Karlsruhe Deutschland
- Department of Chemical Engineering and Biotechnology University of Cambridge West Cambridge Site, Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - Christopher Barner‐Kowollik
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
- Centre for Materials Science Queensland University of Technology (QUT) 2 George Street Brisbane QLD 4000 Australien
- School of Chemistry and Physics Queensland University of Technology (QUT) 2 George Street Brisbane QLD 4000 Australien
| | - Guillaume Delaittre
- Institute of Biological and Chemical Systems (IBCS) Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
- Macromolecular Architectures Institute for Chemical Technology and Polymer Chemistry (ITCP) Karlsruhe Institute of Technology (KIT) Engesserstr. 18 76131 Karlsruhe Deutschland
- Organic Functional Molecules Organic Chemistry University of Wuppertal Gaußstrasse 20 42119 Wuppertal Deutschland
| |
Collapse
|
194
|
Beloqui A, Mane SR, Langer M, Glassner M, Bauer DM, Fruk L, Barner‐Kowollik C, Delaittre G. Hetero-Diels-Alder Cycloaddition with RAFT Polymers as Bioconjugation Platform. Angew Chem Int Ed Engl 2020; 59:19951-19955. [PMID: 32729643 PMCID: PMC7693046 DOI: 10.1002/anie.202005747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Indexed: 12/16/2022]
Abstract
We introduce the bioconjugation of polymers synthesized by RAFT polymerization, bearing no specific functional end group, by means of hetero-Diels-Alder cycloaddition through their inherent terminal thiocarbonylthio moiety with a diene-modified model protein. Quantitative conjugation occurs over the course of a few hours, at ambient temperature and neutral pH, and in the absence of any catalyst. Our technology platform affords thermoresponsive bioconjugates, whose aggregation is solely controlled by the polymer chains.
Collapse
Affiliation(s)
- Ana Beloqui
- Institute of Biological and Chemical Systems (IBCS)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
- Department of Applied Chemistry (UPV/EHU)Avda. Manuel de Lardizabal 3E-20018Donostia – San SebastianSpain
- IKERBASQUEBasque Foundation for ScienceMaria Diaz de Haro 3E-48013BilbaoSpain
| | - Shivshankar R. Mane
- Institute of Biological and Chemical Systems (IBCS)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
| | - Marcel Langer
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
| | - Mathias Glassner
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
| | - Dennis M. Bauer
- Center for Functional Nanostructures (CFN)Karlsruhe Institute of Technology (KIT)Wolfgang-Gaede-Straße 1a76131KarlsruheGermany
| | - Ljiljana Fruk
- Center for Functional Nanostructures (CFN)Karlsruhe Institute of Technology (KIT)Wolfgang-Gaede-Straße 1a76131KarlsruheGermany
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeWest Cambridge Site, Philippa Fawcett DriveCambridgeCB3 0ASUK
| | - Christopher Barner‐Kowollik
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
- Centre for Materials ScienceQueensland University of Technology (QUT)2 George StreetBrisbaneQLD4000Australia
- School of Chemistry and PhysicsQueensland University of Technology (QUT)2 George StreetBrisbaneQLD4000Australia
| | - Guillaume Delaittre
- Institute of Biological and Chemical Systems (IBCS)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
- Macromolecular ArchitecturesInstitute for Chemical Technology and Polymer Chemistry (ITCP)Karlsruhe Institute of Technology (KIT)Engesserstr. 1876131KarlsruheGermany
- Organic Functional MoleculesOrganic ChemistryUniversity of WuppertalGaußstrasse 2042119WuppertalGermany
| |
Collapse
|
195
|
Simultaneous Monitoring and Comparison of Multiple Product Quality Attributes for Cell Culture Processes at Different Scales Using a LC/MS/MS Based Multi-Attribute Method. J Pharm Sci 2020; 109:3319-3329. [DOI: 10.1016/j.xphs.2020.07.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/07/2020] [Accepted: 07/27/2020] [Indexed: 01/02/2023]
|
196
|
Ren CD, Qi W, Wyatt EA, Yeary J, Westland K, Berke M, Rathore N. Application of a High Throughput and Automated Workflow to Therapeutic Protein Formulation Development. J Pharm Sci 2020; 110:1130-1141. [PMID: 33203511 DOI: 10.1016/j.xphs.2020.10.040] [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: 06/30/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Rapid and efficient formulation development is critical to successfully bringing therapeutic protein drug products into a competitive market under increasingly aggressive timelines. Conventional application of high throughput techniques for formulation development have been limited to lower protein concentrations, which are not applicable to late stage development of high concentration therapeutics. In this work, we present a high throughput (HT) formulation workflow that enables screening at representative concentrations via integration of a micro-buffer exchange