1
|
Patil CD, Tejasvi Mutukuri T, Santosh Arte K, Huang Y, Radhakrishnan V, Tony Zhou Q. Effects of buffers on spray-freeze-dried/lyophilized high concentration protein formulations. Int J Pharm 2025; 668:124974. [PMID: 39571769 DOI: 10.1016/j.ijpharm.2024.124974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Solid-state protein formulations are known to exhibit enhanced storage stability compared to their liquid dosage form counterparts. pH is one of the factors affecting the stability of protein formulations. The pH of protein formulations in the solution could be influenced by the buffer used, directly impacting their solid-state stability. During lyophilization, buffer components may interact with other formulation components present in the protein formulations, causing a pH shift. This study aimed to investigate the effects of phosphate buffer and amino acid buffers (such as histidine and/or arginine) on the physical properties and accelerated storage stability of spray freeze-dried or lyophilized protein formulations. A model protein, bovine serum albumin (BSA), was used to prepare high-concentration protein formulations. The formulations consisted of BSA, trehalose, and mannitol in an 80:15:5 ratio (w/w), respectively. Various buffers were utilized in the preparation of protein formulations, and the resultant solid formulations underwent screening via accelerated stability study using size exclusion chromatography (SEC). The combination of phosphate and arginine buffers resulted in increased monomer loss in the accelerated storage stability study. Additional characterizations, including solid-state Fourier transform infrared spectroscopy (ssFTIR) and powder X-ray diffraction (PXRD), were conducted. While these analyses did not definitively elucidate the mechanism behind the observed instability, their outcomes provide valuable insights for further investigation, highlighting the need for future research in this area.
Collapse
Affiliation(s)
- Chanakya D Patil
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Tarun Tejasvi Mutukuri
- Injectable Drug Product Development, Alexion Pharmaceuticals, Inc. (AstraZeneca Rare Disease Unit), New Haven, CT 06510, USA
| | - Kinnari Santosh Arte
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Yijing Huang
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Vinay Radhakrishnan
- Injectable Drug Product Development, Alexion Pharmaceuticals, Inc. (AstraZeneca Rare Disease Unit), New Haven, CT 06510, USA
| | - Qi Tony Zhou
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
2
|
Lo Presti K, Jégo M, Frieß W. "The More, the Better?": The Impact of Sugar-to-Protein Molar Ratio in Freeze-Dried Monoclonal Antibody Formulations on Protein Stability. Mol Pharm 2024. [PMID: 39564766 DOI: 10.1021/acs.molpharmaceut.4c01174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Lyophilization is widely used to ensure the stability of protein drugs by minimizing chemical and physical degradation in the dry solid state. To this end, proteins are typically formulated with sugars that form an amorphous immobilizing matrix and stabilize hydrogen bonds replacing water molecules. The optimal amount of sugar required and protein stability at low excipient-to-protein molar ratios are not well understood. We investigated this by focusing on the physical stability of formulations, reflecting highly concentrated monoclonal antibody (mAb) lyophilizates at low sucrose to mAb ratios between 25:1 and 360:1. Additionally, the impact of different excipient types, buffer concentrations, and polysorbates was studied. The mAb stability was evaluated over up to three months at 25 and 40 °C. We investigated the "the more, the better" approach regarding excipient usage in protein formulation and the existence of a potential stabilizing threshold. Our findings show efficient monomeric content preservation even at low molar ratios, which could be explained based on the water replacement theory. We identified an exponential correlation between the sucrose to protein molar ratio and aggregate formation and found that there is no molar ratio threshold to achieve minimum stabilization. Sucrose demonstrated the best stabilization effect. Both mannitol, used as a cryoprotectant at low concentrations, and arginine reduced aggregation compared to the pure mAb formulation. The higher ionic strength of 5 mM histidine buffer enhanced protein stability compared to that of 0.1 mM histidine buffer, which was more pronounced at lower molar ratios. The addition of polysorbate 20 contributed an additional interfacial stabilizing effect, complementing the cryoprotective and lyoprotective properties of sucrose. Overall, a model could be developed to optimize the quantity of sugar required for protein stabilization and facilitate a more rational design of protein lyophilizates. The molar ratio of sugar to protein for high-concentration mAb products is limited by the acceptable tonicity, but we showed that sufficient stabilization can be achieved even at low molar ratios.
Collapse
Affiliation(s)
- Ken Lo Presti
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Mathilde Jégo
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, Munich 81377, Germany
- Université Claude Bernard, 43 Bd du 11 Novembre 1918, Villeurbanne 69100, France
| | - Wolfgang Frieß
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, Munich 81377, Germany
| |
Collapse
|
3
|
Sheehan K, Jeon H, Corr SC, Hayes JM, Mok KH. Antibody Aggregation: A Problem Within the Biopharmaceutical Industry and Its Role in AL Amyloidosis Disease. Protein J 2024:10.1007/s10930-024-10237-6. [PMID: 39527351 DOI: 10.1007/s10930-024-10237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Due to the large size and rapid growth of the global therapeutic antibody market, there is major interest in understanding the aggregation of protein products as it can compromise efficacy, concentration, and safety. Various production and storage conditions have been identified as capable of inducing aggregation of polyclonal and monoclonal antibody (mAb) therapies such as low pH, freezing, light exposure, lyophilisation and increased ionic strength. The addition of stabilising excipients to these therapeutics helps to combat the formation of aggregates with future aggregation inhibition mechanisms involving the introduction of point mutations and glycoengineering within aggregation prone regions (APRs). Antibody aggregation also plays an integral role in the pathogenesis of a condition known as amyloid light chain (AL) amyloidosis which is characterised by the production of improperly folded and amyloidogenic immunoglobulin light chains (LCs). Current diagnostic tools rely heavily on histological staining with their future moving towards amyloid component identification and proteomic analysis. For many years, treatment options designed for multiple myeloma (MM) have been applied to AL amyloidosis patients by depleting plasma cell numbers. More recently, treatment strategies more specific to this condition have been developed with many designed to recognize amyloid fibrils and trigger their degradation without causing systemic plasma cell cytotoxicity. Amyloid fibrils in AL disease and aggregates in antibody therapeutics are both formed through the oligomerisation of misfolded / modified proteins attempting to reach a thermodynamically stable, free energy minimum that is lower than the respective monomers themselves. Although the final morphologies are different, by understanding the principles underlying such aggregation, we expect to find common insights that may contribute to the development of new and effective methods of antibody aggregation and/or amyloidosis management. We envision that this area of research will continue to be very relevant in both industry and clinical settings.
Collapse
Affiliation(s)
- Kate Sheehan
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - Hyesoo Jeon
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- Lonza Biologics Tuas Pte. Ltd., 35 Tuas South Ave 6, Singapore, 637377, Republic of Singapore
| | - Sinéad C Corr
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jerrard M Hayes
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - K H Mok
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| |
Collapse
|
4
|
Lv JY, Ingle RG, Wu H, Liu C, Fang WJ. Histidine as a versatile excipient in the protein-based biopharmaceutical formulations. Int J Pharm 2024; 662:124472. [PMID: 39013532 DOI: 10.1016/j.ijpharm.2024.124472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
Adequate stabilization is essential for marketed protein-based biopharmaceutical formulations to withstand the various stresses that can be exerted during the pre- and post-manufacturing processes. Therefore, a suitable choice of excipient is a significant step in the manufacturing of such delicate products. Histidine, an essential amino acid, has been extensively used in protein-based biopharmaceutical formulations. The physicochemical properties of histidine are unique among amino acids and could afford multifaceted benefits to protein-based biopharmaceutical formulations. With a pKa of approximately 6.0 at the side chain, histidine has been primarily used as a buffering agent, especially for pH 5.5-6.5. Additionally, histidine exhibited several affirmative properties similar to those of carbohydrates (e.g., sucrose and trehalose) and could therefore be considered to be an alternative approach to established protein-based formulation strategies. The current review describes the general physicochemical properties of histidine, lists all commercial histidine-containing protein-based biopharmaceutical products, and discusses a brief outline of the existing research focused on the versatile applications of histidine, which can act as a buffering agent, stabilizer, cryo-/lyo-protectant, antioxidant, viscosity reducer, and solubilizing agent. The interaction between histidine and proteins in protein-based biopharmaceutical formulations, such as the Donnan effect during diafiltration of monoclonal antibody solutions and the degradation of polysorbates in histidine buffer, has also been discussed. As the first review of histidine in protein biopharmaceuticals, it helps to deepen our understanding of the opportunities and challenges associated with histidine as an excipient for protein-based biopharmaceutical formulations.