system with automated analytical instruments. The operational recommendations associated with the use of such HT systems as well as the efficiencies gained (reduction in hands-on time and run time by over 70% and 30%, respectively), which enable practical characterization of an expanded formulation design space, are discussed. To demonstrate that the workflow is fit for purpose, the formulation properties and stability profiles (SEC and CEX) from samples generated by the HT workflow were compared to those processed by ultrafiltration/diafiltration, and the results were shown to be in good agreement. This approach was further applied to two case studies, one focused on a formulation screen that studied the effects of pH and excipient on viscosity and stability, and the other focused on selection of an appropriate viscosity mimic solution for a protein product.
Collapse
Affiliation(s)
- Cindy D Ren
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320.
| | - Wei Qi
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Emily A Wyatt
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Jeffrey Yeary
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | | | - Michael Berke
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Nitin Rathore
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| |
Collapse
|
197
|
Ibeanu N, Egbu R, Onyekuru L, Javaheri H, Tee Khaw P, R. Williams G, Brocchini S, Awwad S. Injectables and Depots to Prolong Drug Action of Proteins and Peptides. Pharmaceutics 2020; 12:E999. [PMID: 33096803 PMCID: PMC7589296 DOI: 10.3390/pharmaceutics12100999] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Proteins and peptides have emerged in recent years to treat a wide range of multifaceted diseases such as cancer, diabetes and inflammation. The emergence of polypeptides has yielded advancements in the fields of biopharmaceutical production and formulation. Polypeptides often display poor pharmacokinetics, limited permeability across biological barriers, suboptimal biodistribution, and some proclivity for immunogenicity. Frequent administration of polypeptides is generally required to maintain adequate therapeutic levels, which can limit efficacy and compliance while increasing adverse reactions. Many strategies to increase the duration of action of therapeutic polypeptides have been described with many clinical products having been developed. This review describes approaches to optimise polypeptide delivery organised by the commonly used routes of administration. Future innovations in formulation may hold the key to the continued successful development of proteins and peptides with optimal clinical properties.
Collapse
Affiliation(s)
- Nkiruka Ibeanu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Raphael Egbu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Lesley Onyekuru
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Hoda Javaheri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Gareth R. Williams
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Steve Brocchini
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Sahar Awwad
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| |
Collapse
|
198
|
Darriba ML, Cerutti ML, Bruno L, Cassataro J, Pasquevich KA. Stability Studies of the Vaccine Adjuvant U-Omp19. J Pharm Sci 2020; 110:707-718. [PMID: 33058898 PMCID: PMC7815325 DOI: 10.1016/j.xphs.2020.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 01/18/2023]
Abstract
Unlipidated outer membrane protein 19 (U-Omp19) is a novel mucosal adjuvant in preclinical development to be used in vaccine formulations. U-Omp19 holds two main properties, it is capable of inhibiting gastrointestinal and lysosomal peptidases, increasing the amount of co-administered antigen that reaches the immune inductive sites and its half-life inside cells, and it is able to stimulate antigen presenting cells in vivo. These activities enable U-Omp19 to enhance the adaptive immune response to co-administrated antigens. To characterize the stability of U-Omp19 we have performed an extensive analysis of its physicochemical and biological properties in a 3-year long-term stability study, and under potentially damaging freeze-thawing and lyophilization stress processes. Results revealed that U-Omp19 retains its full protease inhibitor activity, its monomeric state and its secondary structure even when stored in solution for 36 months or after multiple freeze-thawing cycles. Non-enzymatic hydrolysis resulted the major degradation pathway for storage in solution at 4 °C or room temperature which can be abrogated by lyophilization yet increasing protein tendency to form aggregates. This information will play a key role in the development of a stable formulation of U-Omp19, allowing an extended shelf-life during manufacturing, storage, and shipping of a future vaccine containing this pioneering adjuvant.