Collapse
Affiliation(s)
- Jia-Yi Lv
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Taizhou Institute of Zhejiang University, Taizhou, Zhejiang 317000, China; School of Pharmaceutical Sciences, Xiamen University, 4221 Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Rahul G Ingle
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education & Research (Deemed to University), Sawangi, Wardha, India
| | - Hao Wu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Cuihua Liu
- Bio-Thera Solutions, Ltd, Guangzhou, Guangdong 510530, China
| | - Wei-Jie Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Taizhou Institute of Zhejiang University, Taizhou, Zhejiang 317000, China; Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua, 321000, China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
5
|
Brom JA, Petrikis RG, Nieukirk GE, Bourque J, Pielak GJ. Protecting Lyophilized Escherichia coli Adenylate Kinase. Mol Pharm 2024; 21:3634-3642. [PMID: 38805365 DOI: 10.1021/acs.molpharmaceut.4c00356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Drying protein-based drugs, usually via lyophilization, can facilitate storage at ambient temperature and improve accessibility but many proteins cannot withstand drying and must be formulated with protective additives called excipients. However, mechanisms of protection are poorly understood, precluding rational formulation design. To better understand dry proteins and their protection, we examine Escherichia coli adenylate kinase (AdK) lyophilized alone and with the additives trehalose, maltose, bovine serum albumin, cytosolic abundant heat soluble protein D, histidine, and arginine. We apply liquid-observed vapor exchange NMR to interrogate the residue-level structure in the presence and absence of additives. We pair these observations with differential scanning calorimetry data of lyophilized samples and AdK activity assays with and without heating. We show that the amino acids do not preserve the native structure as well as sugars or proteins and that after heating the most stable additives protect activity best.
Collapse
Affiliation(s)
- Julia A Brom
- Department of Chemistry, University of North Carolina at Chapel Hill (UNC-CH), 3250 Genome Sciences Building, Chapel Hill, North Carolina 27599-3290, United States
| | - Ruta G Petrikis
- Department of Chemistry, University of North Carolina at Chapel Hill (UNC-CH), 3250 Genome Sciences Building, Chapel Hill, North Carolina 27599-3290, United States
| | - Grace E Nieukirk
- Department of Chemistry, University of North Carolina at Chapel Hill (UNC-CH), 3250 Genome Sciences Building, Chapel Hill, North Carolina 27599-3290, United States
| | - Joshua Bourque
- Department of Chemistry, University of North Carolina at Chapel Hill (UNC-CH), 3250 Genome Sciences Building, Chapel Hill, North Carolina 27599-3290, United States
| | - Gary J Pielak
- Department of Chemistry, University of North Carolina at Chapel Hill (UNC-CH), 3250 Genome Sciences Building, Chapel Hill, North Carolina 27599-3290, United States
- Department of Biochemistry & Biophysics, UNC-CH, Chapel Hill, North Carolina 27599, United States
- Lineberger Cancer Center, UNC-CH, Chapel Hill, North Carolina 27599, United States
- Integrative Program for Biological and Genome Sciences, UNC-CH, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
6
|
T T Nguyen K, Zillen D, Lasorsa A, van der Wel PCA, Frijlink HW, L J Hinrichs W. Combinations of arginine and pullulan reveal the selective effect of stabilization mechanisms on different lyophilized proteins. Int J Pharm 2024; 654:123938. [PMID: 38408554 DOI: 10.1016/j.ijpharm.2024.123938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
The stability of lactate dehydrogenase (LDH) and β-galactosidase (β-gal), incorporated in arginine/pullulan (A/P) mixtures at various weight ratios by lyophilization, was determined. The physicochemical characteristics of various A/P mixtures were assessed. With decreasing A/P ratios, the glass transition temperature of the formulations increased. Furthermore, arginine crystallization due to high relative humidity (RH) exposure was prevented at an A/P weight ratio of 4/6 or less. When stored at 0 % RH / 60 °C for 4 weeks, arginine was superior to pullulan as stabilizer. During storage at 43 % RH / 30 ℃ for 4 weeks, the enzymatic activity of LDH was best retained at an A/P weight ratio of 2/8, while β-gal activity was relatively well-retained at A/P weight ratios of both 8/2 and 2/8. LDH seemed to be more prone to degradation in the rubbery state. In the glassy state, β-gal degraded faster than LDH. Solid-state nuclear magnetic resonance spectroscopy showed that (labeled) arginine experienced a different interaction in the two protein samples, reflecting a modulation of long-range correlations of the arginine side chain nitrogen atoms (Nε, Nη). In summary, LDH stabilization in the A/P matrix requires vitrification. Further stabilization difference between LDH and β-gal may be dependent on the interaction with arginine.
Collapse
Affiliation(s)
- Khanh T T Nguyen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Daan Zillen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Alessia Lasorsa
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, the Netherlands
| | - Patrick C A van der Wel
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, the Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
7
|
Ren S. Effects of arginine in therapeutic protein formulations: a decade review and perspectives. Antib Ther 2023; 6:265-276. [PMID: 38075239 PMCID: PMC10702853 DOI: 10.1093/abt/tbad022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/05/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2024] Open
Abstract
Arginine (Arg) is a natural amino acid with an acceptable safety profile and a unique chemical structure. Arg and its salts are highly effective in enhancing protein refolding and solubilization, suppressing protein-protein interaction and aggregation and reducing viscosity of high concentration protein formulations. Arg and its salts have been used in research and 20 approved protein injectables. This review summarizes the effects of Arg as an excipient in therapeutic protein formulations with the focus on its physicochemical properties, safety, applications in approved protein products, beneficial and detrimental effects in liquid and lyophilized protein formulations when combined with different counterions and mechanism on protein stabilization and destabilization. The decade literature review indicates that the benefits of Arg overweigh its risks when it is used appropriately. It is recommended to add Arg along with glutamate as a counterion to high concentration protein formulations on top of sugars or polyols to counterbalance the negative effects of Arg hydrochloride. The use of Arg as a viscosity reducer and protein stabilizer in high concentration formulations will be the inevitable future trend of the biopharmaceutical industry for subcutaneous administration.
Collapse
Affiliation(s)
- Steven Ren
- CMC Management, WuXi Biologics, 7 Clarke Drive, Cranbury, NJ 08512, USA
| |
Collapse
|
8
|
Härdter N, Geidobler R, Presser I, Winter G. Accelerated Production of Biopharmaceuticals via Microwave-Assisted Freeze-Drying (MFD). Pharmaceutics 2023; 15:pharmaceutics15051342. [PMID: 37242584 DOI: 10.3390/pharmaceutics15051342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recently, attention has been drawn to microwave-assisted freeze-drying (MFD), as it drastically reduces the typically long drying times of biopharmaceuticals in conventional freeze-drying (CFD). Nevertheless, previously described prototype machines lack important attributes such as in-chamber freezing and stoppering, not allowing for the performance of representative vial freeze-drying processes. In this study, we present a new technical MFD setup, designed with GMP processes in mind. It is based on a standard lyophilizer equipped with flat semiconductor microwave modules. The idea was to enable the retrofitting of standard freeze-dryers with a microwave option, which would reduce the hurdles of implementation. We aimed to collect process data with respect to the speed, settings, and controllability of the MFD processes. Moreover, we studied the performance of six monoclonal antibody (mAb) formulations in terms of quality after drying and stability after storage for 6 months. We found drying processes to be drastically shortened and well controllable and observed no signs of plasma discharge. The characterization of the lyophilizates revealed an elegant cake appearance and remarkably good stability in the mAb after MFD. Furthermore, overall storage stability was good, even when residual moisture was increased due to high concentrations of glass-forming excipients. A direct comparison of stability data following MFD and CFD demonstrated similar stability profiles. We conclude that the new machine design is highly advantageous, enabling the fast-drying of excipient-dominated, low-concentrated mAb formulations in compliance with modern manufacturing technology.
Collapse
Affiliation(s)
- Nicole Härdter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Raimund Geidobler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Pharmaceutical Development Biologicals, 88397 Biberach an der Riß, Germany
| | - Ingo Presser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Pharmaceutical Development Biologicals, 88397 Biberach an der Riß, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
9
|
Perodeau J, Arbogast LW, Nieuwkoop AJ. Solid-State NMR Characterization of Lyophilized Formulations of Monoclonal Antibody Therapeutics. Mol Pharm 2023; 20:1480-1489. [PMID: 36702622 DOI: 10.1021/acs.molpharmaceut.2c00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Monoclonal antibodies (mAbs) are an important and growing class of biotherapeutic drugs. Method development for the characterization of critical quality attributes, including higher-order structure (HOS), of mAbs remains an area of active inquiry. Recently, solution-state nuclear magnetic resonance (NMR) spectroscopy has received increased attention and is a means for reliable, high-resolution HOS characterization of aqueous-based preparations of mAbs. While mAbs are predominantly formulated in solution, up to 20% are prepared as solid amorphous powders and techniques for the robust characterization of HOS in the solid state remain limited. We propose here the use of solid-state NMR (ssNMR) fingerprinting to inform directly on the HOS of solid preparations of mAbs. Using lyophilized samples of the NISTmAb reference material prepared with different formulation conditions, we demonstrate that 1H-13C cross-polarization (hC-CP) buildup spectral series at natural isotopic abundance mAb samples are sensitive to differences in formulation. We also demonstrate that principal component analysis (PCA) can be used to differentiate the samples from one another in a user-independent manner while also highlighting areas where expert analysis can provide structural details about important molecular interactions in solid-phase protein formulations. Results from this study contribute to establishing the foundation for the use of ssNMR for HOS characterization of solid-phase biotherapeutics.
Collapse
Affiliation(s)
- Jacqueline Perodeau
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey08854, United States
| | - Luke W Arbogast
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, Rockville, Maryland20850, United States
| | - Andrew J Nieuwkoop
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey08854, United States
| |
Collapse
|
10
|
Moreira AS, Bezemer S, Faria TQ, Detmers F, Hermans P, Sierkstra L, Coroadinha AS, Peixoto C. Implementation of Novel Affinity Ligand for Lentiviral Vector Purification. Int J Mol Sci 2023; 24:3354. [PMID: 36834764 PMCID: PMC9966744 DOI: 10.3390/ijms24043354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
The use of viral vectors as therapeutic products for multiple applications such as vaccines, cancer treatment, or gene therapies, has been growing exponentially. Therefore, improved manufacturing processes are needed to cope with the high number of functional particles required for clinical trials and, eventually, commercialization. Affinity chromatography (AC) can be used to simplify purification processes and generate clinical-grade products with high titer and purity. However, one of the major challenges in the purification of Lentiviral vectors (LVs) using AC is to combine a highly specific ligand with a gentle elution condition assuring the preservation of vector biological activity. In this work, we report for the first time the implementation of an AC resin to specifically purify VSV-G pseudotyped LVs. After ligand screening, different critical process parameters were assessed and optimized. A dynamic capacity of 1 × 1011 total particles per mL of resin was determined and an average recovery yield of 45% was found for the small-scale purification process. The established AC robustness was confirmed by the performance of an intermediate scale providing an infectious particles yield of 54%, which demonstrates the scalability and reproducibility of the AC matrix. Overall, this work contributes to increasing downstream process efficiency by delivering a purification technology that enables high purity, scalability, and process intensification in a single step, contributing to time-to-market reduction.
Collapse
Affiliation(s)
- Ana Sofia Moreira
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
- ITQB Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sandra Bezemer
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | - Tiago Q. Faria
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Frank Detmers
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | - Pim Hermans
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | | | - Ana Sofia Coroadinha
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Cristina Peixoto
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| |
Collapse
|
11
|
Thakral S, Sonje J, Munjal B, Bhatnagar B, Suryanarayanan R. Mannitol as an Excipient for Lyophilized Injectable Formulations. J Pharm Sci 2023; 112:19-35. [PMID: 36030846 DOI: 10.1016/j.xphs.2022.08.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022]
Abstract
The review summarizes the current state of knowledge of mannitol as an excipient in lyophilized injectable small and large molecule formulations. When compared with glycine, the physicochemical properties of mannitol make it a desirable and preferred bulking agent. Though mannitol is a popular bulking agent in freeze-dried formulations, its use may pose certain challenges such as vial breakage or its existence as a metastable crystalline hemihydrate in the final cake, necessitating appropriate mitigation strategies. The understanding of the phase behavior of mannitol in aqueous systems, during the various stages of freeze-drying, can be critical for the optimization of freeze-drying cycle parameters in multi-component formulations. Finally, using a decision tree as a guiding tool, we demonstrate the use of orthogonal techniques for attaining a stable and cost-effective lyophilized drug product containing mannitol.
Collapse
Affiliation(s)
- Seema Thakral
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America.
| | - Jayesh Sonje
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Bhushan Munjal
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Bakul Bhatnagar
- Pfizer Inc., BioTherapeutics, Pharmaceutical Sciences, 1 Burtt Road, Andover, MA 01810, United States of America
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
12
|
Castañeda Ruiz AJ, Shetab Boushehri MA, Phan T, Carle S, Garidel P, Buske J, Lamprecht A. Alternative Excipients for Protein Stabilization in Protein Therapeutics: Overcoming the Limitations of Polysorbates. Pharmaceutics 2022; 14:2575. [PMID: 36559072 PMCID: PMC9781097 DOI: 10.3390/pharmaceutics14122575] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Given their safety and efficiency in protecting protein integrity, polysorbates (PSs) have been the most widely used excipients for the stabilization of protein therapeutics for years. In recent decades, however, there have been numerous reports about visible or sub-visible particles in PS-containing biotherapeutic products, which is a major quality concern for parenteral drugs. Alternative excipients that are safe for parenteral administration, efficient in protecting different protein drugs against various stress conditions, effective in protein stabilization in high-concentrated liquid formulations, stable under the storage conditions for the duration of the product's shelf-life, and compatible with other formulation components and the primary packaging are highly sought after. The aim of this paper is to review potential alternative excipients from different families, including surfactants, carbohydrate- and amino acid-based excipients, synthetic amphiphilic polymers, and ionic liquids that enable protein stabilization. For each category, important characteristics such as the ability to stabilize proteins against thermal and mechanical stresses, current knowledge related to the safety profile for parenteral administration, potential interactions with other formulation components, and primary packaging are debated. Based on the provided information and the detailed discussion thereof, this paper may pave the way for the identification or development of efficient excipients for biotherapeutic protein stabilization.
Collapse
Affiliation(s)
- Angel J. Castañeda Ruiz
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, 53121 Bonn, Germany
| | | | - Tamara Phan
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Stefan Carle
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Julia Buske
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, 53121 Bonn, Germany
| |
Collapse
|
13
|
Mehanna MM, Abla KK. Recent Advances in Freeze-Drying: Variables, Cycle Optimization, and Innovative Techniques. Pharm Dev Technol 2022; 27:904-923. [PMID: 36174214 DOI: 10.1080/10837450.2022.2129385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Freeze-drying (FD) is the most substantial drying technique utilized in the pharmaceutical and biopharmaceutical industries. It is a drying process where the solvent is crystallized at low temperatures and then sublimed from the solid-state directly into the vapor phase. Although FD possesses several merits as its suitability for thermolabile materials and its ability to produce dry products with high-quality attributes, it is a complex and prolonged process that requires optimization of both; process and formulation variables. This review attains to disassemble freeze-drying complications through a detailed explanation of the lyophilization concept, stages, the factors influencing the process including controlled ice nucleation, and the modified and innovative freeze-drying technologies proposed in recent years to overcome the shortage of traditional freeze-drying. In addition, this work points out the quality by design (QbD), critical quality of attributes (CQAs), limitations, and drawbacks of lyophilization.HighlightsLyophilization is a propitious drying technique for thermolabile materials.Optimizing the lyophilization cycle requires controlling the process parameters.The formulation excipients and the dispersion medium play crucial roles in designing a successful process.Numerous approaches were developed to ameliorate the lyophilization performance.
Collapse
Affiliation(s)
- Mohammed M Mehanna
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Kawthar K Abla
- Pharmaceutical Nanotechnology Research Lab, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| |
Collapse
|
14
|
Luther DC, Nagaraj H, Goswami R, Çiçek YA, Jeon T, Gopalakrishnan S, Rotello VM. Direct Cytosolic Delivery of Proteins Using Lyophilized and Reconstituted Polymer-Protein Assemblies. Pharm Res 2022; 39:1197-1204. [PMID: 35297498 PMCID: PMC10587898 DOI: 10.1007/s11095-022-03226-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Cytosolic delivery of proteins accesses intracellular targets for chemotherapy and immunomodulation. Current delivery systems utilize inefficient endosomal pathways of uptake and escape that lead to degradation of delivered cargo. Cationic poly(oxanorbornene)imide (PONI) polymers enable highly efficient cytosolic delivery of co-engineered proteins, but aggregation and denaturation in solution limits shelf life. In the present study we evaluate polymer-protein nanocomposite vehicles as candidates for lyophilization and point-of-care resuspension to provide a transferrable technology for cytosolic protein delivery. METHODS Self-assembled nanocomposites of engineered poly(glutamate)-tagged (E-tagged) proteins and guanidinium-functionalized PONI homopolymers were generated, lyophilized, and stored for 2 weeks. After reconstitution and delivery, cytosolic access of E-tagged GFP cargo (GFPE15) was assessed through diffuse cytosolic and nuclear fluorescence, and cell killing with chemotherapeutic enzyme Granzyme A (GrAE10). Efficiency was quantified between freshly prepared and lyophilized samples. RESULTS Reconstituted nanocomposites retained key structural features of freshly prepared assemblies, with minimal loss of material. Cytosolic delivery (> 80% efficiency of freshly prepared nanocomposites) of GFPE15 was validated in several cell lines, with intracellular access validated and quantified through diffusion into the nucleus. Delivery of GrAE10 elicited significant tumorigenic cell death. Intracellular access of cytotoxic protein was validated through cell viability. CONCLUSION Reconstituted nanocomposites achieved efficient cytosolic delivery of protein cargo and demonstrated therapeutic applicability with delivery of GrAE10. Overall, this strategy represents a versatile and highly translatable method for cytosolic delivery of proteins.