Collapse
Affiliation(s)
- M Laura Darriba
- Instituto de Investigaciones Biotecnológicas (UNSAM-CONICET), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - María L Cerutti
- Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina.
| | - Laura Bruno
- Instituto de Investigaciones Biotecnológicas (UNSAM-CONICET), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas (UNSAM-CONICET), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Karina A Pasquevich
- Instituto de Investigaciones Biotecnológicas (UNSAM-CONICET), Universidad Nacional de San Martín, Buenos Aires, Argentina.
| |
Collapse
|
199
|
Fang WJ, Liu JW, Zheng HJ, Shen BB, Wang X, Kong Y, Jing ZY, Gao JQ. Protein Sub-Visible Particle and Free Radical formation of a Freeze-Dried Monoclonal Antibody Formulation During Dropping. J Pharm Sci 2020; 110:1625-1634. [PMID: 33049261 DOI: 10.1016/j.xphs.2020.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/24/2020] [Accepted: 10/06/2020] [Indexed: 01/31/2023]
Abstract
Dropping during shipping and handling of liquid biopharmaceutical formulations has long been known to cause protein degradation and aggregation. On the other hand, accidental dropping of freeze-dried protein formulations is generally considered not a major issue for biopharmaceutical quality. Reports of stability and especially the underling degradation mechanism(s) during shipping and handling of freeze-dried protein formulations were rarely seen in literature. In this manuscript, we report an interesting phenomenon in which repeated dropping of freeze-dried monoclonal antibody X (mAb-X) formulation powder resulted in significant protein sub-visible particles (SbVPs) in the reconstituted liquid as determined by the sensitive particle analyzing technique micro-flow imaging (MFI). Free radicals were observed after repeated dropping by electron paramagnetic resonance (EPR). Formation of SbVPs could be partially inhibited by the free radical scavengers methionine and 3-carbamoyl-2,2,5,5-tetramethyl-1-pyrrolidin-yloxy free radical (CTPO). The amount of free radicals and SbVPs was correlated to the sample temperature during dropping. Therefore we propose that the high temperature formed during dropping was probably the root cause for protein aggregation and free radical formation, which could further cause protein aggregation. Our observations suggest that similar to liquid protein formulations, dropping of freeze-dried protein formulations should also be avoided or mitigated.
Collapse
Affiliation(s)
- Wei-Jie Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016 China.
| | - Jia-Wei Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016 China
| | - Hong-Jian Zheng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016 China
| | - Bin-Bin Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016 China
| | - Xinyu Wang
- Department of Chemistry, Zhejiang University, Hangzhou, 310013 China
| | - Yi Kong
- The First People's Hospital of Xiaoshan District, Hangzhou, 311200 China
| | - Zhen-Yi Jing
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016 China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
| |
Collapse
|
200
|
Controlling Ice Nucleation during Lyophilization: Process Optimization of Vacuum-Induced Surface Freezing. Processes (Basel) 2020. [DOI: 10.3390/pr8101263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Biopharmaceuticals are often lyophilized to improve their storage stability. Controlling ice nucleation during the freezing step of the lyophilization process is desired to increase homogeneity of product properties across a drug product batch and shorten the primary drying time. The present communication summarizes the process optimization of the freezing process when using vacuum-induced surface freezing to control ice nucleation, in particular for amorphous samples. We characterized freeze-dried samples for solid state properties, and compared these to uncontrolled nucleated samples using bovine serum albumin (BSA) as a model protein. Freezing parameters were optimized to obtain complete nucleation, adequate cake resistance during the subsequent lyophilization cycle, and elegant cakes. We highlight the challenges associated with vacuum-induced surface freezing and propose optimized freezing parameters to control ice nucleation, enabling manufacturing of amorphous samples.
Collapse
|