Collapse
Affiliation(s)
- David C Luther
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Yağız Anıl Çiçek
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA.
| |
Collapse
|
15
|
Yang C, Yu C, Zhang M, Yang X, Dong H, Dong Q, Zhang H, Li L, Guo X, Zang H. Investigation of protective effect of ethanol on the natural structure of protein with infrared spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 271:120935. [PMID: 35121476 DOI: 10.1016/j.saa.2022.120935] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
The stability of biological drugs with protein as an active substance depends heavily on the retention of natural protein structure during freeze-drying. Stabilizers have become important substances in the process of protein freeze-drying. In order to further understand the mechanism of the interaction between protein and stabilizers, human serum albumin (HSA) and simple hydroxyl compound ethanol were used as models. Infrared (IR) spectroscopy combined with chemometrics was implemented to investigate the changes of secondary structure and hydration of HSA when different concentrations of ethanol were considered as interference. Through the analysis of the protein secondary structure and hydrated layer, we found that the addition of ethanol-d6 increased the α-helix of HSA and reduced the disordered structure. The hydrogen bond structure around HSA was enhanced and intermolecular aggregation was reduced through the action of the water molecules. The hypothesis was verified by circular dichroism (CD) and transmission electron microscopy (TEM) observation by adding different concentrations of ethanol-d6. It was found that a small amount of ethanol could protect the native conformation of HSA. In conclusion, this study revealed the mechanism of ethanol as a protein protector, provided a new idea for protein purification process and a theoretical basis for biomolecular interaction.
Collapse
Affiliation(s)
- Cui Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chen Yu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengqi Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangchun Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hailing Dong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qin Dong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hui Zhang
- National Glycoengineering Research Center, Shandong University, Jinan, Shandong 250012, China; Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Jinan, Shandong 250012, China
| | - Lian Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Chemical Biology (Ministry of Education), Shandong University, Jinan, Shandong 250012, China
| | - Xueping Guo
- Bloomage Biotechnology Corporation Limited, Tianchen Street 678, Jinan, Shandong 250012, China
| | - Hengchang Zang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; National Glycoengineering Research Center, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Chemical Biology (Ministry of Education), Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
16
|
Munjal B, Patel SM, Suryanarayanan R. Role of Arginine Salts in Preventing Freezing-induced Increase in Subvisible Particles in Protein Formulations. Int J Pharm 2022; 619:121694. [PMID: 35331829 DOI: 10.1016/j.ijpharm.2022.121694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
While arginine hydrochloride (ArgHCl) has emerged as a potential stabilizer of protein drugs in liquid formulations the purpose of this manuscript was to evaluate its stabilization potential in frozen solutions. The phase behavior of frozen AgHCl solutions was investigated by differential scanning calorimetry and low temperature powder X-ray diffractometry. The aggregation of β-galactosidase was evaluated following freeze-thaw cycling in ArgHCl solutions with and without mannitol. ArgHCl (5% w/v) was retained amorphous in frozen aqueous solutions and effectively inhibited protein aggregation even after 5 freeze-thaw cycles. Annealing frozen arginine solution (5% w/v) containing mannitol (10% w/v) induced mannitol crystallization which in turn facilitated crystallization of ArgHCl. The stabilizing effect of ArgHCl was completely lost in the presence of mannitol. Use of alternate arginine salts (aspartate, glutamate, and acetate) allowed selective crystallization of mannitol while the arginine was retained amorphous and stabilized the protein.
Collapse
Affiliation(s)
- Bhushan Munjal
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Sajal M Patel
- Dosage Form Design & Development, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
17
|
Optimization of Heat-Resistance Technology for a Duck Hepatitis Lyophilized Live Vaccine. Vaccines (Basel) 2022; 10:vaccines10020269. [PMID: 35214727 PMCID: PMC8880185 DOI: 10.3390/vaccines10020269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 02/05/2023] Open
Abstract
In this study, to improve the quality of a live attenuated vaccine for duck viral hepatitis (DHV), the lyophilization of a heat-resistant duck hepatitis virus vaccine was optimized. The optimized heat protectors were made of 10% sucrose, 1.2% pullulan, 0.5% PVP, and 1% arginine, etc., with a titer freeze-drying loss of ≤0.50 Lg. The vaccine product’s valence measurements demonstrated the following: the vaccine could be stored at 2–8 °C for 18 months with a virus titer loss ≤0.91 Lg; at 37 °C for 10 days with a virus valence loss ≤0.89 Lg; and at 45 °C for 3 days with a virus titer loss ≤0.90 Lg. Regarding safety, no deaths occurred in two-day-old ducklings immunized with a 10 times dose vaccine; their energy, diet, and weight gain were all normal, demonstrating that the DHV heat-resistant vaccines were safe for ducklings and did not cause any immune side effects. Duck viral hepatitis freeze-dried vaccine began to produce antibodies at 7 d after immunization, reached above 5.0 on 14 d, and reached above 7.0 on 21 d, showing a continuous upward trend. This indicates that duck viral hepatitis vaccine has a good immunogen level. The optimization of the freeze-drying process saves costs and also improves the quality of the freeze-drying products, which provides important theoretical and technical support for the further study of vaccine products.
Collapse
|
18
|
Wang SS, Yan YS, Ho K. US FDA-approved therapeutic antibodies with high-concentration formulation: summaries and perspectives. Antib Ther 2021; 4:262-272. [PMID: 34909579 PMCID: PMC8664682 DOI: 10.1093/abt/tbab027] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/06/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Thirty four (34) of the total US FDA approved 103 therapeutic antibody drugs, accounts for one third of the total approved mAbs, are formulated with high protein concentration (100 mg/mL or above) which are the focus of this article. The highest protein concentration of these approved mAbs is 200 mg/mL. The dominant administration route is subcutaneous (76%). Our analysis indicates that it may be rational to implement a platform formulation containing polysorbate, histidine and sucrose to accelerate high concentration formulation development for antibody drugs. Since 2015, the FDA approval numbers are significantly increased which account for 76% of the total approval numbers, i.e., 26 out of 34 highly concentrated antibodies. Thus, we believe that the high concentration formulations of antibody drugs will be the future trend of therapeutic antibody formulation development, regardless of the challenges of highly concentrated protein formulations.
Collapse
Affiliation(s)
- Shawn Shouye Wang
- CMC Management, WuXi Biologics, 1 Cedarbrook Drive, Cranbury, NJ 08512, USA
| | - Yifei Susie Yan
- Biologics CMC Leadership training program, WuXi Biologics, Palo Alto, CA, USA
| | - Kin Ho
- CMC Management, WuXi Biologics, 1 Cedarbrook Drive, Cranbury, NJ 08512, USA
| |
Collapse
|
19
|
Sharma A, Khamar D, Cullen S, Hayden A, Hughes H. Innovative Drying Technologies for Biopharmaceuticals. Int J Pharm 2021; 609:121115. [PMID: 34547393 DOI: 10.1016/j.ijpharm.2021.121115] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 01/30/2023]
Abstract
In the past two decades, biopharmaceuticals have been a breakthrough in improving the quality of lives of patients with various cancers, autoimmune, genetic disorders etc. With the growing demand of biopharmaceuticals, the need for reducing manufacturing costs is essential without compromising on the safety, quality, and efficacy of products. Batch Freeze-drying is the primary commercial means of manufacturing solid biopharmaceuticals. However, Freeze-drying is an economically unfriendly means of production with long production cycles, high energy consumption and heavy capital investment, resulting in high overall costs. This review compiles some potential, innovative drying technologies that have not gained popularity for manufacturing parenteral biopharmaceuticals. Some of these technologies such as Spin-freeze-drying, Spray-drying, Lynfinity® Technology etc. offer a paradigm shift towards continuous manufacturing, whereas PRINT® Technology and MicroglassificationTM allow controlled dry particle characteristics. Also, some of these drying technologies can be easily scaled-up with reduced requirement for different validation processes. The inclusion of Process Analytical Technology (PAT) and offline characterization techniques in tandem can provide additional information on the Critical Process Parameters (CPPs) and Critical Quality Attributes (CQAs) during biopharmaceutical processing. These processing technologies can be envisaged to increase the manufacturing capacity for biopharmaceutical products at reduced costs.
Collapse
Affiliation(s)
- Ashutosh Sharma
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Main Campus, Cork Road, Waterford X91K0EK, Ireland.
| | - Dikshitkumar Khamar
- Sanofi, Manufacturing Science, Analytics and Technology (MSAT), IDA Industrial Park, Waterford X91TP27, Ireland
| | - Sean Cullen
- Gilead Sciences, Commercial Manufacturing, IDA Business & Technology Park, Carrigtwohill, Co. Cork T45DP77, Ireland
| | - Ambrose Hayden
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Main Campus, Cork Road, Waterford X91K0EK, Ireland
| | - Helen Hughes
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Main Campus, Cork Road, Waterford X91K0EK, Ireland
| |
Collapse
|
20
|
Das TK, Sreedhara A, Colandene JD, Chou DK, Filipe V, Grapentin C, Searles J, Christian TR, Narhi LO, Jiskoot W. Stress Factors in Protein Drug Product Manufacturing and Their Impact on Product Quality. J Pharm Sci 2021; 111:868-886. [PMID: 34563537 DOI: 10.1016/j.xphs.2021.09.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 01/22/2023]
Abstract
Injectable protein-based medicinal products (drug products, or DPs) must be produced by using sterile manufacturing processes to ensure product safety. In DP manufacturing the protein drug substance, in a suitable final formulation, is combined with the desired primary packaging (e.g., syringe, cartridge, or vial) that guarantees product integrity and enables transportation, storage, handling and clinical administration. The protein DP is exposed to several stress conditions during each of the unit operations in DP manufacturing, some of which can be detrimental to product quality. For example, particles, aggregates and chemically-modified proteins can form during manufacturing, and excessive amounts of these undesired variants might cause an impact on potency or immunogenicity. Therefore, DP manufacturing process development should include identification of critical quality attributes (CQAs) and comprehensive risk assessment of potential protein modifications in process steps, and the relevant steps must be characterized and controlled. In this commentary article we focus on the major unit operations in protein DP manufacturing, and critically evaluate each process step for stress factors involved and their potential effects on DP CQAs. Moreover, we discuss the current industry trends for risk mitigation, process control including analytical monitoring, and recommendations for formulation and process development studies, including scaled-down runs.
Collapse
Affiliation(s)
- Tapan K Das
- Bristol Myers Squibb, Biologics Development, New Brunswick, New Jersey 08903, USA.
| | | | - James D Colandene
- GlaxoSmithKline, Biopharmaceutical Product Sciences, 1250 S Collegeville Road, Collegeville, PA 19425, USA
| | - Danny K Chou
- Compassion BioSolution, LLC, Lomita, CA 90717, USA
| | | | - Christoph Grapentin
- Lonza AG, Drug Product Services, Hochbergerstrasse 60G, 4057 Basel, Switzerland
| | - Jim Searles
- Pfizer Inc., Biotherapeutics Pharmaceutical Sciences Research and Development, 875 Chesterfield Pkwy W, Chesterfield, MO 63017 USA
| | | | | | - Wim Jiskoot
- Leiden University, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands; Coriolis Pharma, Martinsried, Germany
| |
Collapse
|
21
|
Tang M, Hattori Y. Effect of using amino acids in the freeze-drying of siRNA lipoplexes on gene knockdown in cells after reverse transfection. Biomed Rep 2021; 15:72. [PMID: 34405044 DOI: 10.3892/br.2021.1448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/25/2021] [Indexed: 11/06/2022] Open
Abstract
Recently, small interfering RNA (siRNA)/cationic liposome complexes (siRNA lipoplexes) have become a crucial research tool for studying gene function. Easy and reliable siRNA transfection with a large set of siRNAs is required for the successful screening of gene function. Reverse (Rev)-transfection with freeze-dried siRNA lipoplexes is validated for siRNA transfection with a large set of siRNAs in a multi-well plate. In our previous study, it was shown that Rev-transfection with siRNA lipoplexes freeze-dried in disaccharides or trisaccharides resulted in long-term stability with a high level of gene-knockdown activity. In the present study, the effects of amino acids used as cryoprotectants in the freeze-drying of siRNA lipoplexes on gene knockdown via Rev-transfection were assessed. A total of 15 types of amino acids were used to prepare freeze-dried siRNA lipoplexes, and it was found that the freeze-drying of siRNA lipoplexes with amino acid concentrations >100 mM strongly suppressed targeted gene expression regardless of the amino acid type; however, some amino acids strongly upregulated or downregulated gene expression in the cells transfected with negative control siRNA. Amongst the amino acids tested, the presence of asparagine showed specific gene-knockdown activity, forming large cakes after freeze-drying and retaining a favorable siRNA lipoplex size after rehydration. These findings provide valuable information regarding amino acids as cryoprotectants for Rev-transfection using freeze-dried siRNA lipoplexes for the efficient delivery of siRNA into cells.
Collapse
Affiliation(s)
- Min Tang
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Yoshiyuki Hattori
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| |
Collapse
|
22
|
Thakral S, Sonje J, Munjal B, Suryanarayanan R. Stabilizers and their interaction with formulation components in frozen and freeze-dried protein formulations. Adv Drug Deliv Rev 2021; 173:1-19. [PMID: 33741437 DOI: 10.1016/j.addr.2021.03.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/06/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
This review aims to provide an overview of the current knowledge on protein stabilization during freezing and freeze-drying in relation to stress conditions commonly encountered during these processes. The traditional as well as refined mechanisms by which excipients may stabilize proteins are presented. These stabilizers encompass a wide variety of compounds including sugars, sugar alcohols, amino acids, surfactants, buffers and polymers. The rational selection of excipients for use in frozen and freeze-dried protein formulations is presented. Lyophilized protein formulations are generally multicomponent systems, providing numerous possibilities of excipient-excipient and protein-excipient interactions. The interplay of different formulation components on the protein stability and excipient functionality in the frozen and freeze-dried systems are reviewed, with discussion of representative examples of such interactions.
Collapse
|
23
|
Kulkarni SS, Patel SM, Suryanarayanan R, Rinella JV, Bogner RH. Key factors governing the reconstitution time of high concentration lyophilized protein formulations. Eur J Pharm Biopharm 2021; 165:361-373. [PMID: 33974974 DOI: 10.1016/j.ejpb.2021.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/01/2022]
Abstract
Lyophilized protein formulations containing highly concentrated proteins often have long and variable reconstitution times. Reconstitution time is dependent on a number of factors in a complex manner. Furthermore, factors influencing the reconstitution of partially crystalline cakes are reportedly different from those of amorphous cakes. The objectives of this work were to identify the key factors governing reconstitution and understand the mechanisms involved in reconstitution of both amorphous and partially crystalline cakes. Partial crystallinity in the final cake, larger pores and low "concentrated formulation viscosity" (i.e., viscosity near the surface of the dissolving cake) were identified as desirable characteristics for expediting reconstitution. Crystallinity and larger pores dramatically improved wettability and liquid penetration into partially crystalline cakes, ultimately resulting in well dispersed small pieces of partially dissolved cake. The smaller disintegrated cake pieces dissolved faster because of the increased surface area. The amorphous cakes exhibited poorer wettability than partially crystalline cakes. Moreover, the ability of the reconstitution fluid to penetrate the pores, and the resulting cake disintegration was much lower than that observed for partially crystalline cakes. In fact, for some of the amorphous cakes, the reconstitution fluid did not penetrate the cake at all. As a result, the undissolved intact cake or a large cake chunk floated on the reconstitution fluid amidst foam or bubbles generated during reconstitution. Dissolution of the floating cake appeared to proceed via gradual surface erosion where reconstitution time was found to be highly correlated with the viscosity near the surface of the dissolving cake solids. A higher viscosity prolonged reconstitution. Thus, both formulation and processing conditions can be tailored to achieve faster reconstitution. Including a crystallizable excipient proved to be beneficial. Incorporating an annealing step to facilitate crystallization of the crystallizable excipient and to promote larger pores was also found to be advantageous. A viscosity lowering excipient in the formulation could potentially be helpful but needs to be explored further.
Collapse
Affiliation(s)
- Shreya S Kulkarni
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA.
| | - Sajal M Patel
- Dosage Form Design & Development, Biopharmaceutical Development, AstraZeneca, Gaithersburg, MD 20878, USA.
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joseph V Rinella
- Biopharmaceutical Product Sciences, GlaxoSmithKline, Collegeville, PA 19426, USA.
| | - Robin H Bogner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
24
|
Screening of novel excipients for freeze-dried protein formulations. Eur J Pharm Biopharm 2021; 160:55-64. [PMID: 33508435 DOI: 10.1016/j.ejpb.2021.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/22/2022]
Abstract
The typical excipients used as bulking agents and lyoprotectants for freeze-drying are usually limited to only a few selected substances, such as sucrose and mannitol. Considering the sheer diversity amongst proteins, it is doubtful that this limited choice should, in every case, provide the best possible option in order to achieve the most stable product. In this work, a screening of 12 proteins with 64 excipients was conducted in order to increase the knowledge space of potential excipients. Three critical quality attributes (CQAs) of the freeze-dried products, namely the solid state, the cake appearance and the protein integrity based on changes in tryptophan fluorescence were investigated by high throughput X-ray powder diffraction, image analysis and intrinsic fluorescence spectroscopy, respectively. It was found, that in some cases the excipient had a dominating influence on the CQAs, whilst in other cases the CQAs were primarily protein dependent, or that the CQAs were dependent on the combination of both. In the course of this investigation, a general view of potentially relevant excipients, and their interplay with various proteins, was obtained, thereby furthermore paving the way for the use of novel freeze-drying excipients.
Collapse
|
25
|
Seifert I, Friess W. The effect of residual moisture on a monoclonal antibody stability in L-arginine based lyophilisates. Eur J Pharm Biopharm 2021; 158:53-61. [DOI: 10.1016/j.ejpb.2020.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/18/2020] [Accepted: 11/07/2020] [Indexed: 10/23/2022]
|
26
|
Link FJ, Heng JYY. Enhancing the crystallisation of insulin using amino acids as soft-templates to control nucleation. CrystEngComm 2021. [DOI: 10.1039/d1ce00026h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Amino acid as soft templates in promoting nucleation of insulin.
Collapse
Affiliation(s)
- Frederik J. Link
- Department of Chemical Engineering
- Imperial College London, South Kensington Campus
- London SW7 2AZ
- UK
| | - Jerry Y. Y. Heng
- Department of Chemical Engineering
- Imperial College London, South Kensington Campus
- London SW7 2AZ
- UK
- Institute for Molecular Science and Engineering
| |
Collapse
|
27
|
Seifert I, Bregolin A, Fissore D, Friess W. The Influence of Arginine and Counter-Ions: Antibody Stability during Freeze-Drying. J Pharm Sci 2020; 110:2017-2027. [PMID: 33316241 DOI: 10.1016/j.xphs.2020.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 11/16/2022]
Abstract
Amino acids, for example L-arginine, are used in lyophilisation as crystalline bulking, buffering, viscosity reducing or stabilising excipients. In this study, arginine was formulated with different counter ions (hydrochloride, citrate, lactobionate, phosphate, and succinate). A monoclonal antibody was investigated in sugar-free arginine formulations and mixtures with sucrose regarding cake appearance and protein aggregation and fragmentation. Arginine hydrochloride formulations collapsed during lyophilisation due to its low Tg' and partially crystallised during storage, but provided the best protein stability at low antibody concentration, followed by arginine succinate. Arginine citrate/phosphate/lactobionate formulations resulted in amorphous elegant cakes, but inferior protein stability. Addition of sucrose improved cake appearance and protein stability. Arginine phosphate with sucrose resulted in similar protein stability as the sucrose reference. Mixtures of sucrose with arginine hydrochloride/lactobionate/succinate provided better stability than sucrose alone. While 50 mg/mL antibody improved the cake appearance, only arginine lactobionate provided sufficient protein stability next to sucrose. Overall, sugar-free arginine hydrochloride and lactobionate lyophilisates stabilised the antibody comparably or better than sucrose depending on antibody concentration. The best protein stability was found for mixtures of arginine hydrochloride/lactobionate/succinate with sucrose.
Collapse
Affiliation(s)
- Ivonne Seifert
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Alessandro Bregolin
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Turin, Italy
| | - Davide Fissore
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Turin, Italy
| | - Wolfgang Friess
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
28
|
Seifert I, Friess W. Improvement of arginine hydrochloride based antibody lyophilisates. Int J Pharm 2020; 589:119859. [DOI: 10.1016/j.ijpharm.2020.119859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023]
|
29
|
N-Acetylated-L-arginine (NALA) is an enhanced protein aggregation suppressor under interfacial stresses and elevated temperature for protein liquid formulations. Int J Biol Macromol 2020; 166:654-664. [PMID: 33137385 DOI: 10.1016/j.ijbiomac.2020.10.223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022]
Abstract
Even though arginine hydrochloride has been recognized as a protein aggregation suppressor in the biopharmaceutical industry, its use has been questioned due to decreasing transition unfolding temperatures (Tm). Four compounds were designed to enhance the role of arginine by changing the length of the carbon chain with removal or N-acetylation of α-amino group. Biophysical properties were observed by differential scanning calorimetry (DSC), dynamic light scattering (DLS), size-exclusion chromatography (SEC), and flow imaging (FI). N-Acetyl-L-arginine (NALA) performed the best at minimizing decrease in Tm with arginine at different pH. NALA also demonstrated relatively higher colloidal stability than arginine hydrochloride, especially in the acidic pH, thereby reducing agitation stress of IgG. Moreover, NALA exhibited a cooperative effect with commercially used glycine buffer for IVIG to maintain the monomer contents with almost no change and suppressed larger particle formation after agitation with heat. The study concludes that the decreasing Tm of proteins by arginine hydrochloride is due to amide group in the α-carbon chain. Moreover, chemical modification on the group compared to removing it will be a breakthrough of arginine's limitations and optimize storage stability of protein therapeutics.
Collapse
|
30
|
Digital Twin for Lyophilization by Process Modeling in Manufacturing of Biologics. Processes (Basel) 2020. [DOI: 10.3390/pr8101325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Lyophilization stabilizes formulated biologics for storage, transport and application to patients. In process design and operation it is the link between downstream processing and with final formulation to fill and finish. Recent activities in Quality by Design (QbD) have resulted in approaches by regulatory authorities and the need to include Process Analytical Technology (PAT) tools. An approach is outlined to validate a predictive physical-chemical (rigorous) lyophilization process model to act quantitatively as a digital twin in order to allow accelerated process design by modeling and to further-on develop autonomous process optimization and control towards real time release testing. Antibody manufacturing is chosen as a typical example for actual biologics needs. Literature is reviewed and the presented procedure is exemplified to quantitatively and consistently validate the physical-chemical process model with aid of an experimental statistical DOE (design of experiments) in pilot scale.
Collapse
|
31
|
Butreddy A, Dudhipala N, Janga KY, Gaddam RP. Lyophilization of Small-Molecule Injectables: an Industry Perspective on Formulation Development, Process Optimization, Scale-Up Challenges, and Drug Product Quality Attributes. AAPS PharmSciTech 2020; 21:252. [PMID: 32885357 DOI: 10.1208/s12249-020-01787-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Lyophilization is a pivotal manufacturing process to obtain a stable drug product that is unstable as a ready-to-use formulation. Some formulations may require the addition of drug-specific excipients such as stabilizers, buffers, and bulking agents to support the cake appearance and ensure long-term stability of the drug product. Optimization of the lyophilization process parameters at each stage including freezing and primary and secondary drying is important because these parameters can have a direct impact on the process efficiency (shortened cycle time) and product performance (cake appearance and homogeneous moisture content). Several parameters of the formulation, including properties of the active pharmaceutical ingredient, excipients, solvent system, and container closure, determine the success of lyophilization. Development, scale-up, and transfer of the lyophilization cycle are challenging; hence, a comprehensive understanding of the critical parameters related to the formulation, lyophilization process, and lyophilizer design allows designing a quality drug product. One approach for a successful transfer of the lyophilization cycle between the laboratory and commercial-scale lyophilizer is using vial heat transfer coefficient and ice slab test to establish a maximum sublimation rate. This review provides a general overview of the lyophilization process and discusses several key considerations and product development aspects of formulation, process optimization, container closure system, scale-up principles, and drug product quality attributes from the industrial viewpoint. Grapical abstract.
Collapse
|
32
|
Nayak PK, Goode M, Chang DP, Rajagopal K. Ectoine and Hydroxyectoine Stabilize Antibodies in Spray-Dried Formulations at Elevated Temperature and during a Freeze/Thaw Process. Mol Pharm 2020; 17:3291-3297. [PMID: 32672979 DOI: 10.1021/acs.molpharmaceut.0c00395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Maintenance of protein stability during manufacture, storage, and delivery is necessary for the successful development of a drug product. Herein, the utility of two compatible solutes-ectoine and hydroxyectoine-in stabilizing a model protein labeled Fab2 has been investigated. Specifically, the performance of ectoine and hydroxyectoine in stabilizing Fab2 in a spray-dried formulation at elevated temperature and after multiple freeze/thaw cycles has been compared with the performance of a formulation containing trehalose and a formulation containing no excipient as controls. In the solid state at 90 and 37 °C and in freeze concentrate systems, ectoine and hydroxyectoine suppress protein aggregation. Like trehalose, hydroxyectoine also limits N-terminal pyroglutamate formation in Fab2 in the solid state. The extent of protein stabilization is dependent on the excipient concentration in the formulation, but at a 1:1 excipient to protein mass ratio, hydroxyectoine is better than trehalose in stabilizing Fab2. The results presented here suggest that ectoine and hydroxyectoine are effective excipients for stabilizing therapeutic antibodies.
Collapse
Affiliation(s)
- Purnendu K Nayak
- Eurofins Lancaster Laboratories, Lancaster, Pennsylvania 17605, United States
| | - Meghan Goode
- Drug Delivery Department, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Debby P Chang
- Drug Delivery Department, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karthikan Rajagopal
- Drug Delivery Department, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
33
|
Seifert I, Bregolin A, Fissore D, Friess W. Method development and analysis of the water content of the maximally freeze concentrated solution suitable for protein lyophilisation. Eur J Pharm Biopharm 2020; 153:36-42. [PMID: 32526356 DOI: 10.1016/j.ejpb.2020.05.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 11/28/2022]
Abstract
During freeze-drying of a liquid formulation, a freeze-concentrate is formed in the first phase, the freezing step. Understanding the composition of the maximally freeze concentrated solution can help to judge the process stability of biopharmaceuticals during lyophilisation. Our objective was to develop a suitable method to determine the water content of the maximally freeze concentrated solution using differential scanning calorimetry (DSC). Three different methods were compared: (i) the intercept of the glass transition temperature of the maximally freeze concentrated solution Tg' and the melting temperature Tm for a concentration series, (ii) the linear regression of the melting enthalpy starting from the onset of Tg' until the end of the melting event for a concentration series, and (iii) a one-point determination of the amount of unfrozen water. While Method 1 is accurate but requires the analysis of a high number of samples, Method 3 requires only one single sample, with a loss of accuracy. Method 2 works best taking sample preparation and accuracy into account. Various systems containing sugar (sucrose, trehalose) and other excipients (histidine buffer, phosphate buffer, sodium chloride, arginine hydrochloride, arginine citrate) were evaluated with different antibody concentrations to evaluate the composition of the maximally freeze concentrated solution. The freeze concentrates exhibited a water content of 20-30%, slightly dependent on the excipients, but independent of the antibody concentration. The methodology we developed is broadly applicable for the analysis of the composition of maximally freeze concentrated solutions and can help to elucidate protein stability during lyophilisation.
Collapse
Affiliation(s)
- Ivonne Seifert
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Alessandro Bregolin
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Davide Fissore
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Wolfgang Friess
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
34
|
Excipients in freeze-dried biopharmaceuticals: Contributions toward formulation stability and lyophilisation cycle optimisation. Int J Pharm 2020; 576:119029. [DOI: 10.1016/j.ijpharm.2020.119029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/08/2023]
|
35
|
Hristov D, McCartney F, Beirne J, Mahon E, Reid S, Bhattacharjee S, Penarier G, Werner U, Bazile D, Brayden DJ. Silica-Coated Nanoparticles with a Core of Zinc, l-Arginine, and a Peptide Designed for Oral Delivery. ACS APPLIED MATERIALS & INTERFACES 2020; 12:1257-1269. [PMID: 31802658 DOI: 10.1021/acsami.9b16104] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanoparticle constructs for oral peptide delivery at a minimum must protect and present the peptide at the small intestinal epithelium in order to achieve oral bioavailability. In a reproducible, scalable, surfactant-free process, a core was formed with insulin in ratios with two established excipients and stabilizers, zinc chloride and l-arginine. Cross-linking was achieved with silica, which formed an outer shell. The process was reproducible across several batches, and physicochemical characterization of a single batch was confirmed in two independent laboratories. The silica-coated nanoparticles (SiNPs) entrapped insulin with high entrapment efficiency, preserved its structure, and released it at a pH value present in the small intestine. The SiNP delivered insulin to the circulation and reduced plasma glucose in a rat jejunal instillation model. The delivery mechanism required residual l-arginine in the particle to act as a permeation enhancer for SiNP-released insulin in the jejunum. The synthetic process was varied in terms of ratios of zinc chloride and l-arginine in the core to entrap the glucagon-like peptide 1 analogue, exenatide, and bovine serum albumin. SiNP-delivered exenatide was also bioactive in mice to some extent following oral gavage. The process is the basis for a platform for oral peptide and protein delivery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Geraldine Penarier
- Sanofi Recherche & Développement , PSO/LGCR , Bâtiment BLP, rue du Pr Blayac , 34184 Montpellier Cedex 4 , France
| | - Ulrich Werner
- Sanofi-Aventis Deutschland GmbH . Industriepark Höchst , K703 65926 Frankfurt , Germany
| | - Didier Bazile
- Sanofi Recherche & Développement , CMC External Innovation , 82, avenue Raspail , 94250 Gentilly Cedex , France
| | | |
Collapse
|
36
|
Gitter JH, Geidobler R, Presser I, Winter G. Microwave-Assisted Freeze-Drying of Monoclonal Antibodies: Product Quality Aspects and Storage Stability. Pharmaceutics 2019; 11:E674. [PMID: 31842296 PMCID: PMC6956074 DOI: 10.3390/pharmaceutics11120674] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 11/29/2022] Open
Abstract
In order to overcome the downside of long conventional freeze-drying (CFD) process times for monoclonal antibody formulations, microwave-assisted freeze-drying (MFD) was introduced. Recently, the general applicability and potential shortening of drying times were shown. However, little is known about the storage stability of MFD products compared to CFD references. Additionally, batch homogeneity issues were seen within MFD in the past. In this study, we examined four different formulations of two different monoclonal antibodies using three different glass-forming excipients: sucrose, trehalose, and arginine phosphate. These formulations were freeze-dried with two different drying protocols (CFD and MFD), stored for 24 weeks, and analyzed for solid-state and protein-related quality attributes. Moreover, a new microwave generator setup was investigated for its potential to improve batch homogeneity. In all investigated formulations, comparable stability profiles were found, although the classical magnetron generator led to inferior batch homogeneity with respect to residual moisture distribution. In contrast, the new MFD setup indicated the potential to approximate batch homogeneity to the level of CFD. However, for future applications, there is an unabated need for new machine designs to comply with pharmaceutical manufacturing requirements.
Collapse
Affiliation(s)
- Julian Hendryk Gitter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Raimund Geidobler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Pharmaceutical Development Biologicals, 88397 Biberach an der Riß, Germany
| | - Ingo Presser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Pharmaceutical Development Biologicals, 88397 Biberach an der Riß, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
37
|
Gervasi V, Cullen S, McCoy T, Crean A, Vucen S. Application of a mixture DOE for the prediction of formulation critical temperatures during lyophilisation process optimisation. Int J Pharm 2019; 572:118807. [PMID: 31678526 DOI: 10.1016/j.ijpharm.2019.118807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/12/2019] [Accepted: 10/16/2019] [Indexed: 01/27/2023]
Abstract
During lyophilisation cycle design, primary drying parameters (chamber pressure and shelf temperature) are adjusted to maximize the sublimation rate and prevent cake collapse, by maintaining the product continuously below its critical temperatures. The objective of this study was to employ mixture design of experiments to generate empirical models capable of predicting glass transition of the maximally freeze concentrated solution (Tg') and collapse temperature (Tc) of amorphous protein (BSA and IgG1) formulations. Additionally, the models developed aid the design of high concentration protein formulations with maximised critical temperatures to obtain shorter and more cost-effective lyophilisation cycles. Formulations contain sucrose as cryo/lyo-protectant and arginine/arginine-HCl as multifunctional excipient (e.g. solubility enhancer, viscosity and aggregation suppressor). The impact of formulation components at varied ratios on critical temperatures was evaluated; the amorphous excipients decrease critical temperatures, on the contrary, the protein increases critical temperatures. The robustness of the empirical models generated with BSA formulations was verified with BSA and IgG1 formulations. The models showed greater accuracy in predicting Tg' than the Fox-Flory equation. For the first time, empirical models are reported to predict both critical temperatures. Finally, unconventional collapse events observed for formulations with and without arginine/arginine-HCl at different protein concentrations are also discussed.
Collapse
Affiliation(s)
- V Gervasi
- Synthesis and Solid State Pharmaceutical Centre (SSPC), School of Pharmacy, University College Cork, Cork, Ireland; Manufacturing Science Department, Sanofi, Waterford, Ireland
| | - S Cullen
- Manufacturing Science Department, Sanofi, Waterford, Ireland
| | - T McCoy
- Global Biologics Drug Product Development (BioDPD), Sanofi R&D, Framingham, MA, USA
| | - A Crean
- Synthesis and Solid State Pharmaceutical Centre (SSPC), School of Pharmacy, University College Cork, Cork, Ireland
| | - S Vucen
- Synthesis and Solid State Pharmaceutical Centre (SSPC), School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
38
|
Abstract
The pharmaceutical and chemical industries depend on additives to protect enzymes and other proteins against stresses that accompany their manufacture, transport, and storage. Common stresses include vacuum-drying, freeze-thawing, and freeze-drying. The additives include sugars, compatible osmolytes, amino acids, synthetic polymers, and both globular and disordered proteins. Scores of studies have been published on protection, but the data have never been analyzed systematically. To spur efforts to understand the sources of protection and ultimately develop more effective formulations, we review ideas about the mechanisms of protection, survey the literature searching for patterns of protection, and then compare the ideas to the data.
Collapse
Affiliation(s)
- Samantha Piszkiewicz
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Gary J. Pielak
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
39
|
Anumalla B, Prabhu NP. Counteracting Effect of Charged Amino Acids Against the Destabilization of Proteins by Arginine. Appl Biochem Biotechnol 2019; 189:541-555. [PMID: 31056736 DOI: 10.1007/s12010-019-03026-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/22/2019] [Indexed: 10/26/2022]
Abstract
Studies on osmolyte-induced effects on proteins help in enhancing protein stability under stressed conditions for various applications. Using mixtures of osmolytes could indeed widen their applications. The combinatorial effects of osmolytes with methylamines are majorly found in the literature; however, such studies are limited on the amino acid class of osmolytes. The present study examines the effect of charged amino acids Arg, Asp, and Lys on the stability of RNase A and α-LA. The thermal stabilities of the proteins in the presence of osmolytes are monitored by absorption changes, and the structural changes are analyzed using fluorescence quenching and near-UV circular dichroism (CD). These results are compared with our previous report on the effect of Glu. Arg destabilizes both the proteins whereas Asp, Lys, and Glu stabilize the proteins. The extent of stability provided by Asp and Glu is almost same and higher than Lys in RNase A. However, the stability acquired in the presence of Asp and Lys is comparable for α-LA and Glu provides higher stability. Further, the quenching and CD results suggest that the addition of amino acids do not alter the structure of the proteins significantly. The counteracting abilities of the stabilizing amino acids (stAAs) against Arg are then investigated. The results show that Glu could counteract Arg at the lowest fraction in the mixture. Lys requires nearly equimolar concentration whereas Asp needs almost double the concentration to counteract Arg induced destabilization of the proteins. At higher concentrations, the counteracting ability of Asp and Lys is similar for both the proteins. The counteracting ratio might slightly vary among the proteins, and it is not necessary that the amino acid providing higher stability to the protein could more effectively counteract Arg. This could be due to the change in the extent of preferential hydration of the proteins by stAAs in the presence of Arg. The results suggest that the addition of stAAs could be an effective strategy to increase the protein stability in biotechnology and biopharma applications.
Collapse
Affiliation(s)
- Bramhini Anumalla
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India
| | - N Prakash Prabhu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India.
| |
Collapse
|
40
|
|
41
|
Wang W, Roberts CJ. Protein aggregation – Mechanisms, detection, and control. Int J Pharm 2018; 550:251-268. [DOI: 10.1016/j.ijpharm.2018.08.043] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
|
42
|
Hackl E, Darkwah J, Smith G, Ermolina I. Effect of Arginine on the Aggregation of Protein in Freeze-Dried Formulations Containing Sugars and Polyol: II. BSA Reconstitution and Aggregation. AAPS PharmSciTech 2018; 19:2934-2947. [PMID: 29980982 DOI: 10.1208/s12249-018-1114-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/24/2018] [Indexed: 11/30/2022] Open
Abstract
The current paper continues our study on the ability of L-arginine to prevent/reduce the aggregation of proteins that results from the various stresses during the lyophilisation and/or storage of lyophilized protein-based products. The first part of our study, i.e. formulation development, was devoted to the rational design and optimization of an L-arginine containing lyophilized formulation which can resist the natural tendency of L-arginine to absorb atmosphere moisture. Mannitol and trehalose were chosen among other excipients to be included in the protein-based formulation, as mannitol in a combination with L-arginine has been shown to reduce moisture sorption while trehalose provides a degree of lyoprotection. In the present study, a number of formulations, which comprised bovine serum albumin (BSA) with and without L-arginine, and with five different ratios of trehalose-to-mannitol (from 30:70 to 80:20) were lyophilised and assessed. The internal structures and the moisture sorption/retention of the lyophilized formulations were characterised. To study the effect of L-arginine on BSA solid-phase stability, the lyophilized powder was exposed to accelerated storage conditions (high moisture (75% RH) and temperature (22 or 45 °C)) for up to 24 h. The lyophilized BSA formulations were then reconstituted and solution-state protein aggregation assessed by turbidimetry at 360 nm and fluorescence spectroscopy using the thioflavin T assay. It was demonstrated that L-arginine can be used in protein-based freeze-dried formulations to significantly reduce the aggregation of protein during the manufacturing, storage and subsequent reconstitution. The results also revealed the importance of a sufficient amount of mannitol in the arginine-containing formulations.
Collapse
|
43
|
Boll B, Josse L, Heubach A, Hochenauer S, Finkler C, Huwyler J, Koulov AV. Impact of non-ideal analyte behavior on the separation of protein aggregates by asymmetric flow field-flow fractionation. J Sep Sci 2018; 41:2854-2864. [DOI: 10.1002/jssc.201701457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/14/2018] [Accepted: 04/19/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Björn Boll
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
- Department of Pharmaceutical Sciences; Division of Pharmaceutical Technology; University of Basel; Basel Switzerland
| | - Lena Josse
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
| | - Anja Heubach
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
| | - Sophie Hochenauer
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
| | - Christof Finkler
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences; Division of Pharmaceutical Technology; University of Basel; Basel Switzerland
| | - Atanas V. Koulov
- Analytical Development & Quality Control; Pharma Technical Development Biologics Europe; F. Hoffmann-La Roche Ltd.; Basel Switzerland
| |
Collapse
|
44
|
Fernandez-Moure J, Maisha N, Lavik EB, Cannon JW. The Chemistry of Lyophilized Blood Products. Bioconjug Chem 2018; 29:2150-2160. [PMID: 29791137 DOI: 10.1021/acs.bioconjchem.8b00271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With the development of new biologics and bioconjugates, storage and preservation have become more critical than ever before. Lyophilization is a method of cell and protein preservation by removing a solvent such as water from a substance followed by freezing. This technique has been used in the past and still holds promise for overcoming logistic challenges in safety net hospitals with limited blood banking resources, austere environments such as combat, and mass casualty situations where existing resources may be outstripped. This method allows for long-term storage and transport but requires the bioconjugation of preservatives to prevent cell destabilization. Trehalose is utilized as a bioconjugate in platelet and red blood cell preservation to maintain protein thermodynamics and stabilizing protein formulations in liquid and freeze-dried states. Biomimetic approaches have been explored as alternatives to cryo- and lyopreservation of blood components. Intravascular hemostats such as PLGA nanoparticles functionalized with PEG motifs, topical hemostats utilizing fibrinogen or chitosan, and liposomal encapsulated hemoglobin with surface modifications are effectively stored long-term through bioconjugation. In thinking about the best methods for storage and transport, we are focusing this topical review on blood products that have the longest track record of preservation and looking at how these methods can be applied to synthetic systems.
Collapse
Affiliation(s)
- Joseph Fernandez-Moure
- Division of Trauma, Surgical Critical Care & Emergency Surgery , Perelman School of Medicine at the University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Nuzhat Maisha
- Department of Chemical, Biochemical & Environmental Engineering , University of Maryland, Baltimore County , Baltimore , Maryland 21250 , United States
| | - Erin B Lavik
- Department of Chemical, Biochemical & Environmental Engineering , University of Maryland, Baltimore County , Baltimore , Maryland 21250 , United States
| | - Jeremy W Cannon
- Division of Trauma, Surgical Critical Care & Emergency Surgery , Perelman School of Medicine at the University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States.,Department of Surgery , Uniformed Services University of the Health Sciences , Bethesda , Maryland 20814 , United States
| |
Collapse
|